Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
MICROBIOTA ENGINEERING
Document Type and Number:
WIPO Patent Application WO/2023/012109
Kind Code:
A2
Abstract:
The invention relates to methods and means for temporally regulating the production of products of interest (eg, proteins or RNAs) in microbiota of subjects, such as gut microbiota of humans or animals.

Inventors:
MUNCK CHRISTIAN (DK)
PORSE ANDREAS (DK)
KRAUSE HAABER JAKOB (DK)
KOVAL LEV (DK)
ALFASTSEN LAUGE (DK)
SEMSEY SZABOLCS (DK)
Application Number:
PCT/EP2022/071562
Publication Date:
February 09, 2023
Filing Date:
August 01, 2022
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
SNIPR BIOME APS (DK)
International Classes:
A61K31/711; A61K35/74; A61P31/04; C12N15/10; C12N15/113; C12N15/63
Attorney, Agent or Firm:
CMS CAMERON MCKENNA NABARRO OLSWANG LLP (GB)
Download PDF:
Claims:
CLAIMS:

1. At least one nucleic acid vector for transfer into a host cell of a microbiota, the vector(s) comprising nucleic acid that comprises

(a) an expressible nucleotide sequence of interest (NS 1) for producing a product of interest (P1) in the host cell; and

(b) an expressible nucleotide sequence (NS2) for producing a regulator product (P2) in the host cell, wherein P2 is operable in the host cell to regulate expression or activity of P1 ; wherein

(c) NS 1 is under the control of a first promoter (optionally a constitutive or inducible promoter) for expression of P1; and

(d) expression or activity of P2 in the host cell is regulatable by exposure of the host cell comprising the at least one vector to a regulator agent (R), thereby regulating the expression or activity of P1.

2. At least one nucleic acid vector according to claim 1 for transfer into a host cell of a microbiota, the vector(s) comprising nucleic acid that comprises a) an expressible nucleotide sequence of interest (NS 1) for producing a product of interest (P1) in the host cell; and b) an expressible nucleotide sequence (NS2) for producing a regulator product (P2) in the host cell, wherein P2 is operable in the host cell to bind to vector nucleic acid to regulate expression of P1; wherein c) NS 1 is under the control of a first promoter (optionally a constitutive or inducible promoter) for expression of P1; and d) NS2 is under the control of a second promoter that is regulatable for expression of P2, wherein binding or exposure of a regulator agent (R) to the vector nucleic acid regulates the second promoter, thereby regulating the expression of P2 and P1; optionally wherein said at least one vector is one vector that comprises both NS1 and NS2.

3. The vector(s) of claim 1 or 2, wherein

(a) P2 is operable in the host cell to bind to the vector nucleic acid to downregulate expression of P1 ; and/or

(b) R upregulates expression of P2. The vector(s) of any preceding claim, wherein P2 is expressible in the host cell for the formation of a nuclease that is operable in the host cell to cut the nucleic acid, optionally wherein the nucleic acid is degraded, thereby downregulating the expression of P1. The vector(s) of any preceding claim, wherein P2 comprises

(a) an RNA-guided nuclease;

(b) an RNA that is operable for guiding an RNA-guided nuclease or a precursor of such an RNA; or

(c) a restriction endonuclease. The vector(s) of claim 5, wherein the guided nuclease is a Cas nuclease, TALEN, meganuclease or zinc finger nuclease, preferably a Cas nuclease. The vector(s) of any one of claims 4-6, wherein the nuclease is operable to cut the nucleic acid at a predetermined sequence motif, optionally a protospacer sequence or restriction site. The vector(s) of claim 7, wherein the nucleic acid comprises a plurality of said motifs. The vector(s) of any preceding claim, wherein P1 is an amino acid, protein or RNA for human or animal therapy. The vector(s) of any preceding claim, wherein P1 is toxic to cells of the same species as the host cell. The vector(s) of any preceding claim, wherein P1 is a transcription or translation regulator in cells of the same species as the host cell. The vector(s) of any preceding claim, wherein R is an amino acid, protein, carbohydrate (optionally a sugar), lipid, metal ion or nucleic acid; or wherein R is a sugar alcohol (optionally xylitol). The vector or any preceding claim, wherein the vector is an ICE (integrative and conjugative element), plasmid (optionally a conjugative plasmid), transduction particle (optionally a phage or nonself-replicative transduction particle) or nanoparticle. The vector(s) of claim 13, wherein the vector comprised by a carrier cell, optionally wherein the vector is a conjugative plasmid comprised by a carrier cell (optionally a bacterial carrier cell) for administration to a microbiota of a human or animal subject. The vector(s) of claim 14, wherein the carrier cell is a cell of commensal or probiotic bacterial cell species of a human or animal microbiota and/or the carrier cell is a cell of a human or animal gut microbiota species. A nucleic acid vector (optionally according to claim 14 or 15) for transfer into a host cell of a microbiota, wherein the vector is comprised by a carrier cell and encodes

(a) a nuclease (optionally an RNA-guided nuclease or restriction endonuclease) that is operable in the carrier cell to cut a chromosome or episome (which is not the vector) of the carrier cell, optionally wherein the chromosome or episome is degraded; and/or

(b) an RNA that is operable in the carrier cell for guiding an RNA-guided nuclease or a precursor of such an RNA, wherein the RNA guides the nuclease to cut a chromosome or episome (which is not the vector) of the carrier cell, optionally wherein the chromosome or episome is degraded; wherein the vector comprises one or more regulatable promoters for regulating expression of the nuclease of (a) and/or the RNA or component of (b) in the carrier cell. The vector of claim 16, wherein the vector comprises an inducible or repressible promoter that regulates expression of the nuclease of (a) and/or the vector comprises an inducible or repressible promoter that regulates expression of the RNA or component of (b), preferably wherein the promoter(s) are inducible promoters. The vector of claim 16 or 17, wherein the guided nuclease is a Cas nuclease, TALEN, meganuclease or zinc finger nuclease, preferably a Cas nuclease. The vector of any one of claims 16-18, wherein the nuclease is the same guided nuclease as recited in claim 5. The vector of any one of claims 16-19, wherein the nuclease is operable to cut the chromosome or episome in the carrier cell at a predetermined sequence motif, optionally a protospacer sequence or restriction site. The vector of any one of claims 16-20, wherein cutting of the carrier cell chromosome or episome kills the carrier cell or reduces growth or proliferation of the carrier cell, preferably wherein the cell is killed. The vector of any one of claim 16-21, wherein the vector comprises an oriT for transfer into the host cell, optionally wherein the vector is a conjugative plasmid. The vector of any one of claims 16-22, wherein

(a) the vector is a conjugative plasmid;

(b) the vector comprises an inducible promoter that regulates expression of the nuclease of (a) and/or the vector comprises an inducible promoter that regulates expression of the RNA or component;

(c) optionally the guided nuclease is a Cas nuclease; and

(d) cutting of the carrier cell chromosome or episome kills the carrier cell or reduces growth or proliferation of the carrier cell. The vector(s) of any preceding claim, wherein the host cell is a cell of a species found in a microbiota (optionally gut microbiota) of humans or animals. The vector(s) of any preceding claim, wherein the host cell is a cell of commensal or probiotic bacterial cell species of a human or animal microbiota. The vector(s) of any one of claims 16 to 25, wherein the species is selected from any species in Table 1, preferably a Bacteroides or Clostridales species. The vector(s) of any preceding claim, wherein

(a) P1 is a protein or RNA for human or animal therapy; and

(b) P2 comprises (i) a crRNA that is operable in the host cell for guiding a Cas nuclease to bind to a protospacer sequence comprised by the nucleic acid for cutting of the protospacer, optionally wherein the nucleic acid is degraded, thereby downregulating the expression of P1, or (ii) a precursor of such an crRNA. A vector or said at least one vector according to any preceding claim for use as a medicament. A host cell comprising nucleic acid that comprises

(a) an expressible nucleotide sequence of interest (NSl) for producing a product of interest (P1) in the host cell; and (b) an expressible nucleotide sequence (NS2) for producing a regulator product (P2) in the host cell, wherein P2 is operable in the host cell to regulate expression or activity of P1; wherein

(c) NS 1 is under the control of a first promoter (optionally a constitutive or inducible promoter) for expression of P1 ; and

(d) expression or activity of P2 in the host cell is regulatable by exposure of the host cell comprising the at least one vector to a regulator agent (R), thereby regulating the expression or activity of P1; wherein the host cell is a bacterial, archaeal or fungal cell. The cell of claim 29, wherein

(a) the nucleic acid is comprised by at least one nucleic acid vector of the cell;

(b) NS1 is comprised by a nucleic acid vector of the cell and NS2 is comprised by a chromosome of the cell;

(c) NS1 is comprised by a chromosome of the cell and NS2 is comprised by a nucleic acid vector of the cell; or

(d) NS1 is comprised by a chromosome of the cell and NS2 is comprised by a chromosome of the cell. The cell of any one of claim 30(a)-(c) wherein the vector or each vector is a conjugative plasmid for transfer to a cell of a microbiota comprised by a human or animal subject. The cell of any one of claims 29-31, wherein the cell is a cell of commensal or probiotic bacterial species of a human or animal microbiota, optionally an E coli cell or aBacteroides cell. The cell of any one of claims 29-32, wherein NS2 is under the control of a second promoter that is regulatable for expression of P2, wherein binding of a regulator agent (R) to the vector nucleic acid regulates the second promoter, thereby regulating the expression of P2 and P1 . The cell of any one of claims 29-33, wherein P2 comprises an RNA-guided nuclease (optionally a Cas nuclease), wherein the nuclease is operable to cut the nucleic acid at a predetermined sequence motif. The cell of claim 34, wherein

(a) the sequence motif is comprised by a chromosome of the cell and the cutting kills the cell; or wherein the sequence motif is comprised by a gene (on a chromosome or vector of the cell) comprising NS 1 for production of P1 and the cutting down-regulates the production of P1; or (b) the sequence motif is comprised by a said vector comprising NS1. The cell of any one of claims 29-35 for treating or preventing a disease or condition in a human or animal subject, wherein the cell is administered to a microbiota (optionally a gut microbiota) of the subject to produce P1 in the subject thereby treating or preventing the disease or condition in the subject. The cell of any one of claims 29-36 comprising a vector according to any one of claims 1-28. A pharmaceutical composition comprising a vector or said at least one vector, or a cell of any preceding claim and a pharmaceutically-acceptable carrier, diluent or excipient, optionally an antacid. A tablet, suppository, pill, capsule, or liquid formulation for administration to the gastrointestinal tract of a human or animal subject, wherein the tablet, suppository, pill, capsule or liquid formulation comprises a vector or said at least one vector, or a cell according to any one of claims 1 to 37. The tablet, pill or capsule of claim 39, wherein the tablet pill, or capsule comprises an enteric coating. The tablet, pill, capsule or liquid formulation of claim 39 or 40 for use as an orally-administered medicament. A method of temporally regulating the production of an expression product in a human or animal subject, the method comprising

(a) administering to a microbiota (optionally a gut microbiota) of the subject at least one vector comprising nucleic acid, wherein the microbiota comprises a host cell and the nucleic acid encodes a product of interest (P1); optionally wherein the administering is oral or topical administration;

(b) allowing transfer of the nucleic acid into the host cell comprised by the microbiota and expression of P1 in the host cell;

(c) after step (b) exposing the microbiota to a regulator agent (R) that upregulates production of an RNA-guided nuclease/guide RNA complex in the host cell that is capable of targeting a protospacer comprised by the nucleic acid, wherein the nuclease cuts the nucleic acid and expression of P1 is rendered non-functional (optionally by degradation of the cut nucleic acid in the cell), wherein the nuclease (or a component thereof) and/or RNA (or a component thereof) is encoded by the nucleic acid. A method of temporally regulating the production of an expression product in a human or animal subject, the method comprising

(a) administering to a microbiota (optionally a gut microbiota) of the subject a vector that encodes a product of interest (P1), said at least one vector wherein said vector(s) encode a product of interest (P1), cell, composition, tablet, suppository, pill, capsule, or liquid formulation according to any one of claims 1-41; optionally wherein the administering is oral or topical administration;

(b) allowing transfer of the nucleic acid into a host cell comprised by the microbiota and expression of P1 in the host cell; and

(c) after step (b) exposing the microbiota to R (such as by administering R to the subject), wherein R regulates the second promoter, thereby regulating the expression of P2 and P1 . A method of

(a) treating or preventing a disease or condition in a human or animal subject by temporally regulating the production of P1 according to the method claim 42 or 43; or

(b) modifying a microbiota (eg, a gut microbiota) of a human or animal subject by temporally regulating the production of P1 according to the method claim 42 or 43. The method of claim 42, 43 or 44, wherein P1 is a therapeutically or prophylactically useful expression product in the subject. The method of any one of claims 42-45, wherein in step (c) P1 expressed from the nucleic acid is the regulator agent (R) or is a component of a pathway that produces R, whereby a P1 expression feedback loop negatively regulates further expression of P1. The method of any one of claims 42-45, wherein R upregulates the second promoter in step (c) and P2 downregulates the expression of P1; and optionally wherein the upregulation of the second promoter causes the production of a guided nuclease or restriction endonuclease that cuts the nucleic acid in the host cell, wherein the nucleic acid is degraded, thereby downregulating the expression of P1. The method of any one of claims 42-45, wherein P2 is capable of upregulating the expression of P1 in the host cell, wherein R downregulates the second promoter in step (c) whereby the expression of P1 is downregulated. The vector, said at least one vector, cell, composition, tablet, suppository, pill, capsule, or liquid formulation according to any one of claims 1-41 for use in the method of any one of claims 42-48, optionally the method of claim 44(a). A cell (optionally according to any one of claims 29-37), comprising a nucleic acid, wherein the nucleic acid comprises a gene encoding a product of interest (P1), the gene comprising a nucleotide sequence (NS1) encoding P1 and a regulatory region 5’ of NS1 that comprises a promoter (Px) for controlling the expression ofNSl, wherein the combination of Px and NS1 is heterologous to the cell and Px is regulatable by xylitol or xylose. The cell of claim 50, wherein the promoter is a xylitol or xylose regulatable promoter of aMorganella species, optionally M morganii. The cell of claim 50 or 51, wherein the promoter comprises SEQ ID NO: 3 or a nucleotide sequence that is at least 70% identical to SEQ ID NO: 3. The cell of any one of claims 50-52, wherein Px is homologous to a xylitol or xylose regulatable promoter of Morganella morganii. The cell of any one of claims 50-53, wherein the cell genome encodes a repressor that is capable of repressing Px, wherein xylitol and/or xylose is capable of de-repressing the repressor. The cell of claim 54, wherein the repressor is encoded by SEQ ID NO: 1 or a nucleotide sequence that is at least 70% identical to SEQ ID NO: 1. The cell of any one of claims 50-55, wherein the cell comprises a xylitol transporter, optionally a xylitol ABC transporter. The cell of any one of claims 50-56, wherein the cell is devoid of a xylitol isomerase gene. A nucleic acid vector comprising a gene as recited in any one of claims 50-57. The vector of claim 58, wherein the vector is a plasmid (optionally a conjugative plasmid), transposon, phagemid or a phage.

Description:
MICROBIOTA ENGINEERING

TECHNICAL FIELD

The invention relates to methods and means for temporally regulating the production of products of interest (eg, proteins or RNAs) in microbiota of subjects, such as gut microbiota of humans or animals. For example, in this way microbiota can usefully be modified for expression of products in a controlled and optionally reversible manner.

BACKGROUND

Mounting evidence undoubtedly links homeostasis in human microbiotas to human health and conversely, microbial dysbiosis is linked to a variety of disease conditions. For example, dysbiosis such as overgrowth of the bacterial pathogen C. difficile in the gut microbiota is linked to severe diarrhoea caused by production of toxins secreted from the C. difficile. Another example of disease related to microbial dysbiosis is inflammatory bowel disease (IBD), in which an inflammatory state of the intestines is at least partly caused by the metabolome associated with overgrowth of Enterobacteriaceae and under representation of bacterial species capable of producing short-chain fatty acids.

Reverting a disease condition by restoring a healthy microbiota by faecal microbial transplants (FMT), in which the faecal microbiota from a healthy donor is transplanted into the gut of the patient suffering from microbial dysbiosis has been proven effective to treat for example recurrent C. difficile infections. However, FMT is a relatively uncontrolled process and also associated with risks as demonstrated by fatalities caused by bacterial infections that could be traced back to a FMT. A more controlled approach in which single strains or consortia of defined strains are added to the dysbiotic gut environment is currently being heavily investigated. Such approaches require the displacement of strains by colonizing new strains added in the consortia

In some cases, target compounds of interest are identified, such as the presence of bacterial toxins (against which antibodies have been generated) or the absence of IL-22 associated with disease. The delivery, however, of relevant proteinaceous compounds locally in the intestinal tract is inefficient and cumbersome due to instability of the proteins in the gut environment.

SUMMARY OF THE INVENTION

Hence, local in situ production of relevant molecules (e.g. enzymes, antibodies, nanobodies, small molecules) intracellularly or secreted by live bacteria which are established and already present in the microbiota would allow targeted intervention with minimal impact on the native microbiota. The invention addresses this need and how to control such production in situ in microbiota.

The invention thus provides the following configurations

In a First Configuration

At least one nucleic acid vector for transfer into a host cell of a microbiota, the vector(s) comprising nucleic acid that comprises

(a) an expressible nucleotide sequence of interest (NS 1) for producing a product of interest (P1) in the host cell; and

(b) an expressible nucleotide sequence (NS2) for producing a regulator product (P2) in the host cell, wherein P2 is operable in the host cell to regulate expression or activity of P1 ; wherein

(c) NS 1 is under the control of a first promoter (eg, a constitutive or inducible promoter) for expression of P1; and

(d) expression or activity of P2 in the host cell is regulatable by exposure of the host cell comprising the at least one vector to a regulator agent (R), thereby regulating the expression or activity of P1.

In an embodiment there is provided: -

At least one nucleic acid vector for transfer into a host cell of a microbiota, the vector(s) comprising nucleic acid that comprises

(a) an expressible nucleotide sequence of interest (NS1) for producing a product of interest (P1) in the host cell; and

(b) an expressible nucleotide sequence (NS2) for producing a regulator product (P2) in the host cell, wherein P2 is operable in the host cell to regulate expression of P1; wherein

(c) NS1 is under the control of a first promoter (eg, a constitutive or inducible promoter) for expression of P1; and

(d) expression of P2 in the host cell is regulatable by exposure of the host cell comprising the at least one vector to a regulator agent (R), thereby regulating the expression P1.

At least one nucleic acid vector for transfer into a host cell of a microbiota, the vector(s) comprising nucleic acid that comprises

(a) an expressible nucleotide sequence of interest (NS1) for producing a product of interest (P1) in the host cell; and (b) an expressible nucleotide sequence (NS2) for producing a regulator product (P2) in the host cell, wherein P2 is operable in the host cell to regulate expression of P1; wherein

(c) NS1 is under the control of a first promoter (eg, a constitutive or inducible promoter) for expression of P1; and

(d) NS2 is under the control of a second promoter that is regulatable for expression of P2, wherein binding of a regulator agent (R) to the vector nucleic acid regulates the second promoter, thereby regulating the expression of P2 and P1.

Preferably, NS1 and NS2 are comprised by the same nucleic acid vector. In another configuration, exposure of a regulator agent (R) to the vector nucleic acid regulates the second promoter, thereby regulating the expression of P2 and P1.

Preferably, P2 is operable in the host cell to bind to vector nucleic acid to regulate expression of P1.

In an embodiment, the invention provides a host cell comprising said at least one vector.

In an embodiment, P1 is a protein (eg, an enzyme) of a metabolic pathway in the host cell, wherein said activity is an activity (eg, enzymatic activity) of P1 in the pathway. In an example, P1 is a protein inhibitor and the activity is inhibitor activity (eg, inhibitor of a component of a metabolic pathway). In an example, P1 is a binding agent, eg, an antibody or antibody fragment, such as a single domain antibody (eg, a nanobody) or an scFv.

In an embodiment, P2 is an inhibitor of P1 expression. In an embodiment, P2 activity is inhibition of P1 expression. In an embodiment, P2 is an enhancer of P1 expression. In an embodiment, P2 activity is enhancement of P1 expression.

In a First Aspect of the First Configuration

A nucleic acid vector for transfer into a host cell of a microbiota, the vector comprising a nucleic acid that comprises

(a) an expressible nucleotide sequence of interest (NSl) for producing a product of interest (P1) in the host cell; and

(b) an expressible nucleotide sequence (NS2) for producing a regulator product (P2) in the host cell, wherein P2 is operable in the host cell to bind to the vector nucleic acid to regulate expression of P1; wherein (c) NS1 is under the control of a first promoter (eg, a constitutive or inducible promoter) for expression of P1; and

(d) NS2 is under the control of a second promoter that is regulatable for expression of P2, wherein binding of a regulator agent (R) to the vector nucleic acid regulates the second promoter, thereby regulating the expression of P2 and P1.

In a Second Aspect of the First Configuration

P2 is expressible in the host cell for the formation of a nuclease that is operable in the host cell to cut the vector nucleic acid, wherein the nucleic acid is degraded, thereby downregulating the expression of P1. This is useful to control the expression of P1 temporally (ie, for a predetermined window of time) in a subject, such as in a human or animal microbiota. For example, in this way microbiota can usefully be modified for expression of P1 in a controlled and optionally reversible manner.

In a Third Aspect of the First Configuration

Each vector (such as according to the First Aspect) is a conjugative plasmid. This is useful to enable spreading of the vector nucleic acid - and thus expression of P1 - within a targeted microbiota. When coupled with the First Aspect of the First Configuration, there is provided a powerful way to controllably modify the extent and timing of P1 expression in the microbiota.

In a Fourth Aspect of the First Configuration

The host cell is a cell of commensal or probiotic bacterial cell species of a human or animal microbiota, preferably a Bacteroides species. Such species are present and maintained in native microbiota of humans and animals and thus, the invention provides means for relatively stable and controllable microbiota modification for temporally regulating the P1 expression.

In a Second Configuration

A method of temporally regulating the production of an expression product in a human or animal subject, the method comprising a) administering to a microbiota (eg, a gut microbiota) of the subject said at least one vector comprising nucleic acid, wherein the microbiota comprises a host cell (eg, bacterial cell) and the nucleic acid encodes a product of interest (P1); optionally wherein the administering is oral or topical administration; b) allowing transfer of the nucleic acid into the host cell comprised by the microbiota and expression of P1 in the host cell; and c) after step (b) exposing the microbiota to a regulator agent (R) that regulates the expression or activity of a regulator product (P2) in the host cell wherein P2 is operable in the host cell to regulate expression or activity of P1, optionally wherein R upregulates production of an RNA-guided nuclease/guide RNA complex in the host cell that is capable of targeting a protospacer comprised by the nucleic acid, wherein the nuclease cuts the nucleic acid and expression of P1 is rendered non-functional (eg, by degradation of the cut nucleic acid in the cell), wherein the nuclease (or a component thereof) and/or RNA (or a component thereof) is encoded by the nucleic acid.

In a First Aspect of the Second Configuration

A method of temporally regulating the production of an expression product in a human or animal subject, the method comprising a) administering to a microbiota (eg, a gut microbiota) of the subject a said vector of the First Aspect of the First Configuration, wherein the microbiota comprises a host cell (eg, bacterial cell) and the nucleic acid encodes a product of interest (P1); optionally wherein the administering is oral or topical administration. b) allowing transfer of the nucleic acid into the host cell comprised by the microbiota and expression of P1 in the host cell; and c) after step (b) exposing the microbiota to a regulator agent (R) that regulates the expression or activity of P2, optionally wherein R upregulates production of an RNA-guided nuclease/guide RNA complex in the host cell that is capable of targeting a protospacer comprised by the nucleic acid, wherein the nuclease cuts the nucleic acid and expression of P1 is rendered non-functional (eg, by degradation of the cut nucleic acid in the cell), wherein the nuclease (or a component thereof) and/or RNA (or a component thereof) is encoded by the nucleic acid.

In a Third Configuration

A method of temporally regulating the production of an expression product in a human or animal subject, the method comprising

(a) administering to a microbiota (eg, a gut microbiota) of the subject said at least one vector of the invention; optionally wherein the administering is oral or topical administration;

(b) allowing transfer of the nucleic acid into a host cell comprised by the microbiota and expression of P1 in the host cell; and

(c) after step (b) exposing the microbiota to R (eg, by administering R to the subject), wherein R regulates the second promoter, thereby regulating the expression of P2 and P1 .

In a First Aspect of the Second or Third Configuration

A method of a) treating or preventing a disease or condition in a human or animal subject by temporally regulating the production of P1 according to the method of the Second or Third Configuration; or b) modifying a microbiota (eg, a gut microbiota) of a human or animal subject by temporally regulating the production of P1 according to the method of the Second or Third Configuration.

In a Fourth Configuration

A nucleic acid vector for transfer into a host cell of a microbiota, wherein the vector is comprised by a carrier cell (eg, a bacterial cell) and encodes a) a nuclease (optionally an RNA-guided nuclease or restriction endonuclease) that is operable in the carrier cell to cut a chromosome or episome (which is not the vector of the invention) of the carrier cell, optionally wherein the chromosome or episome is degraded; and/or b) an RNA that is operable in the carrier cell for guiding an RNA-guided nuclease or a precursor of such an RNA, wherein the RNA guides the nuclease to cut a chromosome or episome (which is not the vector) of the carrier cell, optionally wherein the chromosome or episome is degraded; wherein the vector comprises one or more regulatable promoters for regulating expression of the nuclease of (a) and/or the RNA or component of (b) in the carrier cell.

In a Fifth Configuration

A method of engineering a microbiome, the method comprising contacting the microbiome with a plurality of vectors as described herein and optionally allowing transfer of said vector nucleic acid into target cells of the microbiota.

A modified microbiota obtained or obtainable by the method herein, optionally wherein the microbiota is comprised by a pharmaceutical composition for use as a medicament to treat a disease or condition in a human or animal subject.

In a Sixth Configuration

A host cell comprising nucleic acid that comprises

(a) an expressible nucleotide sequence of interest (NS 1) for producing a product of interest (P1) in the host cell; and

(b) an expressible nucleotide sequence (NS2) for producing a regulator product (P2) in the host cell, wherein P2 is operable in the host cell to regulate expression or activity of P1; wherein

(c) NS1 is under the control of a first promoter (eg, a constitutive or inducible promoter) for expression of P1; and

(d) expression or activity of P2 in the host cell is regulatable by exposure of the host cell comprising the at least one vector to a regulator agent (R), thereby regulating the expression or activity of P1; wherein the host cell is a bacterial, archaeal or fungal cell.

In a Seventh Configuration

In a first Aspect

A cell (optionally according to any other Configuration), comprising a nucleic acid, wherein the nucleic acid comprises a gene encoding a product of interest (P1), the gene comprising a nucleotide sequence (NS1) encoding P1 and a regulatory region 5’ of NS1 that comprises a promoter (Px) for controlling the expression ofNSl, wherein the combination of Px and NS1 is heterologous to the cell and Px is regulatable by xylitol.

In a second Aspect

A cell (optionally according to any other Configuration), comprising a nucleic acid, wherein the nucleic acid comprises a gene encoding a product of interest (P1), the gene comprising a nucleotide sequence (NS1) encoding P1 and a regulatory region 5’ ofNSl that comprises a promoter (Px) for controlling the expression ofNSl, wherein the combination of Px and NS1 is heterologous to the cell and Px is regulatable by xylose.

In a third Aspect

A nucleic acid vector comprising a gene as recited in the first or second Aspect.

BRIEF DESCRIPTION OF FIGURES

Figure 1. plasmid map of pl364. The plasmid is annotated with relative gene locations and their functions. The plasmid contains the functions of plasmid replication (pBBR ori+rep) and RP4 origin of transfer (mob region encompassing oriT and relaxase). araC - regulatory gene pBAD - inducible promoter regulated by the presence of arabinose cas3 - gene encoding E. coli Type I-E Cas3 protein casA-E - Genes encoding Cascade proteins Prham - inducible promoter regulated by the presence of rhamnose

Bba B1003 Terminator - Artificial terminator rmtB 16S rRNA methylase gene. Provides resistance to amikacin and gentamicin sfgfP gene encoding superfolder green fluorescent protein kanR - kanamycin resistance gene mob - mobilization gene. Requered for conjugal transfer of DNA pBBRori - origin of replication pBBR rep - gene encoding replication initiantion protein

CRISPR array - consists of E. coli direct repeats and one spacer

Figure 2. Conjugative transfer of pl364. Transconjugants formed when pl364 is conjugated from the JKE201 strain encoding the RP4 transfer genes. A control lacking the oriT was included.

Figure 3. GFP expression from pl364. The expressing level of GFP from cells harvested at an OD600 of 1. The b52 strain was included as an autofluorescence control.

Figure 4. The self-targeting plasmid is lost rapidly upon CRISPR/cas induction. Proportion of GFP- positive colonies for induced and non-induced b52 containing the pl364 self-targeting plasmid over time.

Figure 5. PCR screening for the pl364 plasmid backbone. Gel electrophoresis showing no presence of the pl364 in all picked GFP negative colonies from the 24h timepoint.

Figure 6. Chromosomal CRISPR/cas targeting allows for rapid removal of plasmid host. The change in host cell viable counts upon induction of CRISPR/cas system targeting the chromosome (IptA) of A. coli MG1655. Error-bars show standard deviation of 3 replicates.

Figure 7. Various schematics illustrating pathways that may be use the invention.

Figure 8: (i) Overview of GFP reporter plasmids with the xylitol-inducible regulatory system, (ii) Depicted is a genomic region from Morganella morganii strain ZJG812 (genome ID: CP064831.1) with structural genes highlighted related to xylitol uptake and metabolism: NAD(P)-dependent alcohol dehydrogenase (IZ184_04875 gene), xylulokinase (xylB), sugar ABC transporter ATP- binding protein (IZ184_04865 gene), ABC transporter permease (IZ184_04860 gene), substratebinding domain-containing protein (IZ184_04855). These genes are expressed from a negatively inducible promoter (promoter) controlled by a LacI family DNA-binding transcriptional regulator (IZ184_04885 gene). A predicted D-lyxose/D-mannose family sugar isomerase (IZ184_04880 gene) is found in-between the promoter and the repressor. Gene annotations were made based on a sequence similarity search against the public database.

Figure 9: Activity of xylitol-inducible promoter in response to xylitol. GFP reporter plasmids encoding the xylitol-inducible promoter system with a full-length transcriptional regulator (pSNP1902) or with a loss-of-function transcriptional regulator (pSNP103) were tested within a bSNP463 strain background in response to increasing amounts of xylitol in the growth medium (LB). Data shown is the fluorescence emission after 24 hours of incubation (normalized, see Example 2, section 3.2.2). Error bars indicate standard deviations based on three biological replicates.

Figure 10: Activity of xylitol-inducible promoter in a minimal medium with increasing concentrations of xylitol. GFP reporter plasmids encoding the xylitol-inducible promoter system with a full-length transcriptional regulator (pSNP1902) was tested within a MG1655 (bSNP230) strain background growing in a minimal medium supplemented with glycerol and increasing amounts of xylitol. Data shown here is the fluorescence emission after 24 hours of incubation (normalized, see Example 2, section 3.2.2). Error bars indicate standard deviations based on three biological replicates.

Figure 11: Growth of reporter plasmid carrying strain in minimal medium with and without glycerol and/or xylitol. The E. coli strain MG1655 (bSNP230) carrying the xylitol-inducible GFP reporter plasmid pSNP1902 was grown for 24 hours in minimal medium supplemented with glycerol (0.4 %(v/v)) with or without xylitol (0.5% (w/v)). Error bars indicate standard deviations based on two biological replicates. The trends from these growth profiles (i.e. higher amounts of xylitol leads to slower growth) was also observed from the growth profiles of the remaining strains.

Figure 12: Activity of the promoter in the presence of the xylitol ABC transporter system. E coli bSNP230 strains containing the GFP reporter plasmid (pSNP1902) either with or without a coresident plasmid containing the xylitol ABC transporter (pSNP1939) were grown for 24-hours in minimal medium supplemented with 0.4% (v/v) glycerol (A) or 0.4% (w/v) glucose (B) in the presence or absence of xylitol. The cell-density adjusted fluorescence emission after 24 hours of incubation are shown. Error bars indicate standard deviations based on two (A) and three (B) biological replicates, respectively. Figure 13: Growth profiles of the ABC transporter-carrying strains on minimal medium in the absence or presence of xylitol. bSNP230 strains containing the GFP reporter plasmid (pSNP1902) either with or without a co-resident plasmid containing the xylitol ABC transporter (pSNP1939) were grown for 24-hours in minimal medium supplemented with 0.4% (v/v) glycerol (A) or 0.4% (w/v) glucose (B) in the presence or absence of xylitol. Error bars indicate standard deviations based on two (A) and three (B) biological replicates, respectively.

DETAILED DESCRIPTION

The invention relates to methods and means for temporally regulating the production of products of interest (herein called P1, eg, proteins or RNAs) in microbiota of subjects, such as gut microbiota of humans or animals. For example, in this way microbiota can usefully be modified for expression of products in a controlled and possibly reversible manner. This can be useful to enable controlled engineering of microbiota of subjects in a way that enables, for example, production of desirable levels of P1 followed by controlled reduction in expression, eg, for reversion to a pre -engineered level of P1. To this end, the invention provides the following illustrative embodiments.

In a First Configuration, there is provided:

At least one nucleic acid vector for transfer into a host cell of a microbiota, the vector(s) comprising nucleic acid that comprises

(a) an expressible nucleotide sequence of interest (NSl) for producing a product of interest (P1) in the host cell; and

(b) an expressible nucleotide sequence (NS2) for producing a regulator product (P2) in the host cell, wherein P2 is operable in the host cell to bind to vector nucleic acid to regulate expression of P1; wherein

(c) NSl is under the control of a first promoter (eg, a constitutive or inducible promoter) for expression of P1 ; and

(d) NS2 is under the control of a second promoter that is regulatable for expression of P2, wherein binding of a regulator agent (R) to the vector nucleic acid regulates the second promoter, thereby regulating the expression of P2 and P1 (eg, R binds an operator that is operatively connected to the second promoter).

Said “at least one vector” may relate to one vector or a plurality of vectors, eg, first and second vectors, eg, 2 vectors. Herein, where features are described in the context of one vector or “the vector”, the skilled person will realise that the features may apply mutatis mutandis to said “at least one vector” such as a first and a second vector, or such as a plurality of vectors. The one vector may be further according to the vector of the Fourth Configuration. Each of said first and second, 2 or plurality of vectors may be further according to the vector of the Fourth Configuration. This is advantageous to limit the presence or spread ofNSl and/or NS2 (thus limiting P1 and/or P2 respectively) in the microbiota or subject comprising the microbiota.

For example, said at least one vector comprises a first vector and a second vector, wherein the first vectosr comprises NS 1 (and optionally not NS2) and the second vector comprises NS2 (and optionally not NS 1) and the vectors are capable of co-existing in the host cell for expression of P1 and P2. Thus, the first vector may comprise NS1 and not NS2; and the second vector may comprise NS2 and not NS1. The first and second vectors are capable of being transferred into the same host cell and co-existing in the cell, whereby P1 and P2 can be expressed in the cell. For example, said at least one vector is one vector that comprises both NS 1 and NS2.

A microbiota may be in any environment, eg, in soil or a waterway, comprised by a plant, or comprised by a human or animal subject. As the skilled person will know, a microbiota may comprise bacteria, archaea, fungi and viruses.

A First Aspect of the First Configuration provides:

A nucleic acid vector for transfer into a host cell of a microbiota, the vector comprising a nucleic acid that comprises a) An expressible nucleotide sequence of interest (NS1) for producing a product of interest (P1) in the host cell; and b) An expressible nucleotide sequence (NS2) for producing a regulator product (P2) in the host cell, wherein P2 is operable in the host cell to bind to the vector nucleic acid to regulate expression of P1;

Wherein c) NS 1 is under the control of a first promoter (eg, a constitutive or inducible promoter) for expression of P1; and d) NS2 is under the control of a second promoter that is regulatable for expression of P2, wherein binding of a regulator agent (R) to the vector nucleic acid regulates the second promoter, thereby regulating the expression of P2 and P1.

As more fully described herein, some advantages of specific aspects of the invention may be

• inducible self-targeting of the vector to remove nucleotide sequence(s) of interest previously spread in a microbiota to which the vector has been exposed. This would leave the mictobiota without a trace of the vector nucleic acid or with reduced prevalence of the vector nucleic acid, such as once a desired expression level of a NS 1 product has been achieved;

® killing the donor (carrier) bacteria by a CRISPR/cas (or other nuclease) vector-bome system that, upon induction, cuts the donor cell genome (eg, chromosome) and kills the donor cells - this being useful to clear the microbiota of donor cells (and thus reduce transmission of the vector in the microbiota) once a desired level of NS1 product expression has been achieved - and also useful as a safety off-switch system to reduce the vector nucleic acid in microbiota and other environments;

® real-time regulation of production of NS1 product; feedback loop(s) can be employed to remove the vector nucleic acid as response to the produced compound or physiological signal(s);

® localized on/off switches to ensure production ofNSl product at certain spatial sites, in an environment, eg, in a microbiota (eg, gut microbiota) of a human or animal subject.

Each host cell may be a bacterial, fungal (eg, yeast) or archaeal cell. Preferably, each host cell is a microbial cell. Preferably, each host cell is a bacterial cell. Preferably, each host cell is an archaeal cell.

Preferably, each host cell is a cell of a commensal or probiotic bacterial cell species found in human or animal microbiota. Preferably, each host cell is a cell of a commensal or probiotic bacterial cell species of a human or animal microbiota. Preferably, each host cell is a microbial (eg, bacterial) cell of a human or animal gut microbiota species.

The nucleic acid may be DNA or RNA. For example, the nucleic acid is DNA.

A host cell with reference to the First to Third Configurations is interchangeably referred to herein as a target cell. A host cell with reference to the Fourth Configurations is interchangeably referred to herein as a carrier or donor cell.

In an example, P1 is secreted from the host cell(s). In an example, P1 is expressed from NS1 in the host cell as a amino acid sequence comprising a signal peptide for secretion of P1 from the cell. In an example, P1 is not secreted from the host cell(s). P1 may be a protein or RNA (eg, a mRNA).

In an embodiment, P1 is not expressed in the microbiota prior to carrying out the method of the invention. P2 may upregulate P1 expression. In an embodiment, P1 is expressed in the subject prior to carrying out the method of the invention, wherein carrying out the method causes at least a 50, 60, 70, 80, 90, 100, 200, 300, 400, 500,600, 700, 800, 900, 1000, 2000, 3000, 4000 or 5000% increase in the expression of P1 in the subject. The increase in expression may be determined by determining the relative levels of P1 in a tissue or fluid sample (eg, blood sample) that has been obtained from the patient. The increase in expression may be determined by determining the relative levels of P1 in a faecal sample that has been obtained from the patient.

P2 may upregulate P1 expression. In an embodiment, P1 is expressed in the microbiota (eg, gut microbiota of a human or animal subject) prior to carrying out the method of the invention, wherein carrying out the method causes at least a 50, 60, 70, 80, 90, 100, 200, 300, 400, 500, 600, 700, 800, 900, 1000, 2000, 3000, 4000 or 5000% increase in the expression of P1 in the microbiota. The increase in expression may be determined by determining the relative levels of P1 in a sample of the microbiota that has been previously obtained from the patient. The increase in expression may be determined by determining the relative levels of P1 in a tissue or fluid sample (eg, blood sample) that has been obtained from the patient. The increase in expression may be determined by determining the relative levels of P1 in a faecal sample that has been obtained from the patient (eg, wherein the microbiota is a gut microbiota).

P2 may downregulate P1 expression. In an embodiment, the method comprises administering Rto the subject in step (c) of the method of the invention, whereby the expression of P1 in the subject is decreased by at least a 50, 60, 70, 80, 90, 100, 200, 300, 400, 500,600, 700, 800, 900, 1000, 2000, 3000, 4000 or 5000% compared to the expression of P1 immediately before carrying out step (c). The decrease in expression may be determined by determining the relative level of P1 in a tissue or fluid sample (eg, blood sample) that has been obtained from the patient immediately before carrying out step (c) and comparing the level with the level of P1 in a similar sample (ie, tissue or fluid sample respectively) obtained after step (c) has been performed (eg, 0.5, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18 19, 20, 21, 22, 23 or 24 hours, or 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13 or 14 days, or 3, 4, 5, 6, 7, or 8 weeks after the commencement of step (c)). The decrease in expression may alternatively be determined by determining the relative levels of P1 in faecal samples that have been obtained from the patient.

P2 may downregulate P1 expression. In an embodiment, the method comprises administering Rto the subject in step (c) of the method of the invention, whereby the expression of P1 is decreased in the microbiota by at least a 50, 60, 70, 80, 90, 100, 200, 300, 400, 500,600, 700, 800, 900, 1000, 2000, 3000, 4000 or 5000% compared to the expression of P1 immediately before carrying out step (c). The decrease in expression may be determined by determining the relative level of P1 in a tissue or fluid sample (eg, blood sample) that has been obtained from the patient immediately before carrying out step (c) and comparing the level with the level of P1 in a similar sample (ie, tissue or fluid sample respectively) obtained after step (c) has been performed (eg, 0.5, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18 19, 20, 21, 22, 23 or 24 hours, or 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13 or 14 days, or 3, 4, 5, 6, 7, or 8 weeks after the commencement of step (c)). The decrease in expression may alternatively be determined by determining the relative levels of P1 in faecal samples that have been obtained from the patient.

The second promoter may be an inducible promoter (wherein induction of promoter causes an increase in P2 expression) or a repressible promoter (wherein repression of promoter causes a reduction in P2 expression). Thus, binding of R to the nucleic acid may induce or repress the second promoter.

For example, a promoter herein is selected from P1ac and Ptac (comprising lacO operator, lacl repressor), Ptet (comprising tetO operator, tetR repressor) and ParaBad (comprising araO operator, araC repressor). As will be familiar to the skilled person, it is possible to synthetically produce repressible promotors from native constitutive promoters by adding the tetO operator (or an analogue thereof) to the sequence and express tetR or analogues to repress them.

P2 may comprise a nucleic acid (eg, an RNA) or a protein (eg, a peptide). For example, P2 is a silencing RNA or protein that is capable of binding to the nucleic acid to inhibit expression of P1, eg, by binding to the first promoter a sequence that overlaps with the first promoter, or by binding to NS1.

P2 may be operable in the host cell to bind to the vector nucleic acid to downregulate expression of P1; and/or R may upregulate expression of P2. P2 may be operable in the host cell to bind to the vector nucleic acid to upregulate expression of P1 .

P2 may be expressible in the host cell for the formation of a nuclease that is operable in the host cell to cut the nucleic acid. Preferably, the cut nucleic acid is degraded (optionally the vector comprising the nucleic acid is degraded), thereby downregulating the expression of P1 in the host cell.

Preferably, the cut nucleic acid is degraded, thereby downregulating the expression of P1 in the microbiota. P2 itself can be the nuclease or a component thereof (wherein the component combines with one or more other components in the host cell to form the nuclease, such as an RNA-guided nuclease). Examples of suitable nucleases are an RNA-guided endonuclease or restriction endonuclease. For example the nuclease is a restriction nuclease selected from Aatll, AbaSI, Acc65I, AccI, Acil, Acll, Acul, Afel, Aflll, Afllll, Agel, AhdI, Alel, Alul, Alwl, AlwNI, Apal, ApaLI, Apol, Asci, Asel, AsiSI, Aval, Avail, Avril, BaeGI, Bael, BamHI, BanI, Banll, BbsI, BbvCI, Bbvl, BccI, BceAI, Bcgl, BciVI, Bell, Bfal, Bgll, Bglll, BlpI, BmgBI, BmrI, Bmtl, Bpml, BpuEI, BpulOI, BsaAI, BsaBI, BsaHI, Bsal, BsaJI, BsaWI, BsaXI, BseRI, BseYI, Bsgl, BsiEI, BsiHKAI, BsiWI, BslI, BsmAI, BsmBI, BsmFI, BsmI, BspCNI, BspEI, BspHI, Bsp 12861, BspMI, BsrBI, BsrDI, BsrFI, BsrGI, BsrI, BssHII, BssSI, BstAPI, BstBI, BstEII, BstNI, BstUI, BstXI, BstYI, BstZ17I, Bsu36I, Btgl, BtgZI, BtsCI, BtsIMutl, BtsI, Cac8I, Clal, CspCI, CviAII, CviKI-1, CviQI, Ddel, Dpnl, Dral, Dralll, DrdI, Eael, EagI, Earl, Ecil, Eco53kl, EcoNI, EcoO109I, EcoP15I, EcoRI, EcoRV, Esp3I, Fatl, Faul, Fnu4HI, FokI, Fsel, FspEI, FspI, Haell, Haelll, Hgal, Hhal, Hindi, Hindlll, Hinfl, HinP1I, Hpal, HphI, HpyAV, HpyCH4III, HpyCH4IV, HpyCH4V, Hpy99I, Hpyl88I, Hpyl66II, Hpyl88III, I-Ceul, I-Scel, KasI, Kpnl, LpnPI, Mbol, MboII, Mfel, MluCI, Mlul, Mlyl, Mmel, Mnll, MscI, Msel, MslI, MspAlI, MspI, MspJI, Mwol, Nad, Narl, Nb.BbvCI, Nb.BsmI, Nb.BsrDI, Nb.BssSI, Nb.BtsI, Neil, Ncol, Ndd, NgoMIV, Nhd, Nlalll, NlalV, NmeAIII, Notl, Nrul, Nsil, NspI, Nt.AlwI, Nt.BbvCI, Nt.BsmAI, Nt.BspQI, Nt.BstNBI, Nt.CviPII, Pad, PaqCI, Pcil, PflMI, PI- PspI, P1-Scd, P1d, P1uTI, Pmd, Pmll, PpuMI, PshAI, Psil, PspGI, PspOMI, PspXI, PstI, Pvul, PvuII, Rsal, RsrII, Sad, Sadi, Sall, SapI, Sau96I, Sbfl, Seal, ScrFI, SexAI, SfaNI, Sfd, Sfil, Sfol, SgrAI, Smal, Smll, SnaBI, Spd, SphI, Srfl, SspI, Stul, StyD4I, Styl, Swal, TaqI, Tfil, Tsd, Tsp45I, TspRI, Tthl 1 II PflFI, Xbal, Xcml, Xhol, Xhol PaeR7I, Xmal, Xmal TspMI, XmnI and Zral.

In an example, P2 comprises a guided nuclease that is programmable in the host cell to guide the nuclease to a target nucleotide sequence comprised by the nucleic acid, wherein the nuclease is capable of cutting the target sequence, whereby the nucleic acid sequence is degraded and P1 expression is inhibited from the nucleic acid.

In an example, the nuclease is operable to cut a target site comprised by the vector nucleic acid, wherein the target site is

(i) comprised by NS 1 ;

(ii) not comprised by NS 1 ;

(iii) comprised by NS2;

(iv) not comprised by NS2;

(v) comprised by a nucleotide sequence of the vector that encodes a CRISPR/cas system or a component thereof (eg, wherein the component is a nucleotide sequence that encodes a Cas, or wherein the component encodes a crRNA or guide RNA; (vi) not comprised by a nucleotide sequence of the vector that encodes a CRISPR/cas system or a component thereof (eg, wherein the component is a nucleotide sequence that encodes a Cas, or wherein the component encodes a crRNA or guide RNA;

(vii) comprised by a nucleotide sequence that encodes a restriction endonuclease;

(viii) not comprised by a nucleotide sequence that encodes a restriction endonuclease;

(ix) comprised by the first promoter; or

(x) comprised by the second promoter.

A CRISPR/cas system comprises at least one Cas (eg, Cas3 (optionally also Cascade Cas, eg, CasA- E), Cas9, Casl2 or Casl3) and a cognate guide RNA that is capable of forming a Cas/guide RNA complex for recognising and binding to a protospacer sequence. In the above examples, the protospacer is comprised by the target site. The guide RNA may be a single guide RNA.

P2 may comprise a) an RNA-guided nuclease; b) an RNA that is operable for guiding an RNA-guided nuclease or a precursor of such an RNA; or c) a restriction endonuclease.

Optionally, the precursor is an RNA, pre-cRNA or tracrRNA.

In an example, P2 is capable of binding to an operator (O) that is comprised by the nucleic acid and operatively connected to the first promoter (eg O is 5’ of the first promoter, eg,withing 200 or 100 kb 5’ of the promoter), wherein when P2 is bound to O the expression of P1 is reduced (eg, eliminated). In an example, P2 comprises a dead Cas nuclease (eg, a dCas9 or dCas3 or dCasl2 or dCasl3) wherein the dCas is capable of forming a dCas/guide RNA complex in the host cell that recognises and binds to a protospacer comprised by the nucleic acid to interrupt expression of P1 (eg, by interrupting promoter function of the first promoter).

Optionally, the guided nuclease is a Cas nuclease, TALEN, meganuclease or zinc finger nuclease, preferably a Cas nuclease. The nuclease may cut DNA or RNA, preferably DNA.

The nuclease may be operable to cut the nucleic acid at a predetermined sequence motif (a target site), optionally a protospacer sequence or restriction site. The protospacer may be a CRISPR/Cas protospacer. The restriction site may be cut by a restriction endonuclease or any other restriction nuclease disclosed herein. The nucleic acid may comprise a plurality (eg, at least 2 or 3, eg, 2, 3, 4, 5, 6, 7, 8, or 9) of said motifs. This may be useful for efficiency of cutting and destruction of the nucleic acid (or vector comprising the nucleic acid). P1 may be an amino acid, protein (eg, peptide or a polypeptide) or RNA (eg, mRNA or silencing RNA) for human or animal therapy. For example P1 is a cytokine, growth factor, enzyme, hormone or antibody (or antibody chain or antibody fragment). For example, P1 is an antibody chain or antibody fragment, eg, a single domain antibody (AKA a dAb) or a nanobody. For example, the chain or fragment is a human antibody chain or fragment. For example, P1 is an antibody heavy chain that forms an antibody with an antibody light chain that is also expressed in the host cell. Preferably, the antibody, chain or fragment is capable of being secreted from the host cell. Preferably, the antibody, chain or fragment is secreted from the host cell. For example, P1 is an incretin, eg, an incretin peptide or a multimer thereof. Optionally, the incretin in selected from GLP-1, GIP, exendin-4 and insulin. For example, P1 comprises an antigen binding site of an antibody or a variable domain (eg, VH and/or VL domain) of an antibody, eg, wherein the antibody is selected from the group consisting of ReoPro™; Abciximab; Rituxan™; Rituximab; Zenapax™; Daclizumab; Simulect™; Basiliximab; Synagis™; Palivizumab; Remicade™; Infliximab; Herceptin™; Mylotarg™; Gemtuzumab; Campath™; Alemtuzumab; Zevalin™; Ibritumomab; Humira™; Adalimumab; Xolair™;

Omalizumab; Bexxar™; Tositumomab; Raptiva™; Efalizumab; Erbitux™; Cetuximab; A vastin™; Bevacizumab; Tysabri™; Natalizumab; Actemra™; Tocilizumab; Vectibix™; Panitumumab; Lucentis™; Ranibizumab; Soliris™; Eculizumab; Cimzia™; Certolizumab; Simponi™; Golimumab, Haris™; Canakinumab; Stelara™; Ustekinumab; Arzerra™; Ofatumumab; Prolia™; Denosumab; Numax™; Motavizumab; ABThrax™; Raxibacumab; Benlysta™; Belimumab; Yervoy™; Ipilimumab; Adcetris™; Brentuximab; Vedotin™; Peseta™; Pertuzumab; pembrolizumab, nivolumab, atezolizumab, Kadcyla™; Ado-trastuzumab; Keytruda™, Opdivo™, Gazyva™ and Obinutuzumab. For example, P is selected from an insulin peptide, incretin peptide or peptide hormone. For example, the antibody is adalimumab. For example, the antibody is pembrolizumab. For example, the antibody is nivolumab. For example, the antibody is atezolizumab. For example, the antibody is dupilumab. For example, the antibody is tocilizumab. For example, the antibody is sarilumab. For example, the antibody is alirocumab. For example, the antibody is evolocumab. In an alternative the antibody is an anti-CD38 antibody, an anti-TNFa antibody, an anti-TNFR antibody, an anti-IL-4Ra antibody, an anti-IL-6R antibody, an anti-IL-6 antibody, an anti-VEGF antibody, an anti- EGFR antibody, an anti-PD-1 antibody, an anti-PD-Ll antibody, an anti-CTLA4 antibody, an anti- PCSK9 antibody, an anti-CD3 antibody, an anti-CD20 antibody, an anti-CD138 antibody, an anti-IL- 1 antibody. In an alternative the antibody is selected from the antibodies disclosed in W02007024715 at page 40, line 23 to page 43, line 23, the disclosure of which is incorporated herein by reference. An antigen herein may be an antigen selected from the group consisting of ABCF1; ACVR1;

ACVR1B; ACVR2; ACVR2B; ACVRL1; ADORA2A; Aggrecan; AGR2; AICDA; AWI; AIG1; AKAP1; AKAP2; AIYIH; AMHR2; ANGPT1; ANGPT2; ANGPTL3; ANGPTL4; ANPEP; APC; APOCI; AR; AZGP1 (zinc-a-glycoprotein); B7.1; B7.2; BAD; BAFF; BAG1; BAI1; BCL2; BCL6; BDNF; BLNK; BLR1 (MDR15); BlyS; BM P1; BMP2; BMP3B (GDFIO); BMP4; BMP6; BM P8; BMPRIA; BMPRIB; BM PR2; BPAG1 (plectin); BRCA1; CI9orflO (IL27w); C3; C4A; C5; C5R1; CANT1; CASP1; CASP4; CAV1; CCBP2 (D6 / JAB61); CCL1 (1-309); CCL11 (eotaxin); CCL13 (MCP-4); CCL15 (MIP-id); CCL16 (HCC-4); CCL17 (TARC); CCL18 (PARC); CCL19 (M IP-3b); CCL2 (MCP-1); MCAF; CCL20 (MIP-3a); CCL21 (MIP-2); SLC; exodus-2; CCL22 (MDC / STC- 1); CCL23 (M PIF-1); CCL24 (MPIF-2 I eotaxin-2); CCL25 (TECK); CCL26 (eotaxin-3); CCL27 (CTACK /ILC) ; CCL28; CCL3 (MIP-la); CCL4 (M IP-lb); CCL5 (RANTES); CCL7 (MCP-3);

CCL8 (mcp-2); CCNA1; CCNA2; CCND1; CCNE1; CCNE2; CCR1 (CKR1 / HM145); CCR2 (mcp- 1RB / RA);CCR3 (CKR3 / CMKBR3); CCR4; CCR5 (CM KBR5 / ChemR13); CCR6 (CMKBR6 / CKR-L3 / STRL22 / DRY6); CCR7 (CKR7 / EBI1); CCR8 (CM KBR8 / TERI / CKR-L1); CCR9 (GPR-9-6); CCRL1 (VSHK1); CCRL2 (L-CCR); CD164; CD19; CD1C; CD20; CD200; CD-22;

CD24; CD28; CD3; CD37; CD38; CD3E; CD3G; CD3Z; CD4; CD40; CD40L; CD44; CD45RB;

CD52; CD69; CD72; CD74; CD79A; CD79B; CD8; CD80; CD81; CD83; CD86; CDH1 (E- cadherin); CDH10; CDH12; CDH13; CDH18; CDH19; CDH20; CDH5; CDH7; CDH8; CDH9;

CDK2; CDK3; CDK4; CDK5; CDK6; CDK7; CDK9; CDKN1A (p2IWapl/Cipl); CDKN1B (p27Kipl); CDKNIC; CDKN2A (pl6INK4a); CDKN2B; CDKN2C; CDKN3; CEBPB; CER1;

CHGA; CHGB; Chitinase; CHST10; CKLFSF2; CKLFSF3; CKLFSF4; CKLFSF5; CKLFSF6;

CKLFSF7; CKLFSF8; CLDN3; CLDN7 (claudin-7); CLN3; CLU (clusterin); CMKLR1; CMKOR1 (RDC1); CNR1; COL18A1; COL1A1; COL4A3; COL6A1; CR2; CRP; CSF1 (M-CSF); CSF2 (GM- CSF); CSF3 (GCSF); CTLA4; CTNNB1 (b-catenin); CTSB (cathepsin B); CX3CL1 (SCYDi) ;

CX3CR1 (V28); CXCL1 (GRO1); CXCLIO (IP-10); CXCL11 (1-TAC / IP-9); CXCL12 (SDF1);

CXCL13; CXCL14; CXCL16; CXCL2 (GR02); CXCL3 (GR03); CXCL5 (ENA-78 I LIX); CXCL6 (GCP-2); CXCL9 (MIG); CXCR3 (GPR9/CKR-L2); CXCR4; CXCR6 (TYMSTR ISTRL33 I Bonzo); CYB5; CYC1; CYSLTR1; DAB2IP; DES; DKFZp451J0118; DNCL1; DPP4; E2F1;

ECGF1; EDG1; EFNAI; EFNA3; EFNB2; EGF; EGFR; ELAC2; ENG; EN01; EN02; EN03; EPHB4; EPO; ERBB2 (Her-2); EREG; ERK8; ESRI; ESR2; F3 (TF); FADD; FasL; FASN; FCER1A;

FCER2; FCGR3A; FGF; FGF1 (aFGF); FGF10; FGF11; FGF12; FGF12B; FGF13; FGF14; FGF16; FGF17; FGF18; FGF19; FGF2 (bFGF); FGF20; FGF21; FGF22; FGF23; FGF3 (int-2); FGF4 (HST); FGF5; FGF6 (HST-2); FGF7 (KGF); FGF8; FGF9; FGFR3; FIGF (VEGFD); FILI (EPSILON); FILI (ZETA); FU12584; FU25530; FLRT1 (fibronectin); FLT1; FOS; FOSL1 (FRA-I); FY (DARC);

GABRP (GABAa); GAGEB1; GAGECI; GALNAC4S-65T; GATA3; GDF5; GFI1; GGT1; GM- CSF; GNAS1; GNRH1; GPR2 (CCRIO); GPR31; GPR44; GPR81 (FKSG80); GRCCIO (CIO); GRP; GSN (Gelsolin); GSTP1; HAVCR2; HDAC4; EDAC5; HDAC7A; HDAC9; HGF; HIF1A; HIP1; histamine and histamine receptors; HLA-A; HLA-DRA; HM74; HM0X1; HUMCYT2A; ICEBERG; ICOSL; 1D2; IFN-a; IFNA1; IFNA2; IFNA4; IFNA5; IFNA6; IFNA7; IFNB1; IFNgamma; TFNW1; IGBP1; IGF1; IGF1R; IGF2; IGFBP2; IGFBP3; IGFBP6; IL-1; IL10; IL10RA; IL10RB; IL11;

IL11RA; IL-12; IL12A; IL12B; IL12RB1; IL12RB2; 1L13; IL13RA1; IL13RA2; 1L14; 1L15;

IL15RA; IL16; 1L17; IL17B; IL17C; IL17R; 1L18; IL18BP; IL18R1; IL18RAP; 1L19; ILIA; IL1B;

IL1F10; IL1F5; IL1F6; IL1F7; IL1F8; IL1F9; IL1HY1; IL1R1; IL1R2; IL1RAP; IL1RAPL1;

IL1RAPL2; IL1RL1;IL1RL2 IL1RN; 1L2; 1L20; IL20RA; IL21R; 1L22; 1L22R; 1L22RA2; 1L23; 1L24; 1L25; 1L26; 1L27; 1L28A; 1L28B; 1L29; IL2RA; IL2RB; IL2RG; 1L3; 1L30; IL3RA; 1L4; IL4R; 1L5; IL5RA; 1L6; IL6R; IL6ST (glycoprotein 130); 1L7; TL7R; 1L8; IL8RA; IL8RB; IL8RB; 1L9; IL9R; ILK; INHA; INHBA; INSL3; INSL4; IRAKI; IRAK2; ITGA1; ITGA2; 1TGA3; ITGA6 (a6 integrin); ITGAV; ITGB3; ITGB4 (b 4 integrin); JAG1; JAK1; JAK3; JUN; K6HF; KAI1; KDR; MTLG; KLF5 (GC Box BP); KLF6; KLK10; KLK12; KLK13; KLK14; KLK15; KLK3; KLK4;

KLK5; KLK6; KLK9; KRT1; KRT19 (Keratin 19); KRT2A; KRTHB6 (hair-specific type II keratin);

LAMA5; LEP (leptin); Lingo-p75; Lingo-Troy; LPS; LTA (TNF-b); LTB; LTB4R (GPR16);

LTB4R2; LTBR; MACMARCKS; MAG or Omgp; MAP2K7 (c-Jun); MDK; M IB1; midkine; M IF; M IP-2; MK167 (Ki-67); MMP2; M MP9; MS4A1; MSMB; MT3 (metallothionectin-ifi); MTSS 1; M UC 1 (mucin); MYC; MYD88; NCK2; neurocan; NFKB 1; NFKB2; NGFB (NGF); NGFR; NgR- Lingo; NgR-Nogo66 (Nogo); NgR-p75; NgR-Troy; NM El (NM23A); NOX5; NPPB; NROB1;

NROB2; NR1D1; NR1D2; NR1H2; NR1H3; NR1H4; NR1I2; NR1I3; NR2C1; NR2C2; NR2E1; NR2E3; NR2F1; NR2F2; NR2F6; NR3C1; NR3C2; NR4A1; NR4A2; NR4A3; NR5A1; NR5A2; NR6A1; NRP1; NRP2; NT5E; NTN4; ODZ1; OPRD1; P2RX7; PAP; PARTI; PATE; PAWR; PCA3; PCNA; PDGFA; PDGFB; PECAM1; PF4 (CXCL4); PGF; PGR; phosphacan; PIAS2; PIK3CG;

PLAU (uPA); PLG; PLXDC1; PPBP (CXCL7); PPID; PR1; PRKCQ; PRKD1; PRL; PROC; PROK2; PSAP; PSCA; PTAFR; PTEN; PTGS2 (COX-2); PTN; RAC2 (p2IRac2); RARB; RGS1; RGS13;

RGS3; RNF110 (ZNF144); ROB02; S100A2; SCGB1D2 (lipophilin B); SCGB2A1 (mammaglobin 2); SCGB2A2 (mammaglobin 1); SCYE1 (endothelial Monocyte-activating cytokine); SDF2;

SERPINA1; SERPINIA3; SERPINB5 (maspin); SERPINE1 (PAT-i); SERPINF1; SHBG; SLA2; SLC2A2; SLC33A1; SLC43A1; SLIT2; SPP1; SPRRIB (Spri); ST6GAL1; STAB1; STAT6; STEAP; STEAP2; TB4R2; TBX21; TCPIO; TDGF1; TEK; TGFA; TGFB1; TGFB1I1; TGFB2; TGFB3;

TGFBI; TGFBR1; TGFBR2; TGFBR3; TH1L; THBSl(thrombospondin-l); THBS2; THBS4; THPO; TIE (Tie-i); T]MP3; tissue factor; TLRIO; TLR2; TLR3; TLR4; TLR5; TLR6; TLR7; TLR8; TLR9; TNF; TNF-a; TNFAIP2 (B94); TNFAIP3; TNFRSF1 1A; TNFRSF1A; TNFRSF1B; TNFRSF21;

TNFRSF5; TNFRSF6 (Fas); TNFRSF7; TNFRSF8; TNFRSF9; TNFSFIO (TRAIL); TNFSF1 1 (TRANCE); TNFSF12 (AP03L); TNFSF13 (April); TNFSF13B; TNFSF14 (HVEM-L); TNFSF1 5 (VEGI); TNFSF1 8; TNFSF4 (0X40 ligand); TNFSF5 (CD40 ligand); TNFSF6 (FasL); TNFSF7 (CD27 ligand); TNFSF8 (CD30 ligand); TNFSF9 (4-1BB ligand); TOLLIP; Toll-like receptors; TOP2A (topoisomerase lia); TP53; TPM 1; TPM2; TRADD; TRAF1; TRAF2; TRAF3; TRAF4; TRAF5; TRAF6; TREM 1; TREM2; TRPC6; TSLP; TWEAK; VEGF; VEGFB; VEGFC; versican; VHL C5; VLA-4; XCL1 (lymphotactin); XCL2 (SCM-lb); XCR1 (GPR5 / CCXCR1); YY1; and ZFPM2.

For example, P1 comprises a VEGF binding site of aflibercept, eg, anti-VEGF FLT1 and/or KDR domain(s).

For example, P1 comprises (or is) an incretin, an insulin peptide, a GLP-1 (glucagon-like peptide- 1 (GLP-1) peptide, a GIP (glucose-dependent insulinotropic polypeptide) peptide, an exendin (eg, exendin-4) peptide, a peptide hormone, a prolactin or prolactin peptide, a ACTH or ACTH peptide, a growth hormone or growth hormone peptide, a vasopressin or vasopressin peptide, an oxytocin or oxytocin peptide, a glucagon or glucagon peptide, a insulin or insulin peptide, a somatostatin or somatostatin peptide, a cholecystokinin or cholecystokinin peptide, a gastrin or gastrin peptide, a leptin or leptin peptide, an antibody binding site (eg, a scFv or Fab) or variable domain thereof, a TCR binding site (eg, a scTCR) or domain thereof, a TCR Vα/Vβ binding site or variable domain thereof, a TCR VΥ/Vδ binding site or variable domain thereof, an antibody single variable domain binding site, or an FcAb binding site. In an example, P1 comprises at least one copy of a GLP-1 and at least one copy of another incretin (eg, an Exendin-4). For example, there is one copy of GLP-1 and one copy of the other increting. Preferably, in the examples in this pargraph, P1 is a secreted or host cell surface-exposed protein. Optionally, any GLP-1 herein is GLP-1 (7-37)-Pro9. Optionally, any incretin herein is an Exendn-4 or Peptide Y. Optionally, any P1 or incretin herein is DURAGLUTIDE™. P1 may comprise an antigen binding site. A binding site herein may, for example, be an antigen (eg, cytokine or growth factor, eg, VEGF or EGFR) binding site of a receptor (eg, KDR or Fit). A binding site herein may, for example, be a binding site of Eyelea™ , A vastin™ or Lucentis™, eg, for ocular or oncological medical use in a human or animal. When the antigen is VEGF, the vector or method may be for treatment or prevention of a cancer or ocular condition (eg, wet or dry AMD or diabetic retinopathy) or as an inhibitor of neovascularisation in a human or animal subject.

For example, P1 is a metabolism pathway component, eg, an enzyme or reagent in the pathway. For example, P1 is an intracellular enzyme in the target cell (ie, host cell). For example, P1 is a secreted enzyme (eg, secreted from the target cell). The pathway herein may be in the target cell or it may be outside the target cell. For example, the pathway is a pathway inside a different cell comprised by the microbiota of which the target cell is a component. The different cell may be a carrier cell or it may be an endogenous cell of the microbiota (ie, any cell of the microbiota except the target cell or a carrier cell). The pathway may comprise one or more as a product or intermediate :-

® a short-chain fatty acid (SCFA)

® a lipid

® an indole derivative or serotonin (eg, wherein the pathway is for the conversion of tryptophan (Trp) to one or more indole derivatives or serotonin)

® a hormones or incretin (eg, GLP-1, oxytocin)

® one or more antigens (eg, to stimulate immune response for vaccination of the subject).

Optionally, R upregulates P2 expression, P2 downregulates P1 expression and optionally P1 is a component in a metabolic pathway (eg, in the microbiota or target cell or in a subject or environment comprising the microbiota or target cell) wherein a production product (X) of the pathway downstream from P1 causes regulation of P1 or P2 expression. See, for example, Scenarios 1, 3 and 4 (Figure 7). In an embodiment, X is R or a precursor of R. In an embodiment, X regulates the first and/or second promoter. In an embodiment, X upregulates P1 or P2 expression. In an embodiment, X upregulates P1 or P2 expression.

Optionally, R upregulates P2 expression, P2 upregulates P1 expression and optionally P1 is a component in a metabolic pathway (eg, in the microbiota or target cell or in a subject or environment comprising the microbiota or target cell) wherein a production product (X) of the pathway downstream from P1 causes regulation of P1 or P2 expression. See, for example, Scenario 2 (Figure 7). In an embodiment, X is R or a precursor of R. In an embodiment, X regulates the first and/or second promoter. In an embodiment, X upregulates P1 or P2 expression. In an embodiment, X upregulates P1 or P2 expression.

In an example, P1 is a secreted or cell-surface exposed protein antigen. This is useful, for example, to vaccinate the subject, eg, where the antigen is an antigen of a pathogen (such as a bacterium or virus, eg, a coat protein, such as spike protein, eg, a SARS-Cov or SARS-Cov-2 or influenza antigen).

In an example, P1 is a secreted antagonist of a target ligand in the subject. For example, binding of P1 to the ligand may inhibit or neutralise the ligand or mark it for destruction in the subject (eg, by immune cells of the subject). For example, the antagonist may comprise an antibody fragment (such as a nanobody or any other antibody single variable domain) comprising a binding site for the target ligand. The ligand may be, for example, ® A curli protein (eg, an E. colt curli protein);

® TMA (trimethylamine);

® A gluten;

® A bile acid;

® Cholesterol or PCSK9; or

® A bacterial toxin (eg, a toxin encoded by pks (eg, E. coli pks), C. difficile toxin, V. cholerae toxin, anthrax toxin or B. fragilis toxin).

In an embodiment, P1 is toxic to cells of the same species as the host cell. This may be useful for killing cells of such species in the microbiota or for reducing the growth or proliferation of such cells, such as when the cells are detrimental to the health of the subject.

In an embodiment, P1 is a transcription or translation regulator in cells of the same species as the host cell. Thus, up- or down-regulation of P1 expression may advantageously act on one or a plurality of genes in the genome of the host cell.

Optionally, R is an amino acid, protein, carbohydrate (eg, a sugar), lipid, metal ion or nucleic acid (eg, RNA). R may be a sugar alcohol, eg, xylitol, glycerol, arabitol, erythritol, isomalt, HSHs, lactitol, maltitol, mannitol or sorbitol, preferably xylitol. R may be an antibiotic. R may be a metabolite of a metabolic pathway that operates in the subject or in the microbiota. R is optionally a metabolite of P1 or a metabolite produced in a pathway comprising P1 . For example, P1 can be a secreted enzyme (ie, secreted from the host cell) that is capable of acting in a pathway in the subject that produces R. For example, P1 can be metabolised in a pathway that produces R (eg, P1 is metabolised to directly produce R, or to indirectly produce R). The pathway may be inside the target host cell or may be outside the target host cell (eg, in a neighbouring cell in the microbiota of which the target cell is a component). The effect of R on P1 expression may be dose dependent (or it may not). The skilled person will readily be able to determine (eg, by way of titrating doses in assays) the appropriate amount of R to use to provide a desired effect on the microbiota.

As exemplified herein, R may be xylitol. Operons of genes related to the conversion/utilization of sugars can be induced by the sugars themselves by virtue of their binding to the relevant transcriptional repressor proteins. For the present study it was decided to investigate a negatively repressible promoter predicted to be induced by xylitol. This 5 -carbon sugar alcohol has several advantages. First of all, xylitol is generally regarded as safe for human consumption by the FDA (Xiang et al., 2021). Furthermore, over half of ingested xylitol is not adsorbed by human cells instead reaching the gastrointestinal tract where it is taken up by the microbiome (Livesey, 2003). Finally, xylitol serves as a metabolite for some bacteria: It is either taken up directly through an ABC-type transporter complex (Madigan et al., 2015) and/or generated through reduction of the corresponding sugar D-xylose. Followingly, xylitol is typically dehydrogenated and phosphorylated to xylulose-5- phosphate which is further catabolized in the pentose phosphate pathway.

For example, R is an amino acid. For example, R is selected from Alanine, Arginine, Asparagine, Aspartic acid, Cysteine, Glutamic acid, Glutamine, Glycine, Histidine, Isoleucine, Leucine, Lysine, Methionine, Phenylalanine, Proline, Serine, Threonine, Tryptophan, Tyrosine and Valine. In an embodiment, R is Tryptophan. For example, R is a protein (eg, a peptide), eg, R is selected from a nisin, sakacin A and sakacin P, For example, R is a bacteriocin. For example, R is a carbohydrate, eg, R is a sugar, such as selected from 1-arabinose, 1 -rhamnose, xylose and sucrose. For example R is a metal ion, eg, R is selected from Fe 2+ , Mn 2+ , Co 2+ , Hg 2+ and Cu 2+ . For example, R is a lipid, eg, proprionate. For example, R is a fatty acid. For example, R is a nucleic acid, eg, R is an RNA. For example, R is a benzene compound, eg, a substituted benzene compound or benzoic acid.

Optionally, the vector is an ICE (integrative and conjugative element), plasmid (eg, a conjugative plasmid), transduction particle (eg, a phage or non-self-replicative transduction particle) or nanoparticle. In an embodiment, the plasmid comprises an oriT and oriV. The plasmid may be self-conjugating. The plasmid may be a shuttle plasmid, ie, a plasmid that can propagate in at least two different host species. The plasmid may comprise a transposon or an ICE (or mobilizable part thereof) that comprises the nucleic acid comprising NS1 and NS2. For example, the transposon or ICE may be a Bacteroides transposon or ICE. The plasmid may comprise an oriT and be mobilisable in the presence of a conjugative system, eg, a system found on other plasmids or integrative and conjugative element (ICE) in the host cell or microbiota. In an example, a such conjugative system is comprised by the genome of the donor cell or carrier cell. In an example, additionally or alternatively, a conjugative system is comprised by the genome of a recipient cell. The conjugative system may be carried on a chromosome or episome of a carrier cell as described herein (ie, system in trans to the plasmid). The conjugative system may be comprised by the plasmid itself (ie, system in cis). The system may be comprised by cells in the microbiota that neighbour the host cell into which the plasmid has been transferred; in this way the plasmid can be spread between neigbouring cells, thereby propagating the plasmid vector in the microbiota. This can be useful to amplify the P1 expression in the microbiota. Similarly, with a self-conjugating plasmid (ie, the conjugative system is provided along with oriT on the plasmid), the plasmid can spread in the microbiota.

The vector may be a conjugative plasmid comprised by a carrier cell (eg, a bacterial carrier cell). For example, the carrier cell and vector is for administration to a microbiota of a human or animal subject. For example, the carrier cell is a cell of commensal or probiotic bacterial cell species of a human or animal microbiota. Additionally or alternatively, the carrier cell is a cell of a human or animal gut microbiota species.

The conjugation genes of conjugative plasmids such as those of the Inc groups: P, N, W, or X show similarity at the protein level to the VirB system of Agrobacterium, itself constituting a prototypic Type IV secretion system (T4SS). These are often smaller and probably the most minimal systems around. The genes necessary for conjugation of the prototypical VirB plasmid pTI and the protein homologs in the incN (somewhat related) and incF (distantly related) plasmid families are shown in Table 4.

Optionally, the conjugation system is a VirB, IncN or IncF conjugation system, or a homologue or orthologue thereof.

Each vector may be a self-conjugative plasmid comprising an oriT and a conjugation system for transferring the plasmid between cells in the microbiota, wherein the conjugation system is a VirB, IncN or IncF conjugation system, or a homologue or orthologue thereof. The carrier (or donor) cell and said plasmid vector(s) may between them comprise a conjugation system for transferring the plasmid between cells in the microbiota, wherein the conjugation system is a VirB, IncN or IncF conjugation system, or a homologue or orthologue thereof, wherein each vector comprises an oriT. The host (or recipient) cell and said plasmid vector(s) may between them comprise a conjugation system for transferring the plasmid between cells in the microbiota, wherein the conjugation system is a VirB, IncN or IncF conjugation system, or a homologue or orthologue thereof, wherein each vector comprises an oriT.

For example, the system is the system of VirB pasmid pTIm, an IncN plasmid or an IncF plasmid, or the system is a homologue or orthologue thereof.

For example, the system comprises (i) VirB genes virB1-11 and virD4, or homologues or orthologues of said genes; (ii) IncN genes traA-G, traJ, traL, traM, traN and traO, or homologues or orthologues of said genes; (iii) Inc F genes traA-E, traG, traH, traK, traL and ORF 196, or homologues or orthologues of said genes; or (iv) IncF genes traA-I, traK, traL, traM, traW and traU (and optionally at least one or all of traX, traN, finO, trbl and trbB), or homologues or orthologues of said genes. For option (iv), reference is made to Front Mol Biosci., 2016 Nov 10;3 :71. doi:

10.3389/fmolb.2016.00071. eCollection 2016, “Comparative Genomics of the Conjugation Region of F-like P1asmids: Five Shades ofF”, Raul Fernandez-Lopez et al.

The invention also provides, according to the Fourth Configuration:

A nucleic acid vector for transfer into a host cell of a microbiota, wherein the vector is comprised by a carrier cell (eg, a bacterial cell) and encodes c) a nuclease (optionally an RNA-guided nuclease or restriction endonuclease) that is operable in the carrier cell to cut a chromosome or episome (which is not the vector of the invention) of the carrier cell, optionally wherein the chromosome or episome is degraded; and/or d) an RNA that is operable in the carrier cell for guiding an RNA-guided nuclease or a precursor of such an RNA, wherein the RNA guides the nuclease to cut a chromosome or episome (which is not the vector) of the carrier cell, optionally wherein the chromosome or episome is degraded; wherein the vector comprises one or more regulatable promoters for regulating expression of the nuclease of (a) and/or the RNA or component of (b) in the carrier cell.

The vector may have any of the vector features disclosed herein. The episome may be a plasmid.

In an embodiment, the vector comprises an inducible or repressible promoter that regulates expression of the nuclease of (a) and/or the vector comprises an inducible or repressible promoter that regulates expression of the RNA or component of (b), preferably wherein the promoter(s) are inducible promoters. In an alternative, the promoter(s) are repressible promoters. In an example, components (a) and (b) are under the control of different promoters. In an example, components (a) and (b) are under the control a common promoter

In an embodiment, the vector comprises an inducible promoter that regulates expression of the nuclease of (a). In an embodiment, the vector comprises an inducible promoter that regulates expression of the RNA or component of (b). Alternatively, the vector comprises an repressible promoter that regulates expression of the RNA or component of (b).

In an embodiment, the vector comprises an repressible promoter that regulates expression of the nuclease of (a). In an embodiment, the vector comprises an inducible promoter that regulates expression of the RNA or component of (b). Alternatively, the vector comprises an repressible promoter that regulates expression of the RNA or component of (b).

Optionally, the guided nuclease is a Cas nuclease, TALEN, meganuclease or zinc finger nuclease, preferably a Cas nuclease. For example, the guided nuclease is a Cas3. For example, the guided nuclease is a Cas9. For example, the guided nuclease is a Casl2 (eg, Casl2a). For example, the guided nuclease is a Cas 13 (eg, Cas 13a).

The nuclease in the Fourth Configuration is the same guided nuclease as in the First Configuration.

The nuclease in the Fourth Configuration is operable to cut the chromosome or episome in the carrier cell at a predetermined sequence motif, optionally a protospacer sequence or restriction site.

Preferably, cutting of the carrier cell chromosome or episome kills the carrier cell or reduces growth or proliferation of the carrier cell, most preferably wherein the cell is killed. This is useful to reduce the transmission of the vector nucleic acid, such as wherein a microbiota or environment has been exposed to the vector. This provides a useful way of regulating expression of products from the vector, such as expression in recipient cells into which the vector nucleic acid has been transferred.

In an example, the vector comprises an oriT for transfer into the host cell, optionally wherein the vector is a conjugative plasmid.

In a preferred embodiment a) the vector is a conjugative plasmid; b) the vector comprises an inducible promoter that regulates expression of the nuclease of (a) and/or the vector comprises an inducible promoter that regulates expression of the RNA or component; c) optionally the guided nuclease is a Cas nuclease; and cutting of the carrier cell chromosome or episome kills the carrier cell or reduces growth or proliferation of the carrier cell.

According to the Fourth Configuration, there is provided:

A nucleic acid vector for transfer into a host cell of a microbiota, wherein the vector is comprised by a carrier cell (eg, a bacterial cell) and encodes a) a nuclease (optionally an RNA-guided nuclease or restriction endonuclease) that is operable in the carrier cell to cut a chromosome or episome (which is not the vector) of the carrier cell, optionally wherein the chromosome or episome is degraded; and/or b) an RNA that is operable in the carrier cell for guiding an RNA-guided nuclease or a precursor of such an RNA, wherein the RNA guides the nuclease to cut a chromosome or episome (which is not the vector) of the carrier cell, optionally wherein the chromosome or episome is degraded; wherein the vector comprises one or more regulatable promoters for regulating expression of the nuclease of (a) and/or the RNA or component of (b) in the carrier cell; wherein cutting of the carrier cell chromosome or episome kills the carrier cell or reduces growth or proliferation of the carrier cell, preferably wherein the cell is killed.

There is also provides a plurality of carrier cells comprising the vector. The cutting may reduce the number of carrier cells of said plurality at least 10 5 , 10 6 or 10 7 “fold, eg, between 10 5 and 10 7 -fold, or between 10 5 and 10 8 -fold or between 10 5 and 10 9 -fold. The skilled person will be familiar with determining fold-killing or reduction in cells, eg, using a cell sample that is representative of a microbiota or cell population. For example, the extent of killing or reduction is determined using a cell sample, eg, a sample obtained from a subject to which the carrier cells of the invention have been administered, or an environmental sample (eg, aqueous, water or soil sample) obtained from an environment (eg, a water source, waterway or field) that has been contacted with the carrier cells of the invention. For example, the cutting reduces the number of carrier cells of said plurality at least 10 5 , 10 6 or 10 7 -fold and optionally the plurality comprises at least 100,000; 1,000,000; or 10,000,000 carrier cells respectively. Optionally, the plurality of carrier cells is comprised by a cell population of the microbiota, wherein at least 5, 6 or 7 log 10 of cells of the population are killed by the cutting, and optionally the plurality comprises at least 100,000; 1,000,000; or 10,000,000 carrier cells respectively. Optionally, the cutting kills at least 99%, 99.9%, 99.99%, 99.999%, 99.9999% or 99.99999% cells of said plurality of carrier cells.

There is provided a method of killing a plurality of carrier cells comprised by a microbiota, the carrier cells comprising vectors of the Fourth Configurations, wherein the method comprises upregulating expression of the nuclease of (a) and/or the RNA or component of (b) in the carrier cells (eg, by inducing the regulatable promoter(s)) whereby the genomes of the carrier cells are cut and the cells are killed. Preferably, chromosomes of the cells are cut. The method optionally kills at least 99%, 99.9%, 99.99%, 99.999%, 99.9999% or 99.99999% cells of said plurality of carrier cells. In an example, the method kills all (or essentially all) of the cells of said plurality of carrier cells. In an example, the method is kills 100% (or about 100%) of the plurality of carrier cells. Preferably all carrier cells comprised by the microbiota are cells of said plurality.

Preferably, at least 90, 91, 92, 93, 94, 95, 96, 97, 98 or 99% of the carrier cells are killed. A conjugative plasmid herein may be a self-conjugative plasmid (ie, wherein the plasmid comrprises an oriT and encodes all proteins required to mobilise the plasmid for conjugative transfer between cells).

For any Configuration herein, the host cell (eg, donor, carrier or recipient cell) is a cell of a species found in a microbiota (eg, gut microbiota) of humans or animals. In a preferred example, the species is a Bacteroides species. In a preferred example, the species is E coli. The host cell may be a cell of commensal or probiotic bacterial cell species of a human or animal microbiota. For example, the species is selected from any species in Table 1, preferably a Bacteroides species (eg, Bacteroides thetaiotaomicron, Bacteroides vulgatus, Bacteroides uniformis or Bacteroides ovatus) or Clostridales species, (eg, Clostridioides difficle or Clostridium disporicum).

In an embodiment, a) P1 is a protein or RNA (eg, a silencing RNA) for human or animal therapy; and b) P2 comprises (i) a crRNA (eg, comprised by a single guide RNA) that is operable in the host cell for guiding a Cas nuclease to bind to a protospacer sequence comprised by the nucleic acid for cutting of the protospacer, optionally wherein the nucleic acid is degraded, thereby downregulating the expression of P1 or (ii) a precursor of such an crRNA (eg, pre-cRNA).

Human or animal therapy herein may be treatment or prophylaxis of a disease or condition in the human or animal.

For any Configuration herein, a crRNA may be comprised by a guide RNA, such as a single guide RNA. For example, the single guide RNA comprises a crRNA and a tracrRNA that are operable in the recipient and/or carrier (donor) cell with a Cas9 to cut a cognate target nucleic acid sequence.

The invention provides:

A vector according to any Configuration herein for use as a medicament.

The medicament may be for treating or preventing a disease in a human or animal subject, eg, when comprised by a formulation for oral-adminstration to the subject.

A pharmaceutical composition comprising a vector according to any Configuration herein and a pharmaceutically-acceptable carrier, diluent or excipient, optionally an antacid. A tablet, suppository, pill, capsule, or liquid formulation for administration to the gastrointestinal tract of a human or animal subject, wherein the table, suppository, pill, capsule or liquid formulation comprises a vector according to any Configuration herein.

Optionally, the tablet pill, or capsule comprises an enteric coating. Optionally, the tablet, pill, capsule or liquid formulation is for use as an orally-administered medicament.

In an example, the composition, tablet, suppository, pill, capsule or formulation herein comprises a medicament selected from the medicaments listed in Table 3.

The invention provides:

A method of temporally regulating the production of an expression product in a human or animal subject, the method comprising a) administering to a microbiota (eg, a gut microbiota) of the subject a vector comprising a nucleic acid, wherein the microbiota comprises a host cell (eg, bacterial cell) and the nucleic acid encodes a product of interest (P1); optionally wherein the administering is oral or topical administration; b) allowing transfer of the nucleic acid into the host cell comprised by the microbiota and expression of P1 in the host cell; c) after step (b) exposing the microbiota to a regulator agent (R) that upregulates production of an RNA-guided nuclease/guide RNA complex in the host cell that is capable of targeting a protospacer comprised by the nucleic acid, wherein the nuclease cuts the nucleic acid and expression of P1 is rendered non-functional (eg, by degradation of the cut nucleic acid in the cell), wherein the nuclease (or a component thereof) and/or RNA (or a component thereof) is encoded by the nucleic acid.

The invention provides:

A method of temporally regulating the production of an expression product in a human or animal subject, the method comprising a) administering to a microbiota (eg, a gut microbiota) of the subject a vector, composition, tablet, suppository, pill, capsule, or liquid formulation according to any preceding claim; optionally wherein the administering is oral or topical administration; b) allowing transfer of the nucleic acid into a host cell comprised by the microbiota and expression of P1 in the host cell; and c) after step (b) exposing the microbiota to R (eg, by administering R to the subject), wherein R regulates the second promoter, thereby regulating the expression of P2 and P1 . Step (b) may be for a desired time between time points Tl and T2. For example for any of the methods, step (a) of the method is commenced at a first time (Tl) and step (c) at a second time (T2). Optionally, T2 is at least 0.5, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18 19, 20, 21, 22, 23 or 24 hours, or 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13 or 14 days, or 3, 4, 5, 6, 7, or 8 weeks after Tl. Preferably, T2 is at least 0.5 hours after Tl. Preferably, T2 is at least 1 day after Tl. Preferably, T2 is at least 1 week after Tl . Preferably, T2 is at least 1 month after Tl . Preferably, T2 is at least 2 months after Tl. Preferably, T2 is at least 3 months after Tl. Preferably, T2 is at least 3 months after Tl . Preferably, T2 is at least 4 months after Tl . Preferably, T2 is at least 5 months after Tl . Preferably, T2 is at least 6 months after Tl . Preferably, T2 is at least 12 months after Tl . Preferably, T2 is at least 18 months after Tl. For example, T2 is 1-6 (eg, 1, 2, 3, 4, 5 or 6) months after Tl. For example, T2 is 1-8 weeks after Tl. Thus, in this way the method is a method of temporally regulating the expression.

There is provided:

A method of i. treating or preventing a disease or condition in a human or animal subject by temporally regulating the production of P1 according to the method of the invention; or ii. modifying a microbiota (eg, a gut microbiota) of a human or animal subject by temporally regulating the production of P1 according to the method of the invention.

For example, there is provided a method of modifying a metabolome of a human or animal subject by carrying out the method of the invention. For example, expression of P1 causes the secreting or sequestering of one or more metabolites in the subject (eg, in the target or cell comprising the vector nucleic acid). For example, expression of P1 causes altering of a pathway intracellularly in the microbiota of the subject (eg, in the target cell) to cause compound metabolism (e.g. for producing a tryptophan sink, such as wherein the tryptophan is used in the microbiota (eg, in the target cell) for the production of AhR (Aryl Hydrocarbon Receptor) ligands). For example, expression of P1 causes the metabolizing or modifying of a chemical, such as a therapeutic drugs, in the microbiota. P1 may do such “causing” since it is a component of a metabolic pathway in the microbiota (eg, in the target cell), such as wherein P1 is a protein, like an enzyme.

The disease or condition may be any disease or condition described herein. The microbiota of a human or animal may be any microbiota described herein. The administration of step (a) may be oral, topical (eg, by application on skin), buccal, rectal, vaginal, parenteral, intravenously, intramuscularly, inhaled, subcutaneously, ocular or intranasal administration. Preferably oral administration is used. Preferably topical administration is used.

In an example, the vector is comprised by a Faecal Microbial Transplat (FMT). The administration of step (a) may be by rectal administration of an enema or FMT comprising the vector.

In step (b) the vector (eg, a plasmid) comprising the nucleic acid can be transferred. P1 may be a therapeutically or prophylactically useful expression product in the subject.

In an embodiment, n step (c) P1 expressed from the nucleic acid is the regulator agent (R) or is a component of a pathway that produces R, whereby a P1 expression feedback loop negatively regulates further expression of P1. For example, P1 is a metabolism pathway enzyme. For example, P1 is an intracellular enzyme in the target cell. For example, P1 is a secreted enzyme (eg, secreted from the target cell). In an example, expression of P1 causes the production (eg, in the microbiota, eg, in the target cell) of R.

A pathway as mentioned herein may be in the target cell or it may be outside the target cell. For example, the pathway is a pathway inside a different cell comprised by the microbiota of which the target cell is a component. The different cell may be a carrier cell (ie, donor cell) or it may be an endogenous cell of the microbiota (ie, any cell of the microbiome except the target cell or the carrier cell).

In an example, R upregulates the second promoter in step (c) and P2 downregulates the expression of P1; and optionally wherein the upregulation of the second promoter causes the production of a guided nuclease or restriction endonuclease that cuts the nucleic acid in the host cell, wherein the nucleic acid is degraded, thereby downregulating the expression of P1.

In an example, P2 is capable of upregulating the expression of P1 in the host cell, wherein R downregulates the second promoter in step (c) whereby the expression of P1 is downregulated.

Downregulation of P1 in step (c) may be by at least 10, 20, 30, 40, 50, 60, 70, 80, 90, or 95% compared to P1 expression in step (b). For example, downregulation of P1 in step (c) may be expression of P1 for a period (PD1) in the presence of Rthat is at least 10, 20, 30, 40, 50, 60, 70, 80, 90, or 95% less compared to P1 expression in step (b) for a period (PD2), wherein PD1 and PD2 are the same length of time. For example, PD1 and PD2 each is 1 minute, 1 hour, 1 day , 1 week, 1 month, 6 months or 12 months. Expression of P1 may be determined in steps (b) and (c) by assessing P1 expression in a sample of the microbiota taken during steps (b) and (c) respectively. For example, when the microbiota is a gut microbiota of a human or animal subject, each sample may be a faecal sample of the subject.

Any upregulation of P1 expression may be an increase in P1 by at least 10, 20, 30, 40, 50, 60, 70, 80, 90, or 95%, such as compared to P1 expression immediately prior step (b). Any downregulation of P1 expression may be an decrease in P1 by at least 10, 20, 30, 40, 50, 60, 70, 80, 90, or 95%, such as compared to P1 expression immediately prior step (b).

Any upregulation of P2 expression may be an increase in P2 by at least 10, 20, 30, 40, 50, 60, 70, 80, 90, or 95%, such as compared to P2 expression immediately prior step (c). Any downreglation of P2 expression may be an decrease in P2 by at least 10, 20, 30, 40, 50, 60, 70, 80, 90, or 95%, such as compared to P2 expression immediately prior step (c).The vector, composition, tablet, suppository, pill, capsule, or liquid formulation described herein may be for use in the method described herein, such as wherein the vector, composition, suppository, pill, capsule or formulation is administered to the subject by oral or topical administration.

The host, carrier and/or target cells may be bacterial cells. In an alternative, instead of bacterial cells, the host, carrier and/or target cells may be archaea.

In an example, the method modifies the genome of the cell, eg, modifies a chromosome or episome (eg, a plasmid) of the host cell. For example, following transfer of the vector nucleic acid into the host cell, a copy of NS 1 is inserted into a chromosome or episome of the host cell. In an example, the expression of a nuclease (eg, P2 or a nuclease that is operable with P2, such as wherein P2 comprises guide RNA) leads to cutting of the NS1 in the chromosome or episome and optionally host cell death.

The carrier cell and target cell may be cells of the same order, family or genus, such as shown in the Examples.

Preferably, the agent comprises a CRISPR/Cas system or component thereof. The agent may be a crRNA or guide RNA that guides a Cas nuclease in the target cell to a target protospacer sequence, wherein the Cas cuts the target sequence and the target cell is killed. For example, the plasmid may encode a plurality of different crRNAs or guide RNAs, such as a first cRNA or gRNA that comprises a spacer sequence that is capable of guiding a Cas in the target cell to a first protospacer sequence and a second cRNA or gRNA that comprises a spacer sequence that is capable of guiding a Cas in the target cell to a second protospacer sequence wherein the protospacer sequences are different (eg, different chromosomal sequences of the target cell). Each protospacer may be comprised by an essential gene, virulence gene or antibiotic resistance gene of the target cell genome. Each protospacer sequence may be from 10 to 60 nucleotides in length, eg, 15 to 50, 15 to 40, 15 to 30 or 15 to 20 nucleotides in length. The target sequence may be a chromosomal sequence of the target cell. The target sequence may be an episomal sequence of the target cell. The plasmid may encode a or said Cas nuclease, optionally a Cas9, Cas3 or Cpfl.

For example, the target cell is comprised by a plant microbiota. The carrier cell may be a Pseudomonas cell, optionally a P fluorescens cell. Optionally, the carrier and target cells are cells of the same genus or species, optionally both are Pseudomonas cells. For example, the target cell is a P syringae or aeruginosa cell and the carrier is a Pseudomonas (eg, P fluorescens) cell.

Preferably, the carrier cells are of a strain or species that is not pathogenic to an organism (eg, a plant, animal or human) that comprises the target cells. The carrier cells may be of a strain or species that is symbiotic or probiotic to an organism (eg, a plant, animal or human) that comprises the target cells, eg, probiotic or symbiotic in the gut of the organism.

For example, the target cell is comprised by a plant microbiota. In an example, the carrier cell comprises a Chitinase class I exoenzyme and/or the carrier cell genome encodes a Chitinase class I exoenzyme. Optionally, the carrier cell in this example is a Pseudomonas, eg, P fluorescens, cell. In an example, the carrier cell comprises a pepl gene. Optionally, the carrier cell in this example is a Pseudomonas, eg, P fluorescens, cell.

In an example, the carrier cell is a motile bacterial cell. Optionally, the the target cell is comprised by a plant microbiota and the carrier cell in this example is a P seudomonas , eg, P fluorescens, cell.

For example, each target cell is a lag phase cell, exponential phase cell or a stationary phase cell. For example, each carrier cell is a lag phase cell, exponential phase cell or a stationary phase cell.

For example, the target cell is comprised by a plant microbiota. Optionally, the target cell is a Pseudomonas (optionally a P fluorescens or P aeruginosa) cell, Erwinia (optionally E carotovora), Xanthomonas, Agrobcaterium, Burkholdi, Clavibacterium, Enterobacteria, Pantoae, Pectobacterium (eg, P atrosepticum), Rhizobium, Streptomyces (eg, S scabies), Xylella (eg, X fastidiosa), Candidatus (eg, C liberibacter), Phytoplasma, Ralstonia (eg, R solanacearum), or Dickeya (eg, D dadantii) cell. Each target cell (eg, the plurality of target cells) may be a cell of a genus or species disclosed in Table 1 or 2. Each target cell (eg, the plurality of target cells) may be comprised by a plant or a plant environment (such as soil) and selected from a genus or species disclosed in Table 1. Each carrier cell (eg, the plurality of carrier cells) may be a cell of a genus or species disclosed in Table 1 or 2.

The method may be carried out in vitro or ex vivo.

The target cell may be comprised by

(a) a plant microbiota (eg, a microbiota of any plant part disclosed herein),

(b) an animal or human microbiota (eg, a microbiota of any human or animal organ or tissue or part disclosed herein); or

(c) a soil, manure, food or beverage microbiota.

For example, the target cell is comprised by a plant leaf, stem, root, seed, bulb, flower or fruit microbiota.

Optionally, a microbiota herein is a gut, lung, kidney, urethral, bladder, blood, vaginal, eye, ear, nose, penile, bowel, liver, heart, tongue, hair or skin microbiota.

For example, the target cell is a cell of a species found in soil.

The method may be carried out using a first cell population comprising a plurality of carrier cells that are contacted with a second cell population comprising a plurality of target cells, wherein copies of said plasmid are conjugatively transferred from carrier cells into target cells, whereby some or all of the cells of the second population are killed or growth or proliferation of cells of the second population is inhibited (eg, by at least 50, 60, 70, 80, 90 or 95%, such as after 5 or 10 hours or 1 day after commencing the method). Preferably cells of the second population are killed.

Optionally, the method of the Second and Third Configurations is carried out on a plurality of target cells comprised by the microbiota by exposing the plurality of target cells to a plurality of the vectors (eg, a plurality of copies of a vector comprising NS 1 and NS2; or a plurality of copies of a first vector comprising NS1 with a plurality of copies of a second vector comprising NS2). The method optionally modifies at least 99%, 99.9%, 99.99%, 99.999%, 99.9999% or 99.99999% cells of said plurality of target cells so that they are capable of expressing P1. In an example, the method is carried out on a population (or said plurality) of said target cells and the method modifies all (or essentially all) of the cells of said population (or said plurality). In an example, the method is carried out on a population (or said plurality) of said target cells and the method modifies 100% (or about 100%) of the cells of said population (or plurality).

Preferably, at least 90, 91, 92, 93, 94, 95, 96, 97, 98 or 99% of the target cells are modified.

Optionally, the target cells are Pseudomonas (eg, P syringae) cells, eg, wherein the cells are comprised by a crop plant, such as a tomoto plant. For example, leaf, fruit, ear, seed, grain, head, pod, stem, trunk, tuber and/or root biomass is increased by the method. For example, leaf or fruit dry biomass, leaf or fruit wet biomass or number of flowers is increased by the method, eg, wherein expression of P1 is beneficial to the health or growth of the plant. For example, average biomass or number is increased over a plurality of plants on which the method of the invention has been practised.

An increase in biomass (eg, average biomass or number) may be an increase by at least 5, 10, 15, 20, 25, 30, 40, or 50% compared to the biomass of plant(s) that have not been exposed to the carrier bacteria, but which comprise the target bacteria. Increases in plant biomass may be determined by measuring the weight of harvested material (eg, fruit, grain, cane, leaves, tubers, nuts or seeds) per area harvested and comparing the measurement of harvested material from plants that have been treated per the invention versus the same area of harvestsed material from plants of the same species and strain grown that have not been treated per the invention, where all plants are grown under the same conditions, eg, in the same field. In some systems units of volume, such as bushels, are used instead of units of weight.

In an Aspect the method is a method of promoting growth of a plant or germination of a plant seed, wherein the method is carried out using a first cell population comprising a plurality of carrier cells that are contacted with a second cell population comprising a plurality of target cells, wherein copies of said nucleic acid are transferred from carrier cells into target cells, whereby some or all of the cells of the second population are modified to express P1, wherein the seed comprises said target cells and said growth or germination is promoted.

Promoting germination may be decreasing the time to onset of germination and/or decreasing the duration of germination. Promoting germination may be increasing the percentage (eg, by at least 5, 10, 15 or 20%) of germination of seeds comprised by a plurality of seeds that are exposed to the carrier cells in the method. Each seed may comprise target cells on the seed surface.

An increase in germination (eg, average germination) in a plurality of seeds exposed to the carrier cells in the method may be obtained, which is an increase by at least 5, 10, 15, 20, 25, 30, 40, or 50% compared to the germination of seeds that have not been exposed to the carrier cells, but which seeds comprise the target bacteria.

The method may be useful for treating pre-emergent seedlings have pathogens present which stop successful germination. Each seedling may comprise target cells on leaves and/or stems of the seedling.

An increase in growth (eg, average growth) in a plurality of seedlings exposed to the carrier cells in the method may be obtained, which is an increase by at least 5, 10, 15, 20, 25, 30, 40, or 50% compared to the growth of seedlings that have not been exposed to the carrier cells, but which seedlings comprise the target bacteria.

In an Aspect the method is a method of increasing leaf chlorophyll (eg, chlorophyll a and/or b) production in a plant, wherein the method is carried out using a first cell population comprising a plurality of carrier cells that are contacted with a second cell population comprising a plurality of target cells, wherein copies of said nucleic acid are transferred from carrier cells into target cells, whereby some or all of the cells of the second population are modified to express P1, wherein the plant comprises said target cells (optionally on leaves and/or stems thereof, or comprised by the apoplast of the plant), whereby target cells are modified and chlorophyll is increased in the plant. Chlorophyll measurement may be measured, for example, by spectrophotometry, high performance liquid chromatography (HPLC) or fluorometry.

In an Aspect the method is a method of modifying target cells comprised by a biofilm, wherein the biofilm is comprised by a subject or comprised on a surface, wherein the biofilm comprises target cells, wherein the method is carried out using a first cell population comprising a plurality of carrier cells that are contacted with a second cell population comprising a plurality of target cells, wherein copies of said nucleic acid are transferred from carrier cells into target cells, thereby modifying the target cells in the biofilm to express P1, optionally wherein the method is carried out ex vivo or in vitro.

The subject may be a human or animal, optionally wherein the surface is a lung surface. The subject may be a plant, optionally wherein the biofilm is comprised by a leaf, trunk, root or stem of the plant.

The surface may be comprised by a domestic or industrial apparatus or container, eg, a fermentation vessel.

There is further provided: -

A carrier bacterial cell (eg, for use in a method according to the invention) for administration to a microbiota comprising a target cell, wherein the carrier cell comprises a conjugative plasmid, the plasmid being a vector of the invention, wherein the carrier cell is capable of conjugating to the target cell wherein the plasmid is transferred into the target cell to modify the target cell to express P1 .

The carrier cell may be any carrier cell or carrier cell disclosed herein. The target cell may be any carrier cell or target cell disclosed herein.

There is provided: -

A carrier cell (eg, a bacterial cell) comprising a vector of the invention. The invention also provides a plurality of such carrier cells (eg, wherein the cells are genetically identical or wherein all of the cells encode the same P1). In an alternative, cells of the plurality encode different P1 proteins.

A pharmaceutical composition comprising a plurality of carrier cells of the invention for administration to a human or animal subject for modifying a plurality of bacterial target cells comprised by the subject to express P1, wherein vectors of the invention (eg, conjugative plasmids) encoding P1 are capable of being introduced from carrier cells into target cells (eg, by conjugation) and P1 is produced in target cells.

Preferably, at least 90, 91, 92, 93, 94, 95, 96, 97, 98 or 99% of the target cells are so modified.

The plurality of target cells may comprise at least 10 7 , 10 8 , 10 9 , 10 10 , 10 11 or 10 12 target cells. For example, the plurality of target cells is comprised by a gut, blood, lung, oral cavity, liver, kidney, bladder, urethra or skin microbiota of the subject.

There is provided:- A method of treating or preventing a disease or condition in a subject, the method comprising contacting the subject (eg, a gut microbiota where the subject is a human or animal) with a composition comprising a plurality of carrier cells of the invention, wherein vectors of the invention (eg, conjugative plasmids) encoding P1 are introduced from carrier cells into target cells (eg, by conjugation) and P1 is produced in target cells, whereby the disease or condition is treated or prevented.

Use of a plurality of carrier cells of the invention in the manufacture of a composition for administration to a subject or environment (eg, soil), for modifying bacterial target cells comprised by the subject or environment to express P1, wherein the target cells are contacted with the carrier cells and vectors of the invention (eg, conjugative plasmids) encoding P1 are introduced from carrier cells into target cells (eg, by conjugation) and P1 is produced in target cells.

For example, the subject is a human or animal. For example, the subject is a mammal. For example, the subject is a bird, fish, protozoan or insect. For example, the animal is a livestock anima. For example, the animal is a dog, cat, horse, cow, sheep or pig.

For example, the subject is a plant and optionally the method comprises contacting the plant (eg, one or more stems and/or one or more leaves of the plant, or the plant apoplast) with the composition comprising a plurality of carrier cells.

There is provided:

Use of a carrier cell of the invention in the manufacture of a composition, for modifying a bacterial target cell ex vivo or wherein the target cell is not comprised by a human or animal (eg, the target cell is comprised by a plant or soil or a human microbiota sample ex vivo), wherein the target cell is contacted with the carrier cell and the carrier cell conjugates to the target cell, whereby the vector nucleic acid is introduced into the target cell, wherein P1 is expressed in the target cell.

For example, each vector is a conjugative plasmid, wherein the carrier cell conjugates (or is capable of conjugating) with the target cell and transfers the plasmid into the target cell by conjugation. Optionally, the plasmid comprises an origin or transer (oriT) and genes for self-conjugation, whereby the plasmid is capable of conjugative transfer from the target cell to a further cell (eg, wherein the target and further cells are comprised by a microbiota). Optionally, the use comprises using a plurality of said carrier cells to modify a plurality of said target cells, wherein the target cells are comprised by a plant or plant environment (eg, soil) and the modifying a) increases (or is for increasing) the biomass of the plant or part thereof (eg, leaf, fruit, ear, seed, grain, head, pod, stem, trunk, tuber and/or root biomass is increased); b) promotes (or is for promoting) germination of one or more seeds of the plant; c) increases (or is for increasing) the amount of leaf chlorophyll of the plant; and/or d) reduces (or is for reducing) a biofilm comprised by the plant, wherein the biofilm comprises target cells (eg, Pseudomonas cells).

Optionally, the target cell or plurality of target cells is in an environment, eg, soil, or in an environment for growing plants. For example, P1 is a plant growth promoter (eg, a fertilizer).

Example Target Cells

For example, each target cell is a Bacteriodes cell, eg, comprised by a human or animal subject. For example, each target cell is a Clostridiales cell, eg, comprised by a human or animal subject.

For example, each target cell is a gram-positive bacterial cell (eg, a Staphylococcus (such as S aureus, eg, methicillin-resistant Staphylococcus aureus (MRSA)), Streptococcus pneumoniae, Clostridium difficile, Enterococcus spp. or Listeria monocytogenes cell). For example, each target cell is a gramnegative bacterial cell (eg, a Acinetobacter baumannii, Escherichia coli, Klebsiella pneumoniae, Stenotrophomonas maltophilia, Campylobacter jejuni, Pseudomonas aeruginosa, Neisseria gonorrhoeae, Vibrio cholerae or Salmonella spp. Cell) For example, each target cell is a cell of a genus or species disclosed in Table 1 herein or Table 2 herein.

Reference is made to Journal of P1ant Pathology (2010), 92 (3), 551-592 Edizioni ETS Pisa, 2010 551, LETTER TO THE EDITOR, “COMPREHENSIVE LIST OF NAMES OF PLANT PATHOGENIC BACTERIA, 1980-2007”, C.T. Bull et al, the disclosure of which is incorporated herein by reference to provide examples of bacterial genera, species and strains of importance to plants and which may be genera, species and strains of target cells of the invention. Examples are disclosed in Table 1 herein.

For example, each target cell is resistant to a fluoroquinolone, P-lactam (eg, methicillin), tetracycline or linezolid antibiotic. For example, each target cell is resistant to vancomycin, eg, wherein the cell is a vancomycin-resistant Enterococcus cell. For example, each target cell is an Azotobacter, Burkholderia, Cupriavidus, Enterococcus, Lysobacter, Paucimonas, Paraburkholderia, Ralstonia, Stenotrophomonas, Variovorax, Xanthomonas or Pseudomonas cell, eg, wherein the target cell is comprised by a plant.

For example, each target cell is an E colt cell.

For example, each target cell is Klebsiella cell, eg, wherein the target cell is comprised by a plant.

For example, each target cell is an Azotobacter, Burkholderia, Cupriavidus, Lysobacter, Paraburkholderia, Ralstonia, Variovorax, Xanthomonas or Pseudomonas cell, eg, wherein the target cell is comprised by a plant.

For example, each target cell is a cell of a Pseudomonas species, optionally wherein the species is selected from Pseudomonas aeruginosa Pseudomonas amygdali, Pseudomonas asturiensis, Pseudomonas avellanae, Pseudomonas cerasi, Pseudomonas chlororaphis, Pseudomonas cichorii, Pseudomonas coronafaciens, Pseudomonas otitidis, Pseudomonas putida, Pseudomonas salegens Pseudomonas savastanoi, Pseudomonas syringae and Pseudomonas viridiflava, eg, wherein the target cell is comprised by a plant.

For example, each target cell is a cell of a species selected from Azotobacter chroococcum, Azotobacter salinestris, Burkholderia ambifaria, Burkholderia cenocepacia, Burkholderia lata, Burkholderia pyrrocinia, Cupriavidus basilensis, Cupriavidus necator, Cupriavidus taiwanensis, Lysobacter gummosus, Paraburkholderia sprentiae, Paraburkholderia terricola, Ralstonia pseudosolanacearum, Ralstonia solanacearum, Variovorax paradoxus, Xanthomonas arboricola, Xanthomonas axonopodis, Xanthomonas campestris Xanthomonas citri, Xanthomonas euvesicatoria and Xanthomonas perforans, eg, wherein the target cell is comprised by a plant.

For example, each target cell is a Stenotrophomonas, Enterococcus, Paucimonas or Pseudomonas cell, eg, wherein the target cell is comprised by a plant.

For example, each target cell is a cell of a Pseudomonas species, optionally wherein the species is selected from Pseudomonas amygdali, Pseudomonas asturiensis, Pseudomonas avellanae, Pseudomonas cerasi, Pseudomonas chlororaphis, Pseudomonas cichorii, Pseudomonas coronafaciens, Pseudomonas putida, Pseudomonas savastanoi, Pseudomonas syringae and Pseudomonas viridiflava, eg, wherein the target cell is comprised by a plant. For example, each target cell is a cell of a species selected from Stenotrophomonas rhizophila, Enterococcus faecalis, Paucimonas lemoignei, Pseudomonas amygdali, Pseudomonas asturiensis, Pseudomonas avellanae, Pseudomonas cerasi, Pseudomonas chlororaphis, Pseudomonas cichorii, Pseudomonas coronafaciens, Pseudomonas putida, Pseudomonas savastanoi, Pseudomonas syringae and Pseudomonas viridiflava, eg, wherein the target cell is comprised by a plant.

Example Carrier Cells

For example, the carrier is an E coli cell (eg, E coli , K12, Nissle or S17 cell), eg, wherein the cell is for adiminstration to a human or animal subject, such as to treat or prevent a disease or condition. For example, each carrier cell is an Bacteroides cell, eg, wherein the cell is for adiminstration to a human or animal subject, such as to treat or prevent a disease or condition. For example, each carrier cell is an Clostridiales cell, eg, wherein the cell is for adiminstration to a human or animal subject, such as to treat or prevent a disease or condition.

For example, each carrier cell is a gram-positive bacterial cell. For example, each carrier cell is a gram -negative bacterial cell. For example, the carrier cell is a cell of a genus or species disclosed in Table 1 ofWO2017211753 (the disclosure of this table and each genus and species individually being incorporated herein for disclosure of cell genus or species that may be used in the present invention).

For example, the carrier cell is a cell of phylum Proteobacteria, class Gammaproteobacteria, order Pseudomonadales or family Pseudomonadaceae. In a preferred example, the carrier is a Pseudomonas (eg, P fluorscens) cell, eg, wherein the target cell is comprised by a plant.

For example, each carrier cell is Klebsiella cell, eg, wherein the target cell is comprised by a plant.

For example, the carrier is a gram positive cell, eg, a Bacillus (such as Bacillus subtilis) or Cloistridiales (such as Clostridium butyricum) cell.

In an example, the subject is a shellfish. The shellfish may be selected from shrimp, crayfish, crab, lobster, clam, scallop, oyster, prawn and mussel.

The subject may be any subject disclosed herein. The subject may be an animal, such as a livestock animal, eg, a bird (such as a poultry bird; or a chicken or a turkey) or swine,

In an alternative, the subject is a plant, eg, and the target bacteria are plant pathogen bacteria. In an example, the target baceteria are Pseudomonas, eg, P syringae or P aeruginosa. In an alternative, the carrier and target cells are archaeal cells. For example the target cells are methanobacterium cells. For example the target cells are methanogen cells. For example, the target cells comprise one or more species of cell selected from:

Methanobacterium bryantii

Methanobacterium formicum

Methanobrevibacter arboriphilicus

Methanobrevibacter gottschalkii

Methanobrevibacter ruminantium

Methanobrevibacter smithii

Methanococcus chunghsingensis

Methanococcus burtonii

Methanococcus aeolicus

Methanococcus deltae

Methanococcus jannaschii

Methanococcus maripaludis

Methanococcus vannielii

Methanocorpusculum labreanum

Methanoculleus bourgensis (Methanogenium olentangyi & Methanogenium bourgense)

Methanoculleus marisnigri

Methanoflorens stordalenmirensis

Methanofollis liminatans

Methanogenium cariaci

Methanogenium frigidum

Methanogenium organophilum

Methanogenium wolfei

Methanomicrobium mobile

Methanopyrus kandleri

Methanoregula boonei

Methanosaeta concilii

Methanosaeta thermophila

Methanosarcina acetivorans

Methanosarcina barkeri

Methanosarcina mazei

Methanosphaera stadtmanae

Methanospirillium hungatei • Methanothermobacter defluvii (Methanobacterium defluvii)

• Methanothermobacter thermautotrophicus (Methanobacterium thermoautotrophicum)

• Methanothermobacter thermoflexus (Methanobacterium thermoflexum)

• Methanothermobacter wolfei (Methanobacterium wolfei)

• Methanothrix sochngenii

Optionally, the target cells are not pathogenic to the subject, for example when the method is a nonmedical method. In an example, the method is a cosmetic method.

In the example, optionally the subject or animal is a livestock animal, such as a cow, sheep, goat or chicken (preferably a cow). Optionally, eg, wherein the subject is an animal (eg, a livestock animal or a wild animal), the target cells are zoonotic bacterial cells, such as cells of a species selected from Bacillus anthracis, Mycobacterium bovis (eg, wherein the animal is a cow), Campylobacter spp (eg. wherein the animal is a poultry animal), Mycobacterium marinum (eg. wherein the animal is a fish), Shiga toxin-producing E. coli (eg. wherein the animal is a ruminant), Listeria spp (eg, wherein the animal is a cow or sheep), Chlamydia abortus (eg, wherein the animal is a sheep), Coxiella burnetii (eg, wherein the animal is a cow, sheep or goat), Salmonella spp (eg, wherein the animal is a poultry animal), Streptococcus suis (eg, wherein the animal is a pig) and Corynebacterium (eg, C ulcerans) (eg, wherein the animal is a cow).

In an example, a plurality of carrier cells as described herein (eg, carrier cells of any configuration, aspect, example or embodiment described herein) is administered to the subject, wherein the carrier cells comprise the nucleic acid encoding P1 .

In an example, each animal is a chicken. In an example, each animal is a cow (eg, a beef or dairy cow).

Optionally the method modifies target cells in the gastrointestinal tract of the subject human or animal; optionally the method modifies target cells in the jejunum, ileum, colon, liver, spleen or caecum of the subject; optionally wherein the animal is a bird and the method modifies target cells in the caecum of the bird. In an example the method is carried out on a group (optionally a flock or herd) of animals, wherein some or all of the animals comprise target cells.

Optionally, the plasmid comprises a RP4 origin of transfer (oriT). The plasmid may be any type of plasmid disclosed herein. P2

P2 may be any antibacterial agent disclosed herein or a component thereof, preferably a guided nuclease that is programmed to cut one or more target sequences in target cells. A suitable nuclease may be a TALEN, meganuclease, zinc finger nuclease or Cas nuclease. For example, the agent comprises one or more components (eg, a Cas nuclease and/or a guide RNA or a crRNA) of a CRISPR/Cas system that is operable in a target cell to cut a protospacer sequence comprised by the target cell (eg, comprised by the vector, such that cut vector is degraded in the target cell). For example, the system is operable to cut at least 2 or 3 different protospacer sequences comprised by the vector of the invention. Optionally, P2 is operable to cut a plurality of different protospacer sequences comprised by the vector (and optionally further operable to cut the carrier cell genome, eg, a carrier cell chromosomal or episomal protospacer sequence, wherein cutting thereof is lethal to the carrier cell - as explained elsewhere, this is useful to reduce or remove carrier cells from the subject or microbiota when desired, such as after a desired level of P1 expression in target cells has been obtained). Optionally, the agent comprises one or more components of a CRISPR/Cas system that is operable to cut at least 2, 3, 4, 5, 6, 7, 8, 9, or 10 different protospacer sequences comprised by the vector and/or carrier cell genome (eg, comprised by the carrier cell chromosome).

In an embodiment, P2

(a) comprises a guided nuclease that is capable in a target cell of recognising and cutting a protospacer sequence comprised by the vector; and/or

(b) encodes a guide RNA or crRNA of a CRISPR/Cas system that operates with a Cas nuclease in a target cell to cut a protospacer sequence comprised by the vector; wherein the nuclease is capable of cutting the vector in the target cell and the vector is degraded in the target cell.

This is useful as an “off switch” to remove the vector from the target cell, eg, for downregulating the capability of P1 expression in a plurality of target cells that have been previously contacted with carrier cells and into which target cells vectors of the invention have been transferred for P1 expression. For example, this usefully can be used to clear the microbiota of a subject so that it can produce less or no P1 after cutting of vector nucleic acid has taken place. In this way, for example, it is possible in a first step to modify a microbiota of a subject (eg, a human gut microbiota) so that it produces P1 (eg, where P1 expression is useful for treatment or prevention of a disease or condition or alternatively for non-medical utility) and thereafter P1 expression can be reduced or removed and the microbiota can be restored to a partially modified or unmodified condition. Any administration of cells to a subject herein may be by oral administration. Any administration of cells to a subject herein may preferably be by administration to the GI tract. Any administration of cells to a subject herein may be by systemic, intranasal or inhaled administration.

There is provided the following definitions:-

Homologue: A gene, nucleotide or protein sequence related to a second gene, nucleotide or protein sequence by descent from a common ancestral DNA or protein sequence. The term, homologue, may apply to the relationship between genes separated by the event of or to the relationship between genes separated by the event of genetic duplication.

Orthologue: Orthologues are genes, nucleotide or protein sequences in different species that evolved from a common ancestral gene, nucleotide or protein sequence by speciation. Normally, orthologues retain the same function in the course of evolution.

Optionally, each P2 is a guide RNA. Optionally, each vector (eg, plasmid) encodes a plurality of guide RNAs or crRNAs of a CRISPR/Cas system wherein the guide RNAs or crRNAs are operable with Cas nuclease in the target cell to recognise a plurality of protospacer sequences comprised by the vector and/or carrier cell genome, optionally wherein the protospacer sequences comprise one or more nucleotide sequences of genes selected . In an example, the vector additionally or alternatively encodes a Cas, eg, a Cas9, Cas3, Cpfl, Casl2, Casl3, CasX or CasY.

In an embodiment, a Cas herein is a Type I, II, III, IV, V or VI Cas, preferably a Type I or II Cas.

In an example, the vector also encodes a Cas3 and cognate Cascade proteins (eg, CasA, B, C, D and E). Optionally, the Cas (and Cascade of present) are E coli Cas (and Cascade).

The plasmid may comprise one or more CRISPR spacers, wherein each spacer consists of 20-40, 25- 35, or 30-35 consecutive nucleotides, eg, consecutive nucleotides selected from

Optionally, the plasmid comprises a RP4 origin of transfer (oriT) and/or a pl5A origin of replication.

In an example, the plasmid is a conjugative phagemid.

In an example, the vector encodes a Cas3 and optionally one or more Cascade proteins (eg, one or more of CasA, B, C, D and E). In an embodiment, the vector encodes a Cas3 and CasA, B, C, D and E. In an embodiment, the vector encodes an E coli Cas3 and CasA, B, C, D and E. Optionally, the guided nuclease (eg, Cas3) is a Type I-A, -B, -C, -D, -E, -F or -U Cas. In an example, P2 in any configuration, aspect, example, option or embodiment herein comprises one or more components of a CRISPR/Cas system that is operable in the target cell to cut a protospacer sequence comprised by the vector. Additionally, in an example, P2 in any configuration, aspect, example, option or embodiment herein comprises one or more components of a CRISPR/Cas system (eg, the same system as in the first sentence in this paragraph) that is operable in the carrier cell to cut a protospacer sequence comprised by the carrier cell genome (eg, a chromosomal or episomal sequence whose cutting is lethal to the carrier cell). This is useful to clear the carrier cell from the subject (eg, from a gut microbiota) when expression of P1 is no longer required.

In an example, the system is operable to cut at least 3 different protospacer sequences comprised by the vector or carrier cell genome.

In an example, the vector

(a) encodes a guided nuclease that is capable of recognising and modifying a carrier cell nucleic acid sequence, wherein the sequence is comprised by an endogenous chromosome or episome of the carrier cell, wherein the nuclease cuts the chromosome or episome to kill the carrier cell or inhibit the growth or proliferation of the target cell; and/or

(b) encodes a guide RNA or crRNA of a CRISPR/Cas system that operates with a Cas nuclease in the carrier cell to cut a protospacer sequence comprised by the cell.

The expression of (a) and/or (b) may be inducible by exposure of the carrier cell to a regulator agent, eg, R. For example, exposure of the carrier and target cells to R may induce production of P2, wherein P2 comprises component (a) and/or (b) whereby vector nucleic acid is cut in the target cell and the genome of the carrier cell is also cut. This kills the carrier cell and leads to degradation of the vector in the target cell. This usefully can then clear the subject of the vector and the carrier cell, eg after a desirable amount of P1 has been expressed in the subject.

Optionally, the Cas, Cascade proteins, gRNAs and crRNAs are E. colt K12 (MG1655) Cas, Cascade proteins, gRNAs and crRNAs respectively. Optionally, the vector is devoid of nucleotide sequences encoding Casl and Cas2 proteins.

In embodiments, by action of P2 (eg, components (a) and/or (b)) in the carrier cells, growth or proliferation of carrier cells is reduced (eg, by at least 40, 50, 60, 70, 80, or 90% compared to growth in the absence of P2 therein). For example, the carrier cells may be comprised by a medicament for treating or preventing a disease or condition in a human or animal; a growth promoting agent for administration to animals for promoting growth thereof; killing zoonositic bacteria in the animals; for administration to livestock as a pesticide; a pesticide to be applied to plants; or a plant fertilizer.

An advantage may be that the carrier cells may be used as producer cells in which vectors of the invention can be replicated (eg, before (eg, in vitro) and/or following administration to the subject).

Example Plasmids

A method of delivery of a vector can be by bacterial conjugation, a natural process whereby a donor bacterium (carrier bacterium) transfers plasmid DNA from itself to a recipient bacterium (target bacterium). Donor bacteria elaborate a surface structure, the pilus which can be considered to be like a syringe or drinking straw through which the DNA is delivered. The donor pilus binds to the surface of a receptive recipient and this event triggers the process of DNA transfer. P1asmids are suitable for this conjugative process, where the plasmid comprises DNA enoding the agent of the invention.

DNA transfer by conjugation may only take place with a ‘susceptible recipient’ but does not generally occur with a recipient carrying a similar type of plasmid. Because conjugation is via pilus bridge, it is possible for that bridge to attach itself not to a recipient but to the donor bacterium. This could result in a futile cycle of transfer of the plasmid DNA to itself. P1asmids thus naturally encode incompatibility factors. One is a surface arrayed protein that prevents the pilus binding to bacterium displaying that surface protein such as itself or any other bacterium carrying the same plasmid.

Additionally, plasmids naturally encode another incompatibility system that closely regulates the copy number of the plasmid inside a bacterium. Thus, should a conjugation event manage to evade surface exclusion and start to transfer DNA by conjugation, the recipient will prevent that plasmid establishing as it already maintains the current copy number and will not accept and maintain a further unwanted additional copy.

In an example of the invention, the plasmid is a member of a plasmid incompatibility group, wherein the target cell does not comprise a plasmid of said group. Optionally, the plasmid of the invention is a member of the incompatibility group P (ie, the plasmid is an incP plasmid). For example within the Enterobacteriaceae the following is a non-exclusive list of potential plasmids that could use for delivery: IncFI, IncFII, IncFIll, IncFIV, IncFV, IncM, Inc9, InclO, Incl, IncA, IncB, IncC, IncH, Incla, Incllc, IncI2, Indy, IncJ, IncL, IncN, Inc2e, IncO, IncP, IncS, IncT and/or IncW. Thus, optionally, the target cell is an Enterobacteriaceae cell and the vector of the invention is a plasmid, wherein the plasmid is selected from an IncFI, IncFII, IncFIll, IncFIV, IncFV, IncM, Inc9, InclO, Incl, IncA, IncB, IncC, IncH, Incla, Indic, Indi, IncI2, Indy, IncJ, IncL, IncN, Inc2e, IncO, IncP, IncS, IncT and IncW plasmid.

Preferably, the subject is a human or animal and the plasmid is an Incl plasmid, eg, an Indi mlncl2 plasmid.

In an example, the carrier cell of the invention comprises two or more plasmids, each plasmid comprising a DNA that encodes a respective P1 and P2 (P1/P2 may be the same in the cells or the cells may comprise different P1 and/or P2). Optionally, a first of said plasmids is a member of a first incompatibility group, wherein the target cell does not comprise a plasmid of said first group, and wherein a second of said plasmids is a member of a second incompatibility group, wherein the target cell does not comprise a plasmid of said second group. For example, a carrier cell may comprise an incP plasmid encoding P1 and P2 (eg, a CRISPR-Cas system or a component thereof (eg, encoding a first crRNA or guide RNA that targets a first protospacer sequence of the vector)) and wherein the carrier cell further comprises an incFl plasmid (eg, encoding P1 and P2 (eg, an anti -carrier cell CRISPR-Cas system or a component thereof (eg, encoding a second crRNA or guide RNA that targets a protospacer sequence of the carrier cell genome)). The protospacers may comprise different nucleotide sequences. Optionally, the carrier cell comprises a group of plasmids comprising 2, 3, 4, 5, 6 or more different types of plasmid, wherein each plasmid is capable of being conjugatively transferred into a target cell, wherein the plasmids encode different P1 and/or P2 products. For example, the plasmids encode different cRNAs or gRNAs that target different protospacers comprisesd by the vector, carrier cell and/or target cell genome. For example, the group of plasmids comprises up to n different types of plasmid, wherein the plasmids are members of up to n different incompatibility groups, eg, groups selected from IncFl, IncFII, IncFIll, IncFIV, IncFV, IncM, Inc9, InclO, Incl, IncA, IncB, IncC, IncH, Incla, Incllc, IncI2, Indy, IncJ, IncL, IncN, Inc2e, IncO, IncP, IncS, IncT and IncW. For example, n=2, 3, 4, 5, 6, 7, 8, 9 or 10.

For example, the carrier cell comprises (i) a first vector (eg, plasmid) that encodes a P2 which is a first type of CRISPR/Cas system that targets a first protospacer comprised by the vector, or encodes a component of said system; and (ii) a second vector (eg, plasmid) that encodes a P2 which is a second type of CRISPR/Cas system that targets a second protospacer comprised by the vector or carrier cell genome (eg, a carrier cell chromosomal sequence), or encodes a component of said system, wherein the first and second types are different. For example, the first type is a Type I system, and the second type is a Type II system (eg, the first vector encodes a Cas3, Cascade and a crRNA or guide RNA that is operable with the Cas3 and Cascade in the target cell to modify the first protospacer; and the second vector encodes a Cas9 and a crRNA or guide RNA that is operable with the Cas9 in the target cell to modify the second protospacer). In an alternative, the Cas3 and Cascade are encoded by endogenous target cell genes, wherein the first vector encodes the crRNA or guide RNA that is operable with the endogenous Cas3 and Cascade in the target cell to modify the first protospacer. In an alternative, the Cas9 is encoded by an endogenous target cell gene, wherein the second vector encodes the crRNA or guide RNA that is operable with the endogenous Cas9 in the target cell to modify the second protospacer. Optionally, the Cas3 and Cascade are encoded by endogenous genes of the target cell and the Cas9 is encoded by the second vector.

Instead of a Type I and Type II system, the invention alternatively provides in an embodiment a first vector (eg, plasmid) encoding a Type I CRISPR/Cas system (or component thereof, eg, a Cas3 or a crRNA or a gRNA) and a second vector (eg, plasmid) encoding a Type III CRISPR/Cas system (or a component thereof). Instead of a Type I and Type II system, the invention alternatively provides in an embodiment a first vector encoding a Type I CRISPR/Cas system (or component thereof) and a second vector encoding a Type IV CRISPR/Cas system (or a component thereof). Instead of a Type I and Type II system, the invention alternatively provides in an embodiment a first vector encoding a Type I CRISPR/Cas system (or component thereof) and a second vector encoding a Type V CRISPR/Cas system (or a component thereof). Instead of a Type I and Type II system, the invention alternatively provides in an embodiment a first vector encoding a Type I CRISPR/Cas system (or component thereof) and a second vector encoding a Type VI CRISPR/Cas system (or a component thereof).

Instead of a Type I and Type II system, the invention alternatively provides in an embodiment a first vector encoding a Type II CRISPR/Cas system (or component thereof, eg, a Cas9 or a crRNA or a gRNA) and a second vector encoding a Type III CRISPR/Cas system (or a component thereof). Instead of a Type I and Type II system, the invention alternatively provides in an embodiment a first vector encoding a Type II CRISPR/Cas system (or component thereof) and a second vector encoding a Type IV CRISPR/Cas system (or a component thereof). Instead of a Type I and Type II system, the invention alternatively provides in an embodiment a first vector encoding a Type II CRISPR/Cas system (or component thereof) and a second vector encoding a Type V CRISPR/Cas system (or a component thereof). Instead of a Type I and Type II system, the invention alternatively provides in an embodiment a first vector encoding a Type II CRISPR/Cas system (or component thereof) and a second vector encoding a Type VI CRISPR/Cas system (or a component thereof).

Instead of a Type I and Type II system, the invention alternatively provides in an embodiment a first vector encoding a Type V CRISPR/Cas system (or component thereof, eg, a Casl2a or a crRNA) and a second vector encoding a Type III CRISPR/Cas system (or a component thereof). Instead of a Type I and Type II system, the invention alternatively provides in an embodiment a first vector encoding a Type V CRISPR/Cas system (or component thereof) and a second vector encoding a Type IV CRISPR/Cas system (or a component thereof). Instead of a Type I and Type II system, the invention alternatively provides in an embodiment a first vector encoding a Type V CRISPR/Cas system (or component thereof) and a second vector encoding a Type V CRISPR/Cas system (or a component thereof). Instead of a Type I and Type II system, the invention alternatively provides in an embodiment a first vector encoding a Type V CRISPR/Cas system (or component thereof) and a second vector encoding a Type VI CRISPR/Cas system (or a component thereof).

Instead of a Type I and Type II system, the invention alternatively provides in an embodiment first and second vectors (eg, plasmids), each encoding a Type I CRISPR/Cas system (or a component thereof). Instead of a Type I and Type II system, the invention alternatively provides in an embodiment first and second vectors, each encoding a Type II CRISPR/Cas system (or a component thereof). Instead of a Type I and Type II system, the invention alternatively provides in an embodiment first and second vectors, each encoding a Type III CRISPR/Cas system (or a component thereof). Instead of a Type I and Type II system, the invention alternatively provides in an embodiment first and second vectors, each encoding a Type IV CRISPR/Cas system (or a component thereof). Instead of a Type I and Type II system, the invention alternatively provides in an embodiment first and second vectors, each encoding a Type V CRISPR/Cas system (or a component thereof). Instead of a Type I and Type II system, the invention alternatively provides in an embodiment first and second vectors, each encoding a Type VI CRISPR/Cas system (or a component thereof).

Optionally, the plasmids are members of different incompatibility groups, eg, groups selected from IncFI, IncFII, IncFIll, IncFIV, IncFV, IncM, Inc9, InclO, Incl, IncA, IncB, IncC, IncH, Incla, Incllc, Indi, IncI2, Indy, IncJ, IncL, IncN, Inc2e, IncO, IncP, IncS, IncT and IncW. In an example here, the target cell is an Enterobacteriaceae cell.

Advantageously, the carrier cells are for treating or preventing a target cell infection in a human or an animal subject (eg, a dog, cat, horse, chicken, cow, sheep, goat, pig, fish or shellfish).

Advantageously, the carrier cells are of a species that is probiotic to said subject or is probioitic to humans or animals (eg, chickens). For example, the carrier cells are probiotic Bacteriodetes (eg, Bacteriodes) cell, eg, wherein the subject is a human. For example, the carrier cells are probiotic Clostridiales cell, eg, wherein the subject is a human. For example, the carrier cells are probiotic E coli cell. For example, the carrier cells are probiotic Bacillus cell, eg, wherein the subject is a plant. Advantageously, each vector (eg, virus or plasmid) encodes one or more guide RNAs or one or more crRNAs that are capable of hybridizing in the target cell to respective vector target nucleic acid sequence(s). For example, each vector encodes 2, 3, 4, 5, 6, 7, 7, 9, or 10 (or more than 10) different gRNAs or different crRNAs that hybridise to a respective target sequence, wherein the target sequences are different from each other. For example, 3 different gRNAs or crRNAs are encoded by each vector. For example, 2 different gRNAs or crRNAs are encoded by each vector. For example, 3 different gRNAs or crRNAs are encoded by each vector. For example, 4 different gRNAs or crRNAs are encoded by each vector. For example, 3 different gRNAs or crRNAs are encoded by each vector. For example, 5 different gRNAs or crRNAs are encoded by each vector. For example, 6 different gRNAs or crRNAs are encoded by each vector. For example, 7 different gRNAs or crRNAs are encoded by each vector. For example, 8 different gRNAs or crRNAs are encoded by each vector. For example, 9 different gRNAs or crRNAs are encoded by each vector. For example, 10 different gRNAs or crRNAs are encoded by each vector. For example, 11 different gRNAs or crRNAs are encoded by each vector. For example, 12 different gRNAs or crRNAs are encoded by each vector. For example, 13 different gRNAs or crRNAs are encoded by each vector.

In an example, the target cells are Salmonella cells (eg, wherein the subject is a chicken). In an example, the target cells are Campylobacter cells (eg, wherein the subject is a chicken). In an example, the target cells are Edwardsiella cells (eg, wherein the subject is a fish or shellfish, eg, a catfish or a shrimp or prawn). In an example, the target cells are Bacteriodetes (eg, Bacteriodes) cells. In an example the target cells are Clostridiales cells. In an example, the target cells are E coli cells.

Optionally, each plasimid comprises an expressible tral and/or tra2 module or a homologue thereof for conjugative transfer of the plasmid between cells. Any episome herein may be a plasmid.

Optionally, each plasimid comprises an expressible operon of a tral and/or tra2 module or a homologue thereof for conjugative transfer of the plasmid between cells.

Optionally, each plasmid is a modified RK2 or R6K plasmid. The modification comprises an insertion of nucleotide sequences encoding P1 and P2.

Optionally, each plasmid comprises an oriV, eg, oriV of an Incl (eg, Indi or IncI2), RK2 or R6K plasmid, or a homologue thereof. Optionally, each plasmid comprises an oriV of an Incl (eg, Indi or IncI2), RK2 or R6K plasmid, or a homologue thereof

Optionally, each plasmid comprises an oriT, eg, oriT of an lncll or IncI2 plasmid. Optionally, each plasmid is a modified an Incl (eg, Indi or IncI2) plasmid. The modification (for the First to Third Configurations, and optionally for the Fourth Configuration) comprises an insertion of nucleotide sequences encoding P1 and P2.

Optionally, P2 comprises one or more components of a CRISPR/Cas system that is operable in the target cell to cut a protospacer sequence comprised by the vector or carrier cell genome, eg, wherein the protospacer sequence is comprised by the carrier cell chromosome.

In an embodiment, the cutting herein kills the carrier cell or causes degradation of the vector in the target cell. In an alternative, the cutting inhibits the growth or proliferation of the target cell.

Optionally, P2 encodes a guide RNA or crRNA of a CRISPR/Cas system that is operable with a Cas nuclease in the target cell to cut a protospacer sequence comprised by the vector.

In an example of the First to Third Configurations, the protospacer is comprised by a gene required for vector viability or maintenance in the cell. In an example of the Fourth Configuration, the protospacer is comprised by a gene required for carrier cell viability.

Optionally, each vector (eg, virus (eg, phage) or plasmid) comprises a gene that encodes a product, wherein the product is essential for survival or proliferation of the carrier cell when in an environment that is devoid of the product, wherein the carrier cell chromosome does not comprise an expressible gene encoding the product and optionally the vector nucleic acid is the only episomal nucleic acid comprised by the carrier cell that encodes the product. For example, the gene is selected from an aroA, argH, hisD, leuB, lysA, metB, proC, thrC, pheA, tyrA, trpC and pflA gene; or wherein the gene is an anti-toxin gene and optionally the vector encodes a cognate toxin.

For example, the carrier cell is an E coli (eg, Nissle, F18 or S17 E coli strain) cell. For example, the carrier cell is a Bacillus (eg, B subtilis), Enterococcus or Lactobacillus cell, eg, wherein the subject is a plant.

Optionally, each carrier cell is for administration to a microbiota of a human or animal subject for medical use. For example, the medical use is for treating or preventing a disease disclosed herein. For example, the medical use is for treating or preventing a condition disclosed herein.

Optionally, the medical use is for the treatment or prevention of a disease or condition mediated by said target cells. Optionally, the medical use is for the treatment or prevention of a disease or condition mediated by cells of a microbiota that also comprises said target cells. For example, P1 is secreted from target cells and acts to kill or modify the growth or metabolism of neighbouring cells in the microbiota.

Optionally, the carrier cell(s) is(are) for administration to a human or animal for enhancing growth or weight of the human or animal. Optionally, the carrier cell(s) is(are) for administration to a human or animal for reducing growth or weight of the human or animal. Optionally, the carrier cell(s) is(are) for administration to a human or animal for reducing obesity in the human or animal.

In an embodiment, the administration is to a human for enhancing the growth or weight of the human. Optionally, the enhancing is not a medical therapy. Optionally, the enhancing is a medical therapy.

Optionally, the use comprises the administration of a plurality of carrier cells to a microbiota (eg, a gut microbiota) of a human or animal subject, wherein the microbiota comprises target cells and the vector nucleic acid is transferred into target cells for expression therein of P1, thereby killing microbiota cells in the subject or reducing the growth or proliferation of microbiota cells.

Optionally, the use comprises the administration of a plurality of carrier cells to a microbiota (eg, a gut microbiota) of a human or animal subject, wherein the microbiota comprises target cells and the vector nucleic acid is transferred into target cells for expression therein of P1, thereby promoting growth or metabolism of microbiota cells in the subject.

Optionally, the use comprises the administration of a plurality of carrier cells to a microbiota (eg, a gut microbiota) of a human or animal subject, wherein the microbiota comprises target cells and the vector nucleic acid is transferred into target cells for expression therein of P1, wherein P1 is a protein (eg, an enzyme) in a metabolic pathway in cells of the microbiota. For example, P1 is secreted by target cells and is taken up by further cells in the microbiota for use of P1 in a metabolic pathway in the further cells.

Optionally, the use comprises the administration of a plurality of carrier cells to a microbiota (eg, a gut microbiota) of a human or animal subject, wherein the microbiota comprises target cells and the vector nucleic acid is transferred into target cells for expression therein of P1, wherein P1 is a protein that is capable of sequestering a substance (eg, a protein, peptide, nucleic acid (eg, RNA), carbohydrate (eg, a sugar or precursor thereof), amino acid, lipid, fatty acid, ion or chemical compound) in the subject. For example, P1 is secreted by target cells and is taken up by further cells in the microbiota for use of P1 as a sequestering agent in the further cells. For example, P1 is secreted by target cells for use of P1 as a sequestering agent in the subject, eg, in the microbiota, an organ, tissue, cell or bloodstream of the subject.

For example, a plant herein in any configuration or embodiment of the invention is selected from a tomato plant, a potato plant, a wheat plant, a com plant, a maize plant, an apple tree, a bean-producing plant, a pea plant, a beetroot plant, a stone fruit plant, a barley plant, a hop plant and a grass. For example, the plant is a tree, eg, palm, a horse chestnut tree, a pine tree, an oak tree or a hardwood tree. For example the plant is a plant that produces fruit selected from strawberries, raspberries, blackberries, reducrrants, kiwi fruit, bananas, apples, apricots, avoocados, cherries, oranges, clementines, satsumas, grapefruits, plus, dates, figs, limes, lemons, melons, mangos, pears, olives or grapes. Optionally, the plant is a dicotyledon. Optionally, the plant is a flowering plant. Optionally, the plant is a monocotyledon.

In an example, the weight (ie, biomass) of a plant is dry weight. For example, the method is for increasing plant dry weight (eg, within 1 or 2 weeks of said administration). Optionally, the increase is an increase of at least 5, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19 or 20% compared to a control plant of the same species or strain to which the administration if carrier cells has not taken place, wherein all plants are kept under the same environmental conditions. For example, such an increase is within 1, 2, 3, 4, 5, 6, or 8 weeks following the first administration of the carrier cells. In an example, the method is for increasing the dry weight of a leaf and/or fruit of the plant, such as a tomato plant.

In an example, the weight is wet weight. For example, the method is for increasing plant wet weight (eg, within 1 or 2 weeks of said administration). Optionally, the increase is an increase of at least 5, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19 or 20% compared to a control plant of the same species or strain to which the administration if carrier cells has not taken place, wherein all plants are kept under the same environmental conditions. For example, such an increase is within 1, 2, 3, 4, 5, 6, or 8 weeks following the first administration of the carrier cells. In an example, the method is for increasing the dry weight of a leaf and/or fruit of the plant, such as a tomato plant.

For example, the microbiota is comprised by a leaf, trunk, root or stem of the plant. The target bacteria (or target cell) may be comprised by a microbiota of a plant. In an example, the microbiota is comprised by a leaf. In an example, the microbiota is comprised by a xylem. In an example, the microbiota is comprised by a phloem. In an example, the microbiota is comprised by a root. In an example, the microbiota is comprised by a tuber. In an example, the microbiota is comprised by a bulb. In an example, the microbiota is comprised by a seed. In an example, the microbiota is comprised by an exocarp, epicarp, mesocarp or endocarp. In an example, the microbiota is comprised by a fruit, eg, a simple fruits; aggregate fruits; or multiple fruits. In an example, the microbiota is comprised by a seed or embryo, eg, by a seed coat; a seed leaf; cotyledons; or a radicle. In an example, the microbiota is comprised by a flower, eg, comprised by a peduncle; sepal: petals; stamen; filament; anther or pistil. In an example, the microbiota is comprised by a root; eg, a tap root system, or a fibrous root system. In an example, the microbiota is comprised by a leaf or leaves, eg, comprised by a leaf blade, petiole or stipule. In an example, the microbiota is comprised by a stem, eg, comprised by bark, epidermis, phloem, cambium, xylem or pith.

For example, the biofilm is comprised by a lung of the subject, eg, wherein the target cells are Pseudomonas (eg, P aeruginosa) cells. This may be useful wherein the subject is a human suffering from a lung disease or condition, such as pneumonia or cystic fibrosis, wherein P1 is a therapeutic protein that is expressed by modified target cells of the biofilm. For example, the biofilm is comprised by an animal or human organ disclosed herein. For example, the biofilm is comprised by a microbiota of a human or animal disclosed herein.

Optionally, said surface is a surface ex vivo, such as a surface comprised by a domestic or industrial apparatus or container.

Optionally, the target cells are comprised by a biofilm, eg, a biofilm as disclosed herein.

There is provided: -

A pharmaceutical composition, livestock growth promoting composition, soil improver, herbicide, plant fertilizer, food or food ingredient sterilizing composition, dental composition, personal hygiene composition or disinfectant composition (eg, for domestic or industrial use) comprising a plurality of the carrier cells.

Herein, a carrier cell is, eg, a commensal or probiotic cell for administration to a human or animal subject. For example, the carrier cell is commensal in a microbiota (eg, gut or blood microbiota) of a human or animal subject, wherein the carrier is for administration to the subject. In an example, a carrier cell is a prokaryotic cell. In an example, a carrier cell is a bacterial cell (and optionally the target cell is a bacterial cell). In an example, a carrier cell is an archaeal cell (and optionally the target cell is an archaeal cell)

Optionally, the carrier cell is a gram-positive bacterial cell and the target cell is a gram-positive bacterial cell.

Optionally, the carrier cell is a gram-positive bacterial cell and the target cell is a gram-negative bacterial cell.

Optionally, the carrier cell is a gram-negative bacterial cell and the target cell is a gram-positive bacterial cell.

Optionally, the carrier cell is a gram-negative bacterial cell and the target cell is a gram-negative bacterial cell.

Optionally, the carrier cell is a Bacteriodes bacterial cell and the target cell is a gram-positive bacterial cell. Optionally, the carrier cell is a Bacteriodes bacterial cell and the target cell is a gramnegative bacterial cell. Optionally, the carrier cell is a Bacteriodes bacterial cell and the target cell is a Bacteriodes bacterial cell. Optionally, the carrier cell is a Bacteriodes bacterial cell and the target cell is an E coli bacterial cell. Optionally, the carrier cell is an E coli bacterial cell and the target cell is a Bacteriodes bacterial cell. For example, in these options the subject is a human or animal, preferably a human.

Optionally, the carrier cell is a Clostridiales bacterial cell and the target cell is a gram-positive bacterial cell. Optionally, the carrier cell is a Clostridiales bacterial cell and the target cell is a gramnegative bacterial cell. Optionally, the carrier cell is a Bacteriodes bacterial cell and the target cell is a Clostridiales bacterial cell. Optionally, the carrier cell is a Clostridiales bacterial cell and the target cell is an E coli bacterial cell. Optionally, the carrier cell is an E coli bacterial cell and the target cell is a Clostridiales bacterial cell. For example, in these options the subject is a human or animal, preferably a human.

Optionally, the carrier cell is an Escherichia (eg, E coli) bacterial cell and the target cell is a grampositive bacterial cell. Optionally, the carrier cell is an Escherichia (eg, E coli) bacterial cell and the target cell is a gram-negative bacterial cell. Optionally, the carrier cell is a Bacteriodes bacterial cell and the target cell is an Escherichia (eg, E coli) bacterial cell. Optionally, the carrier cell is an Escherichia (eg, E coli) bacterial cell and the target cell is an E coli bacterial cell. Optionally, the carrier cell is an A coli bacterial cell and the target cell is an Escherichia (eg, A coli) bacterial cell. For example, in these options the subject is a human or animal, preferably a human.

Optionally, the carrier cell is a Bacillus bacterial cell and the target cell is a gram-positive bacterial cell. Optionally, the carrier cell is a Bacillus bacterial cell and the target cell is a gram-negative bacterial cell. Optionally, the carrier cell is a Bacillus bacterial cell and the target cell is a Salmonella bacterial cell. Optionally, the carrier cell is a Bacillus bacterial cell and the target cell is an E coli bacterial cell. Optionally, the carrier cell is an E coli bacterial cell and the target cell is a Pseudomonas bacterial cell. For example, in these options the subject is a plant.

Optionally, the carrier cell is an A coli bacterial cell and the target cell is a gram-positive bacterial cell.

Optionally, the carrier cell is an E coli bacterial cell and the target cell is a gram-negative bacterial cell.

Optionally, the carrier cell is an E coli bacterial cell and the target cell is a Salmonella bacterial cell.

Optionally, the carrier cell is an E coli bacterial cell and the target cell is an E coli bacterial cell.

Optionally, the carrier cell is an E coli bacterial cell and the target cell is a Pseudomonas bacterial cell.

A Bacillus cell herein is optionally a B subtilis cell.

Optionally, the carrier cell is a probiotic or commensal Bacteriodes bacterial cell for administration to a human or animal subject. Optionally, the carrier cell is a probiotic or commensal Clostridiales bacterial cell for administration to a human or animal subject. Optionally, the carrier cell is a probiotic or commensal E coli bacterial cell for administration to a human or animal subject.

Optionally, the carrier cell is a probiotic or commensal Bacillus bacterial cell for administration to a human or animal subject.

Herein, optionally the plasmid is a closed circular DNA. In an embodiment, the vector (eg, plasmid) nucleic acid is DNA. Optionally, the DNA is dsDNA. In an embodiment, the vector DNA is ssDNA. In an embodiment, the vector (eg, plasmid) nucleic acid is RNA.

In an example, the target cell is a cell of a species that does not cause nosocomial infection in humans.

Optionally, the target cell is comprised by an animal (eg, poultry animal (such as chicken), swine, cow, fish (eg, catfish or salmon) or shellfish (eg, prawn or lobster)) microbiota. Optionally, the microbiota is a gut microbiota. For example, the target cell is a cell comprised by a human or animal (eg, chicken) gut biofilm. For example, the target cell is a cell comprised by a gut biofilm sample ex vivo. For example, the target cell is a cell comprised by a human or animal (eg, chicken) lung biofilm. For example, the target cell is a cell comprised by a lung biofilm sample ex vivo. For example, the target cell is a cell comprised by a human or animal (eg, chicken) skin biofilm. For example, the target cell is a cell comprised by a skin biofilm sample ex vivo.

In an embodiment, each plasmid comprises an oriV and/or an oriT. In an embodiment, each plasmid comprises a bacterial oriV and/or an oriT.

In an embodiment, the plasmid comprises an oriV and does not encode any replication protein (eg,pzr or trfA) that is operable with the oriV to initiate replication of the plasmid.

In an example, the invention relates to a composition comprising a plurality of carrier cells of the invention. Optionally, all of the carrier cells comprise identical said vectors (eg, plasmids).

Optionally, the plurality comprises a first sub-population of carrier cells (first cells) and a second subpopulation of carrier cells (second cells) wherein the first cells comprise indentical first said vectors and the second cells comprise indentical second said vectors (which are different from the first vectors of the first cells). For example, the first vectors encode a first guide RNA or crRNA and the second vectors encode a second guide RNA or crRNA, wherein the first guide RNA/crRNA is capable of hybridizing to a first protospacer sequence comprised by the vector in first target cells; and the second guide RNA/crRNA is capable of hybridizing to a second protospacer sequence in carrier or target cells, wherein the protospacers are different.

Optionally, the composition is comprised by a liquid (eg, an aqueous liquid or in water), the composition comprising the carrier cells at an amount of from 1 x 10 3 to 1 x IO 10 (eg, from 1 x 10 4 to 1 x IO 10 ; from 1 x 10 4 to 1 x 10 9 ; from 1 x 10 4 to 1 x 10 8 ; from 1 x 10 4 to 1 x 10 7 ; from 1 x 10 3 to 1 x IO 10 ; from 1 x 10 3 to 1 x 10 9 ; from 1 x 10 3 to 1 x 10 8 ; from 1 x 10 3 to 1 x 10 7 ; from 1 x 10 5 to 1 x IO 10 ; from 1 x 10 5 to 1 x 10 9 ; from 1 x 10 5 to 1 x 10 8 ; from 1 x 10 5 to 1 x 10 7 ; from 1 x 10 6 to 1 x IO 10 ; from 1 x 10 6 to 1 x 10 9 ; from 1 x 10 6 to 1 x 10 8 ; or from 1 x 10 6 to 1 x 10 7 ) cfu/ml. For example, the liquid is a beverage, such for human or animal consumption. For example, the beverage is a livestock beverage, eg, a poultry beverage (ie, a beverage for consumption by poultry, such as chicken).

In an example, the composition is a dietary (eg, dietary supplement) composition for consumption by humans or animals. In an example, the composition is a slimming composition for consumption by humans or animals. In an example, the composition is a growth promotion composition for consumption by humans or animals. In an example, the composition is a body buidling composition for consumption by humans. In an example, the composition is a probiotic composition for consumption by humans or animals. In an example, the composition is a biocidal composition for consumption by humans or animals. In an example, the composition is a pesticidal composition for consumption by humans or animals. In an example, the composition is a zoonosis control composition for consumption by animals.

In an example, the composition comprises vitamins in addition to the carrier cells. In an example, the composition comprises vitamin A, B (eg, B12), C, D, E and/or K in addition to the carrier cells. In an example, the composition comprises lipids in addition to the carrier cells. In an example, the composition comprises carbohydrates in addition to the carrier cells. In an example, the composition comprises proteins and/or amino acids in addition to the carrier cells. In an example, the composition comprises minerals in addition to the carrier cells. In an example, the composition comprises metal ions (eg, Mg 2+ , Cu 2+ and/or Zn 2+ ) in addition to the carrier cells. In an example, the composition comprises sodium ions, potassium ions, magnesium ions, calcium ions, manganese ions, iron ions, cobalt ions, copper ions, zinc ions and/or molybdenum ions.

In an example, the composition is a plant fertilizer composition. In an example, the composition is a herbicide. In an example, the composition is a pesticide composition for application to plants.

In any embodiment or example, where appropriate: The plants are, for example, crop plants. The plants are, for example, wheat. The plants are, for example, com. The plants are, for example, maize. The plants are, for example, fruiting plants. The plants are, for example, vegetable plants. The plants are, for example, tomato plants. The plants are, for example, potato plants. The plants are, for example, grass plants. The plants are, for example, flowering plants. The plants are, for example, trees. The plants are, for example, shrubs. In an example, the composition is for environmental application, wherein the environment is an outdoors environment (eg, application to a field or waterway or reservoir).

In an example, the composition is comprised by a food or food ingredient (eg, for human or animal consumption). In an example, the composition is comprised by a beverage or beverage ingredient (eg, for human or animal consumption).

In an example the target cell(s) are biofilm cells found in a human, eg, wherein the biofilm is a gut, skin, lung, eye, nose, ear, gastrointestinal tract (GI tract), stomach, hair, kidney, urethra, bronchiole, oral cavity, mouth, liver, heart, anus, rectum, bladder, bowel, intestine, penis, vagina or scrotum biofilm. In an example the target cell(s) are animal biofilm cells, eg, wherein the biofilm is a gut, skin, lung, eye, nose, ear, gastrointestinal tract (GI tract), caecum, jejunum, ileum, colon, stomach, hair, feather, scales, kidney, urethra, bronchiole, oral cavity, mouth, liver, spleen, heart, anus, rectum, bladder, bowel, intestine, penis, vagina or scrotum biofilm. For example, the biofilm is a bird (eg, chicken) caecum biofilm. For example, the biofilm is a bird (eg, chicken) gastrointestinal tract (GI tract), caecum, jejunum, ileum, colon or stomach biofilm.

In an example, any method herein is ex vivo. In an example, a method herein is in vivo. In an example, a method herein is in vitro. In an example, a method herein is carried out in an environment, eg, in a domestic (such as in a house), industrial (such as in a factory) or agricultural environment (such as in a field). In an example, a method herein is carried out in or on a container; or on a surface.

In an example each vector (eg, plasmid) encodes one or more components of a CRISPR/Cas system operable to perform vector protospacer cutting in the target cell (eg, wherein the protospacer comprises 10-20, 10-30, 10-40, 10-100, 12-15 or 12-20 consecutive nucleotides that are capable of hybridizing in the target cell with a crRNA or gRNA encoded by the vector). For example, the system is a Type I, II, III, IV or V CRISPR/Cas system.

In an example, each vector encodes a Cas9 (and optionally a second, different, Cas, such as a Cas3, Cas9, Cpfl, Cas 13a, Cas 13b or Cas 10); and/or a Cas3 (and optionally a second, different, Cas, such as a Cas3, Cas9, Cpfl, Casl3a, Casl3b or CaslO). In an example, each vector encodes a Cas selected from a Cas3, Cas9, Cpfl, Casl3a, Casl3b and CaslO. Additionally or alternatively, the vector encodes a guide RNA or crRNA or tracrRNA. For example, the guide RNA or crRNA or tracrRNA is cognate to (ie, operable with in the target cell) the first Cas. In an example, a Cas herein is a Cas9. In an example, a Cas herein is a Cas3. The Cas may be identical to a Cas encoded by the target bacteria.

In an embodiment, each plasmid is a shuttle vector.

Optionally, the target cell is devoid of a functional endogenous CRISPR/Cas system before transfer therein of the vector, eg, wherein the vector encodes a component of an exogenous CRISPR/Cas system that is functional in the target cell. An embodiment provides a medicament comprising a plurality of carrier cells of the invention, wherein each target cell is optionally according to this paragraph, for administration to a human or animal subject for medical use.

In an example, the composition of the invention is a herbicide, pesticide, insecticide, plant fertilizer or cleaning agent.

Optionally, target bacteria herein are comprised by a microbiota of the subject, eg, a gut microbiota. Alternatively, the microbiota is a skin, scalp, hair, eye, ear, oral, throat, lung, blood, rectal, anal, vaginal, scrotal, penile, nasal or tongue microbiota.

In an example the subject (eg, human or animal) is further administered a medicament simultaneously or sequentially with the carrier cell administration. In an example, the medicament is an antibiotic, antibody or antibody fragment (eg, an scFv, nanobody or Fab), immune checkpoint inhibitor (eg, an anti-PD-1, anti-PD-Ll or anti-CTLA4 antibody), adoptive cell therapy (eg, CAR-T therapy) or a vaccine.

In an embodiment, the vector encodes a guided nuclease, such as a Cas nuclease, TALEN, zinc finger nuclease or meganuclease. Thus, P2 may comprise a guided nuclease, such as a Cas nuclease, TALEN, zinc finger nuclease or meganuclease. Optionally, the vector encodes a restriction nuclease that is capable of cutting the vector and/or genome of the carrier cell (eg, chromosome of the carrier cell).

Optionally, the composition is a pharmaceutical composition for use in medicine practised on a human or animal subject. Optionally, the composition is not a medicament.

In an example, the animal is a livestock or companion pet animal (eg, a cow, pig, goat, sheep, horse, dog, cat or rabbit). In an example, the animal is an insect (an insect at any stage of its lifecycle, eg, egg, larva or pupa). In an example, the animal is a protozoan. In an example, the animal is a cephalopod.

Optionally, the composition is a herbicide, pesticide, food or beverage processing agent, food or beverage additive, petrochemical or fuel processing agent, water purifying agent, cosmetic additive, detergent additive or environmental (eg, soil) additive or cleaning agent.

For example, the carrier bacteria are Lactobacillus (eg, L reuteri or L lactis), E coli, Bacillus or Streptococcus (eg, S thermophilus) bacteria. Usefully, the carrier can provide protection for the plasmid from the surrounding environment. The use of a carrier may be useful for oral administration or other routes where the carrier can provide protection for the vector from the acid stomach or other harsh environments in the subject. Furthermore, the carrier can be formulated into a beverage, for example, a probiotic drink, eg, an adapted Yakult (trademark), Actimel (trademark), Kevita (trademark), Activia (trademark), Jarrow (trademark) or similar drink for human consumption.

Optionally, the carrier cell(s) or composition are for administration to a human or animal subject for medical use, comprising killing target bacteria using P1 or a metabolite thereof that is produced in the target cell, wherein the target bacteria mediate as disease or condition in the subject. In an example, when the subject is a human, the subject is not an embryo. In an example, the carrier cells are probiotic in the subject.

Optionally, the environment is a microbiota of soil; a plant, part of a part (e.g., a leaf, fruit, vegetable or flower) or plant product (e.g., pulp); water; a waterway; a fluid; a foodstuff or ingredient thereof; a beverage or ingredient thereof; a medical device; a cosmetic; a detergent; blood; a bodily fluid; a medical apparatus; an industrial apparatus; an oil rig; a petrochemical processing, storage or transport apparatus; a vehicle or a container.

Optionally, the environment is an ex vivo bodily fluid (e.g., urine, blood, blood product, sweat, tears, sputum or spit), bodily solid (e.g., faeces) or tissue of a human or animal subject that has been administered the composition.

Optionally, the environment is an in vivo bodily fluid (e.g., urine, blood, blood product, sweat, tears, sputum or spit), bodily solid (e.g., faeces) or tissue of a human or animal subject that has been administered the composition. In an embodiment, the plasmid is a phagemid or cloning vector (eg, a shuttle vector, eg, a pUC vector). In an embodiment, the plasmid is a conjugative plasmid.

Optionally, P2 comprises one or more components of a CRISPR/Cas system, eg, a DNA sequence encoding one or more components of Type I Cascade (eg, CasA).

Optionally, P2 comprises a DNA sequence encoding guided nuclease, such as a Cas nuclease, TALEN, zinc finger nuclease or meganuclease.

In an example, the carrier cell(s) or composition are comprised by a medical container, eg, a syringe, vial, IV bag, inhaler, eye dropper or nebulizer. In an example, the carrier cell(s) or composition are comprised by a sterile container. In an example, the carrier cell(s) or composition are comprised by a medically-compatible container. In an example, the carrier cell(s) or composition are comprised by a fermentation vessel, eg, a metal, glass or plastic vessel. In an example, the carrier cell(s) or composition are comprised by an agricultural apparatus. In an example, the carrier cell(s) or composition are comprised by food production or processing apparatus. In an example, the carrier cell(s) or composition are comprised by a horticultural apparatus. In an example, the carrier cell(s) or composition are comprised by a farming apparatus. In an example, the carrier cell(s) or composition are comprised by petrochemicals recovery or processing apparatus. In an example, the carrier cell(s) or composition are comprised by a distillation apparatus. In an example, the carrier cell(s) or composition are comprised by cell culture vessel (eg, having a capacity of at least 50, 100, 1000, 10000 or 100000 litres). Additionally or alternatively, the target cell(s) are comprised by any of these apparatus etc.

In an example, the carrier cell(s) or composition are comprised by a medicament, e,g in combination with instructions or a packaging label with directions to administer the medicament by oral, IV, subcutaneous, intranasal, intraocular, vaginal, topical, rectal or inhaled administration to a human or animal subject. In an example, the carrier cell(s) or composition are comprised by an oral medicament formulation. In an example, the carrier cell(s) or composition are comprised by an intranasal or ocular medicament formulation. In an example, the carrier cell(s) or composition are comprised by a personal hygiene composition (eg, shampoo, soap or deodorant) or cosmetic formulation. In an example, th the carrier cell(s) or composition are comprised by a detergent formulation. In an example, the carrier cell(s) or composition are comprised by a cleaning formulation, eg, for cleaning a medical or industrial device or apparatatus. In an example, the carrier cell(s) or composition are comprised by foodstuff, foodstuff ingredient or foodstuff processing agent. In an example, the carrier cell(s) or composition are comprised by beverage, beverage ingredient or beverage processing agent. In an example, the carrier cell(s) or composition are comprised by a medical bandage, fabric, plaster or swab. In an example, the carrier cell(s) or composition are comprised by a herbicide or pesticide. In an example, the carrier cell(s) or composition are comprised by an insecticide.

In an example, the CRISPR/Cas component(s) are component(s) of a Type I CRISPR/Cas system. In an example, the CRISPR/Cas component(s) are component(s) of a Type II CRISPR/Cas system. In an example, the CRISPR/Cas component(s) are component(s) of a Type III CRISPR/Cas system. In an example, the CRISPR/Cas component(s) are component(s) of a Type IV CRISPR/Cas system. In an example, the CRISPR/Cas component(s) are component(s) of a Type V CRISPR/Cas system. In an example, the CRISPR/Cas component(s) comprise a Cas9-encoding nucleotide sequence (eg, S pyogenes Cas9, S aureus Cas9 or S thermophilus Cas9). In an example, the CRISPR/Cas component(s) comprise a Cas3 -encoding nucleotide sequence (eg, E coli Cas3, C dificile Cas3 or Salmonella Cas3). In an example, the CRISPR/Cas component(s) comprise a Cpf-encoding nucleotide sequence. In an example, the CRISPR/Cas component(s) comprise a CasX-encoding nucleotide sequence. In an example, the CRISPR/Cas component(s) comprise a CasY-encoding nucleotide sequence.

Optionally, target bacteria are gram negative bacteria (eg, a spirilla or vibrio). Optionally, target bacteria are gram positive bacteria. Optionally, target bacteria are mycoplasma, chlamydiae, spirochete or mycobacterium bacteria. Optionally, target bacteria are Streptococcus (eg, pyogenes or thermophilus)' . Optionally, target bacteria are Staphylococcus (eg, aureus, eg, MRSA). Optionally, target bacteria are E. coli (eg, 0157: H7), eg, wherein the Cas is encoded by the vecor or an endogenous target cell Cas nuclease (eg, Cas3) activity is de-repressed. Optionally, target bacteria are Pseudomonas (eg, syringae or aeruginosa). Optionally, target bacteria are Vibro (eg, cholerae (eg, 0139) or vulnificus). Optionally, target bacteria are Neisseria (eg, gonnorrhoeae or meningitidis). Optionally, target bacteria are Bordetella (eg, pertussis). Optionally, target bacteria are Haemophilus (eg, influenzae). Optionally, target bacteria are Shigella (eg, dysenteriae). Optionally, target bacteria are Brucella (eg, abortus). Optionally, target bacteria are Francisella host. Optionally, target bacteria are Xanthomonas. Optionally, target bacteria are Agrobacterium. Optionally, target bacteria are Erwinia. Optionally, target bacteria are Legionella (eg, pneumophila). Optionally, target bacteria are Listeria (eg, monocytogenes). Optionally, target bacteria are Campylobacter (eg, jejuni). Optionally, target bacteria are Yersinia (eg,pestis). Optionally, target bacteria are Borelia (eg, burgdorferi).

Optionally, target bacteria are Helicobacter (eg, pylori). Optionally, target bacteria are Clostridium (eg, dificile or botulinum). Optionally, target bacteria are Erlichia (eg, chaffeensis). Optionally, target bacteria are Salmonella (eg, typhi or enterica, eg, serotype typhimurium, eg, DT 104). Optionally, target bacteria are Chlamydia (eg, pneumoniae). Optionally, target bacteria are Parachlamydia host. Optionally, target bacteria are Corynebacterium (eg, amycolatum). Optionally, target bacteria are Klebsiella (eg, pneumoniae). Optionally, target bacteria are Enterococcus (eg, faecalis or faecim, eg, linezolid-resistant). Optionally, target bacteria are Acinetobacter (eg, baumannii, eg, multiple drug resistant).

Further examples of target cells are as follows

(a) Optionally the target bacteria are Staphylococcus aureus cells, eg, resistant to an antibiotic selected from methicillin, vancomycin, linezolid, daptomycin, quinupristin, dalfopristin and teicoplanin.

(b) Optionally the target bacteria are Pseudomonas aeuroginosa cells, eg, resistant to an antibiotic selected from cephalosporins (eg, ceftazidime), carbapenems (eg, imipenem or meropenem), fluoroquinolones, aminoglycosides (eg, gentamicin or tobramycin) and colistin.

(c) Optionally the target bacteria are Klebsiella (eg, pneumoniae) cells, eg, resistant to carbapenem.

(d) Optionally the target bacteria are Streptoccocus (eg, thermophilus , pneumoniae or pyogenes) cells, eg, resistant to an antibiotic selected from erythromycin, clindamycin, beta-lactam, macrolide, amoxicillin, azithromycin and penicillin.

(e) Optionally the target bacteria are Salmonella (eg, serotype Typhi) cells, eg, resistant to an antibiotic selected from ceftriaxone, azithromycin and ciprofloxacin.

(f) Optionally the target bacteria are Shigella cells, eg, resistant to an antibiotic selected from ciprofloxacin and azithromycin.

(g) Optionally the target bacteria are Mycobacterium tuberculosis cells, eg, resistant to an antibiotic selected from Resistance to isoniazid (INH), rifampicin (RMP), fluoroquinolone, amikacin, kanamycin and capreomycin and azithromycin.

(h) Optionally the target bacteria are Enterococcus cells, eg, resistant to vancomycin.

(i) Optionally the target bacteria are Enterobacteriaceae cells, eg, resistant to an antibiotic selected from a cephalosporin and carbapenem.

(j) Optionally the target bacteria are E. coli cells, eg, resistant to an antibiotic selected from trimethoprim, itrofurantoin, cefalexin and amoxicillin.

(k) Optionally the target bacteria are Clostridium (eg, dificile) cells, eg, resistant to an antibiotic selected from fluoroquinolone antibiotic and carbapenem.

(l) Optionally the target bacteria are Neisseria gonnorrhoea cells, eg, resistant to an antibiotic selected from cefixime (eg, an oral cephalosporin), ceftriaxone (an injectable cephalosporin), azithromycin and tetracycline. (m) Optionally the target bacteria are Acinetoebacter baumannii cells, eg, resistant to an antibiotic selected from beta-lactam, meropenem and a carbapenem.

(n) Optionally the target bacteria are Campylobacter (eg, jejuni) cells, eg, resistant to an antibiotic selected from ciprofloxacin and azithromycin.

(o) Optionally, the target cell(s) produce Beta (P)-lactamase (eg, ESBL-producing E. coli or ESBL-producing Klebsiella).

(p) Optionally, the target cell(s) are bacterial cells that are resistant to an antibiotic recited in any one of examples (a) to (n).

In an example, the target cell(s) is a cell of a species selected from Shigella, E coli, Salmonella, Serratia, Klebsiella, Yersinia, Pseudomonas and Enterobacter, eg, wherein the subject is a plant. Optionally, the composition comprises carrier cells that are each or in combination capable of conjugative transfer of the vector nucleic acid into target cells of species selected from two or more of Shigella, E coli, Salmonella, Serratia, Klebsiella, Yersinia, Pseudomonas and Enterobacter, eg, wherein the subject is a plant.

In an example, the reduction in growth or proliferation of target cells is at least 50, 60, 70, 80, 90 or 95%.

In embodiments, the plasmid contains a screenable or selectable marker gene. For example, the selectable marker gene is an antibiotic resistance gene.

The carrier bacteria can be bacteria of a species or genus as follows. For example, the species is found in warm-blooded animals (eg, livestock vertebrates). For example, the species is found in humans. For example, the species is found in plants. Preferably, non-pathogenic bacteria that colonize the non-sterile parts of the human or animal body (e.g., skin, digestive tract, urogenital region, mouth, nasal passages, throat and upper airway, ears and eyes) are utilized as carrier cells, and in an example the methodology of the invention is used to combat a target cell bacterial infection of such a part of the body of a human or animal. In another embodiment, the infection is systemic infection. Examples of carrier bacterial species include, but are not limited to: non-pathogenic strains of Escherichia coli (E. coli Fl 8, S17 and E. coli strain Nissle), various species of Lactobacillus (such as L. casei, L. plantarum, L. paracasei, L. acidophilus, L. fermentum, L. zeae and L. gasseri), or other nonpathogenic or probiotic skin- or GI colonizing bacteria such as Lactococcus, Bifidobacteria, Eubacteria, and bacterial mini-cells, which are anucleoid cells destined to die but still capable of transferring plasmids (see; e.g., Adler et al., Proc. Natl. Acad. Sci. USA 57; 321-326, 1970; Frazer and Curtiss III, Current Topics in Microbiology and Immunology 69: 1-84, 1975; U.S. Patent No. 4,968,619 to Curtiss III). In some embodiments, the target recipient cells are pathogenic bacteria comprised by a human, animal or plant, eg, on the skin or in the digestive tract, urogenital region, mouth, nasal passage, throat and upper airway, eye(s) and ear(s). Of particular interest for targeting and eradication are pathogenic strains of Pseudomonas aeruginosa, Escherichia coll, Staphylococcus pneumoniae and other species, Enterobacter spp., Enterococcus spp. and Mycobacterium tuberculosis .

The present invention finds use with a wide array of settings or environments, eg, in therapeutic, agricultural, or other settings, including, but not limited to, those described in U.S. patents 6,271,359, 6,261,842, 6,221,582, 6,153,381, 6,106,854, and 5,627,275. Others are also discussed herein, and still others will be readily apparent to those of skill in the art.

A single carrier bacterial strain might harbor more than one type of such vector (eg, differing in the P1 that they encode). Further, in another example two or more different carrier bacterial strains, each containing one or more such vectors, may be combined for producing a plurality of different P1 products in the subject.

The present invention finds utility for treatment of humans and in a variety of veterinary, agronomic, horticultural and food processing applications. For human and veterinary use, and depending on the cell population or tissue targeted for protection, the following modes of administration of the carrier bacteria of the invention are contemplated: topical, oral, nasal, ocular, aural, pulmonary (e.g., via an inhaler), ophthalmic, rectal, urogenital, subcutaneous, intraperitoneal and intravenous. The bacteria may be supplied as a pharmaceutical composition, in a delivery vehicle suitable for the mode of administration selected for the patient being treated. The term "patient" or "subject" as used here may refer to humans or animals (animals being particularly useful as models for clinical efficacy of a particular donor strain, for example, or being farmed or livestock animals). Commercially-relevant animals are chicken, turkey, duck, catfish, salmon, cod, herring, lobster, shrimp, prawns, cows, sheep, goats, pigs, goats, geese or rabbits.

For example, to deliver the carrier bacteria to the gastrointestinal tract or to the nasal passages, the preferred mode of administration may be by oral ingestion or nasal aerosol, or by feeding (alone or incorporated into the subject's feed or food and/or beverage, such as drinking water). In this regard, the carrier cells may be comprised by a food of livestock (or farmed or companion animal), eg, the carrier bacteria are comprised by a feed additive for livestock. Alternatively, the additive is a beverage (eg, water) additive for livestock. It should be noted that probiotic bacteria, such as Lactobacillus acidophilus, are sold as gel capsules containing a lyophilized mixture of bacterial cells and a solid support such as mannitol. When the gel capsule is ingested with liquid, the lyophilized cells are re-hydrated and become viable, colonogenic bacteria. Thus, in a similar fashion, carrier bacterial cells of the present invention can be supplied as a powdered, lyophilized preparation in a gel capsule, or in bulk, eg, for sprinkling onto food or beverages. The re-hydrated, viable bacterial cells will then populate and/or colonise sites throughout the upper and/or lower gastrointestinal system, and thereafter come into contact with the target bacteria.

For topical applications, the carrier bacteria may be formulated as an ointment or cream to be spread on the affected skin surface. Ointment or cream formulations are also suitable for rectal or vaginal delivery, along with other standard formulations, such as suppositories. The appropriate formulations for topical, vaginal or rectal administration are well known to medicinal chemists.

The present invention may be of utility for topical or mucosal administrations to treat a variety of bacterial infections or bacterially related undesirable conditions. Some representative examples of these uses include treatment of (1) conjunctivitis, caused by Haemophilus sp., and corneal ulcers, caused by Pseudomonas aeruginosa,' (2) otitis externa, caused by Pseudomonas aeruginosa,' (3) chronic sinusitis, caused by many Gram-positive cocci and Gram-negative rods, or for general decontamination of bronchii; (4) cystic fibrosis, associated with Pseudomonas aeruginosa,' (5) enteritis, caused by Helicobacter pylori (eg, to treat or prevent gastric ulcers), Escherichia coli, Salmonella typhimurium, Campylobacter or Shigella sp. ; (6) open wounds, such as surgical or non- surgical, eg, as a prophylactic measure; (7) bums to eliminate Pseudomonas aeruginosa or other Gram-negative pathogens; (8) acne, eg, caused by Propionobacter acnes,' (9) nose or skin infection, eg, caused by methicillin resistant Staphylococcus aureus (MSRA); (10) body odor, eg, caused by Gram-positive anaerobic bacteria (i.e., use of carrier cells in deodorants); (11) bacterial vaginosis, eg, associated with Gardnerella vaginalis or other anaerobes; and (12) gingivitis and/or tooth decay caused by various organisms.

In one example, the target cells are E coli cells and the disease or condition to be treated or prevented in a human is a uterine tract infection or a ventilator associated infection, eg, pneumonia, sepsis, septicaemia or HUS.

In other embodiments, the carrier cells of the present invention find application in the treatment of surfaces for the removal or attenuation of unwanted target bacteria or for modification of bacteria on the surfaces, for example use in a method of treating such a surface or an environment comprising target bacteria, wherein the method comprises contacting the surface or environment with carrier bacteria of the invention, allowing transfer of the vector nucleic acid of the invention from the carrier to the target bacteria, and expressing P1 in target cells. For example, surfaces that may be used in invasive treatments such as surgery, catheterization and the like may be treated to prevent infection of a subject by bacterial contaminants on the surface. It is contemplated that the methods and compositions of the present invention may be used to treat numerous surfaces, objects, materials and the like (e.g., medical or first aid equipment, nursery and kitchen equipment and surfaces) to control bacterial contamination thereon.

Pharmaceutical preparations or other compositions comprising the carrier bacteria may be formulated in dosage unit form for ease of administration and uniformity of dosage. Dosage unit form, as used herein, refers to a physically discrete unit of the pharmaceutical preparation appropriate for the patient or plant or environment or surface undergoing treatment. Each dosage should contain a quantity of the carrier bacteria calculated to produce the desired antibacterial effect in association with the selected carrier. Procedures for determining the appropriate dosage unit are well known to those skilled in the art. Dosage units may be proportionately increased or decreased based on the weight of a patient, plant, surface or environment. Appropriate concentrations for achieving eradication of pathogenic target cells (eg, comprised by a tissue of the patient) may be determined by dosage concentration curve calculations, as known in the art.

Other uses for the carrier bacteria of the invention are also contemplated. These include a variety agricultural, horticultural, environmental and food processing applications. For example, in agriculture and horticulture, various plant pathogenic bacteria may be targeted in order to minimize plant disease. One example of a plant pathogen suitable for targeting is Erwinia (eg, E amylovora, the causal agent of fire blight). Similar strategies may be utilized to reduce or prevent wilting of cut flowers. For veterinary or animal farming, the carrier cells of the invention may be incorporated into animal feed (chicken, swine, poultry, goat, sheep, fish, shellfish or cattle feed) to reduce bio-burden or to eliminate certain pathogenic organisms (e.g., Salmonella, such as in chicken, turkey or other poultry). In other embodiments, the invention may be applied on meat or other foods to eliminate unwanted or pathogenic bacteria (e.g., E. coli O157:H7 on meat, or Proteus spp., one cause of "fishy odour" on seafood).

Environmental utilities comprise, for example, engineering carrier bacteria, eg, Bacillus thurengiensis and one of its conjugative plasmids, to deliver and conditionally express an insecticidal agent in addition to or instead of an antibacterial agent (e.g., for the control of mosquitos that disseminate malaria or West Nile virus). In such applications, as well as in the agricultural and horticultural or other applications described above, formulation of the carrier bacteria as solutions, aerosols, or gel capsules are contemplated. In an example the plasmid (eg, where the microbiota is a hunan, anima or plant microbiota) is an engineered RK2 plasmid (ie, a RK2 plasmid that has been modified by recombinant DNA technology or a progeny of such a modified plasmid). P1asmid RK2 is a promiscuous plasmid that can replicate in 29 (and probably many more) gram-negative species (Guiney and Lanka, 1989, p 27-54. In C. M. Thomas (ed) Promiscous plasmids in gram-negative bacteria. London, Ltd London United Kingdom.). P1asmid RK2 is a 60-kb self-transmissible plasmid with a complete nucleotide sequence known (Pansegrau et al., 1994, J. Mol. Biol. 239, 623-663). A minimal replicon derived from this large plasmid has been obtained that is devoid of all its genes except for a trfA gene, that encodes plasmid' s Rep protein called TrfA, and an origin of vegetative replication oriV For a review of RK2 replication and its control by TrfA protein, see Helinski et al., 1996 (In Escherichia coli and Salmonella Cellular and Molecular Biology, Vol. 2 (ed. F. Neidhardt, et al, 2295-2324, ASM Press, Washington D.C.).

In an example the plasmid (eg, where the microbiota is a hunan, anima or plant microbiota) is an engineered R6K plasmid (ie, a R6K plasmid that has been modified by recombinant DNA technology or a progeny of such a modified plasmid).

The present invention is optionally for an industrial or domestic use, or is used in a method for such use. For example, it is for or used in agriculture, oil or petroleum industry, food or drink industry, clothing industry, packaging industry, electronics industry, computer industry, environmental industry, chemical industry, aeorspace industry, automotive industry, biotechnology industry, medical industry, healthcare industry, dentistry industry, energy industry, consumer products industry, pharmaceutical industry, mining industry, cleaning industry, forestry industry, fishing industry, leisure industry, recycling industry, cosmetics industry, plastics industry, pulp or paper industry, textile industry, clothing industry, leather or suede or animal hide industry, tobacco industry or steel industry.

The present invention is optionally for use in an industry or the environment is an industrial environment, wherein the industry is an industry of a field selected from the group consisting of the medical and healthcare; pharmaceutical; human food; animal food; plant fertilizers; beverage; dairy; meat processing; agriculture; livestock farming; poultry farming; fish and shellfish farming; veterinary; oil; gas; petrochemical; water treatment; sewage treatment; packaging; electronics and computer; personal healthcare and toiletries; cosmetics; dental; non-medical dental; ophthalmic; nonmedical ophthalmic; mineral mining and processing; metals mining and processing; quarrying; aviation; automotive; rail; shipping; space; environmental; soil treatment; pulp and paper; clothing manufacture; dyes; printing; adhesives; air treatment; solvents; biodefence; vitamin supplements; cold storage; fibre retting and production; biotechnology; chemical; industrial cleaning products; domestic cleaning products; soaps and detergents; consumer products; forestry; fishing; leisure; recycling; plastics; hide, leather and suede; waste management; funeral and undertaking; fuel; building; energy; steel; and tobacco industry fields.

In an example, the plasmid comprises a CRISPR array, wherein the array comprises one, or two or more different spacers (eg, 2, 3, 4, 5, 6, 7, 8, 9 ,10, 20, 30, 40, 50 or more spacers) for targeting the vector and/or the genome of a carrier bacterium.

In an example, the target bacteria are comprised by an environment as follows. In an example, the environment is a microbiota of a human, eg, the oral cavity microbiota or gut microbiota or the bloodstream. In an example, the environment is not an environment in or on a human. In an example, the environment is not an environment in or on a non-human animal. In an embodiment, the environment is an air environment. In an embodiment, the environment is an agricultural environment. In an embodiment, the environment is an oil or petroleum recovery environment, eg, an oil or petroleum field or well. In an example, the environment is an environment in or on a foodstuff or beverage for human or non-human animal consumption. In an example, the environment is a maritimeenvironment, eg, in seawater or on a boat (eg, in ship or boat ballast water).

In an example, the environment is a a human or animal microbiota (eg, gut, vaginal, scalp, armpit, skin or oral cavity microbiota). In an example, the target bacteria are comprised by a human or animal microbiota (eg, gut, vaginal, scalp, armpit, skin or oral cavity microbiota).

In an example, the carrier bacteria or composition of the invention are administered intranasally, topically or orally to a human or non-human animal, or is for such administration. The skilled person aiming to treat a microbiota of the human or animal will be able to determine the best route of administration, depending upon the microbiota of interest. For example, when the microbiota is a gut microbiota, administration can be intranasally or orally. When the microbiota is a scalp or armpit microbiota, administration can be topically. When the microbiota is in the mouth or throat, the administration can be orally.

In an example, the environment is harboured by a beverage or water (eg, a waterway or drinking water for human consumption) or soil. The water is optionally in a heating, cooling or industrial system, or in a drinking water storage container. In an example, the carrier and/or target bacteraia are Firmicutes selected from Anaerotruncus, Acetanaerobacterium, Acetitomaculum, Acetivibrio, Anaerococcus, Anaerofilum, Anaerosinus, Anaerostipes, Anaerovorax, Butyrivibrio, Clostridium, Capracoccus, Dehalobacter, Dialister, Dorea, Enterococcus, Ethanoligenens, Faecalibacterium, Fusobacterium, Gracilibacter, Guggenheimella, Hespellia, Lachnobacterium, Lachnospira, Lactobacillus, Leuconostoc, Megamonas, Moryella, Mitsuokella, Oribacterium, Oxobacter, Papillibacter, Proprionispira,Pseudobutyrivibrio, Pseudoramibacter, Roseburia, Ruminococcus, Sarcina, Seinonella, Shuttleworthia, Sporobacter, Sporobacterium, Streptococcus, Subdoligranulum, Syntrophococcus, Thermobacillus, Turibacter and Weisella.

In an example, the carrier bacteria, composition, use or method is for reducing pathogenic infections or for re-balancing gut or oral biofilm eg, for treating or preventing obesity or disease in a human or animal; or for treating or preventing a GI condition (such as Crohn’s disease, IBD or colitis). For example, the vector, carrier bacteria, composition, use or method is for knocking-down Salmomnella, Campylobacter, Erwinia, Xanthomonous, Edwardsiella, Pseudomonas, Klebsiella, Pectobacterium, Clostridium dificile or E coli bacteria in a gut biofilm of a human or animal or a plant, preferably in a human or animal.

In an example, the animal is a chicken, eg, and the target bacteria are Salmomnella or Campylobacter. In an example, the animal is a fish (eg, catfish or salmon) or shellfish (eg, prawn or lobster), eg, and the target bacteria are Edwardsiella. In an example, the plant is a potato plant and, eg, the target bacteria are Pectobacterium. In an example, the plant is a cabbage plant and, eg, the target bacteria are Xanthomonous (eg, X campestris). In an example, the plant is a marijuana plant and, eg, the targt bacteria are Pseudomonas (eg, P cannabina or P amygdali), Agrobacterium (eg, A tumefaciens) or Xanthomonas (eg, X campestris). In an example, the plant is a hemp plant and, eg, the targt bacteria are are Pseudomonas (eg, P cannabina or P amygdali), Agrobacterium (eg, A tumefaciens) or Xanthomonas (eg, X campestris).

Optionally, the environment is comprised by, or the target bacteria are comprised by, a gut biofilm, skin biofilm, oral cavity biofilm, throat biofilm, hair biofilm, armpit biofilm, vaginal biofilm, rectal biofilm, anal biofilm, ocular biofilm, nasal biofilm, tongue biofilm, lung biofilm, liver biofilm, kidney biofilm, genital biofilm, penile biofilm, scrotal biofilm, mammary gland biofilm, ear biofilm, urethra biofilm, labial biofilm, organ biofilm or dental biofilm. Optionally, the environment is comprised by, or the target bacteria are comprised by, a plant (eg, a tobacco, crop plant, fruit plant, vegetable plant or tobacco, eg on the surface of a plant or contained in a plant) or by an environment (eg, soil or water or a waterway or acqueous liquid). Diseases & Conditions

In an example, the carrier cell(s) or composition is for treating a disease or condition in an animal or human. In an example, the disease or condition is caused by or mediated by the presence of a protein or metabolite in the human or animal subject and expression of P1 by target cells comprising the vector nucleic acid causes a reduction in the protein or metabolite in the subject. In an example, the disease or condition is caused by or mediated by the absence of a protein or metabolite in the human or animal subject and expression of P1 by target cells comprising the vector nucleic acid causes an increase in the protein or metabolite in the subject. In an example, the disease or condition is caused by an undesirably high level of a protein or metabolite in the human or animal subject and expression of P1 by target cells comprising the vector nucleic acid causes an decrease in the protein or metabolite in the subject. In an example, the disease or condition is caused by an undesirably low level of a protein or metabolite in the human or animal subject and expression of P1 by target cells comprising the vector nucleic acid causes an increase in the protein or metabolite in the subject. The metabolite may be a protein, peptide, amino acid, carbohydrate, sugar, lipid, fatty acid or ion (eg, a metal ion). The metabolite may be toxic to human cells of the human subject. The metabolite may be a hormone, growth factor or antibiotic. The metabolite may be a mineral. The metabolite may be a salt. The metabolite may be a nucleic acid, eg, a RNA (eg, a mRNA) or DNA.

In an example, the disease or condition is a cancer, inflammatory or autoimmune disease or condition, eg, obesity, diabetes IBD, a GI tract condition or an oral cavity condition.

Optionally, the disease or condition of a human or animal subject is selected from

(a) A neurodegenerative disease or condition;

(b) A brain disease or condition;

(c) A CNS disease or condition;

(d) Memory loss or impairment;

(e) A heart or cardiovascular disease or condition, eg, heart attack, stroke or atrial fibrillation;

(f) A liver disease or condition;

(g) A kidney disease or condition, eg, chronic kidney disease (CKD);

(h) A pancreas disease or condition;

(i) A lung disease or condition, eg, cystic fibrosis or COPD;

(j) A gastrointestinal disease or condition;

(k) A throat or oral cavity disease or condition;

(l) An ocular disease or condition;

(m) A genital disease or condition, eg, a vaginal, labial, penile or scrotal disease or condition; (n) A sexually-transmissible disease or condition, eg, gonorrhea, HIV infection, syphilis or Chlamydia infection;

(o) An ear disease or condition;

(p) A skin disease or condition;

(q) A heart disease or condition;

(r) A nasal disease or condition

(s) A haematological disease or condition, eg, anaemia, eg, anaemia of chronic disease or cancer;

(t) A viral infection;

(u) A pathogenic bacterial infection;

(v) A cancer;

(w) An autoimmune disease or condition, eg, SLE;

(x) An inflammatory disease or condition, eg, rheumatoid arthritis, psoriasis, eczema, asthma, ulcerative colitis, colitis, Crohn’s disease or IBD;

(y) Autism;

(z) ADHD;

(aa) Bipolar disorder;

(bb) ALS [Amyotrophic Lateral Sclerosis];

(cc) Osteoarthritis;

(dd) A congenital or development defect or condition;

(ee) Miscarriage;

(ff) A blood clotting condition;

(gg) Bronchitis;

(hh) Dry or wet AMD;

(ii) Neovascularisation (eg, of a tumour or in the eye);

(jj) Common cold;

(kk) Epilepsy;

(11) Fibrosis, eg, liver or lung fibrosis;

(mm) A fungal disease or condition, eg, thrush;

(nn) A metabolic disease or condition, eg, obesity, anorexia, diabetes, Type I or Type II diabetes.

(oo) Ulcer(s), eg, gastric ulceration or skin ulceration;

(pp) Dry skin;

(qq) Sjogren’s syndrome;

(rr) Cytokine storm;

(ss) Deafness, hearing loss or impairment; (tt) Slow or fast metabolism (ie, slower or faster than average for the weight, sex and age of the subject);

(uu) Conception disorder, eg, infertility or low fertility;

(vv) Jaundice;

(ww) Skin rash;

(xx) Kawasaki Disease;

(yy) Lyme Disease;

(zz) An allergy, eg, a nut, grass, pollen, dust mite, cat or dog fur or dander allergy;

(aaa) Malaria, typhoid fever, tuberculosis or cholera;

(bbb) Depression;

(ccc) Mental retardation;

(ddd) Microcephaly;

(eee) Malnutrition;

(fff) Conjunctivitis;

(ggg) Pneumonia;

(hhh) Pulmonary embolism;

(iii) Pulmonary hypertension;

(jjj) A bone disorder;

(kkk) Sepsis or septic shock;

(111) Sinusitus;

(mmm) Stress (eg, occupational stress);

(nnn) Thalassaemia, anaemia, von Willebrand Disease, or haemophilia;

(ooo) Shingles or cold sore;

(ppp) Menstruation;

(qqq) Low sperm count.

Neurodegenerative or CNS diseases or conditions for treatment or prevention by the invention In an example, the neurodegenerative or CNS disease or condition is selected from the group consisting of Alzheimer disease , geriopsychosis, Down syndrome, Parkinson's disease, Creutzfeldt- jakob disease, diabetic neuropathy, Parkinson syndrome, Huntington's disease, Machado-Joseph disease, amyotrophic lateral sclerosis, diabetic neuropathy, and Creutzfeldt Creutzfeldt- Jakob disease. For example, the disease is Alzheimer disease. For example, the disease is Parkinson syndrome.

In an example, wherein the method of the invention is practised on a human or animal subject for treating a CNS or neurodegenerative disease or condition, the method causes downregulation of Treg cells in the subject, thereby promoting entry of systemic monocyte-derived macrophages and/or Treg cells across the choroid plexus into the brain of the subject, whereby the disease or condition (eg, Alzheimer’s disease) is treated, prevented or progression thereof is reduced. In an embodiment the method causes an increase of IFN -gamma in the CNS system (eg, in the brain and/or CSF) of the subject. In an example, the method restores nerve fibre and//or reduces the progression of nerve fibre damage. In an example, the method restores nerve myelin and//or reduces the progression of nerve myelin damage. In an example, the method of the invention treats or prevents a disease or condition disclosed in WO2015136541 and/or the method can be used with any method disclosed in WO2015136541 (the disclosure of this document is incorporated by reference herein in its entirety, eg, for providing disclosure of such methods, diseases, conditions and potential therapeutic agents that can be administered to the subject for effecting treatement and/or prevention of CNS and neurodegenerative diseases and conditions, eg, agents such as immune checkpoint inhibitors, eg, anti- PD-1, anti-PD-Ll, anti-TIM3 or other antibodies disclosed therein).

Cancers for treatment

Cancers that may be treated include tumours that are not vascularized, or not substantially vascularized, as well as vascularized tumours. The cancers may comprise non-solid tumours (such as haematological tumours, for example, leukaemias and lymphomas) or may comprise solid tumours. Types of cancers to be treated with the invention include, but are not limited to, carcinoma, blastoma, and sarcoma, and certain leukaemia or lymphoid malignancies, benign and malignant tumours, and malignancies e.g., sarcomas, carcinomas, and melanomas. Adult tumours/cancers and paediatric tumours/cancers are also included.

Haematologic cancers are cancers of the blood or bone marrow. Examples of haematological (or haematogenous) cancers include leukaemias, including acute leukaemias (such as acute lymphocytic leukaemia, acute myelocytic leukaemia, acute myelogenous leukaemia and myeloblasts, promyeiocytic, myelomonocytic, monocytic and erythroleukaemia), chronic leukaemias (such as chronic myelocytic (granulocytic) leukaemia, chronic myelogenous leukaemia, and chronic lymphocytic leukaemia), polycythemia vera, lymphoma, Hodgkin's disease, non-Hodgkin's lymphoma (indolent and high grade forms), multiple myeloma, Waldenstrom's macroglobulinemia, heavy chain disease, myeiodysplastic syndrome, hairy cell leukaemia and myelodysplasia.

Solid tumours are abnormal masses of tissue that usually do not contain cysts or liquid areas. Solid tumours can be benign or malignant. Different types of solid tumours are named for the type of cells that form them (such as sarcomas, carcinomas, and lymphomas). Examples of solid tumours, such as sarcomas and carcinomas, include fibrosarcoma, myxosarcoma, liposarcoma, chondrosarcoma, osteosarcoma, and other sarcomas, synovioma, mesothelioma, Ewing's tumour, leiomyosarcoma, rhabdomyosarcoma, colon carcinoma, lymphoid malignancy, pancreatic cancer, breast cancer, lung cancers, ovarian cancer, prostate cancer, hepatocellular carcinoma, squamous eel! carcinoma, basal cell carcinoma, adenocarcinoma, sweat gland carcinoma, medullary thyroid carcinoma, papillary thyroid carcinoma, pheochromocytomas sebaceous gland carcinoma, papillary carcinoma, papillary adenocarcinomas, medullary carcinoma, bronchogenic carcinoma, renal cell carcinoma, hepatoma, bile duct carcinoma, choriocarcinoma, Wilms' tumour, cervical cancer, testicular tumour, seminoma, bladder carcinoma, melanoma, and CNS tumours (such as a glioma (such as brainstem glioma and mixed gliomas), glioblastoma (also known as glioblastoma multiforme) astrocytoma, CNS lymphoma, germinoma, medulloblastoma, Schwannoma craniopharyogioma, ependymoma, pineaioma, hemangioblastoma, acoustic neuroma, oligodendroglioma, menangioma, neuroblastoma, retinoblastoma and brain metastases).

Autoimmune disease for treatment or prevention

I- Acute Disseminated Encephalomyelitis (ADEM)

2. Acute necrotizing hemorrhagic leukoencephalitis

3- Addison’s disease

4. Agammaglobulinemia

5. Alopecia areata

6- Amyloidosis

7. Ankylosing spondylitis

8. Anti-GBM/Anti-TBM nephritis

9. Antiphospholipid syndrome (APS)

10- Autoimmune angioedema

I I- Autoimmune aplastic anemia

12. Autoimmune dysautonomia

13. Autoimmune hepatitis

14. Autoimmune hyperlipidemia

15. Autoimmune immunodeficiency

16. Autoimmune inner ear disease (AIED)

17. Autoimmune myocarditis

18. Autoimmune oophoritis

19. Autoimmune pancreatitis

20. Autoimmune retinopathy

21. Autoimmune thrombocytopenic purpura (ATP)

22. Autoimmune thyroid disease 23. Autoimmune urticaria

24. Axonal & neuronal neuropathies

25. Balo disease

26. Behcet’s disease

27. Bullous pemphigoid

28. Cardiomyopathy

29. Castleman disease

30- Celiac disease

31- Chagas disease

32. Chronic fatigue syndrome

33. Chronic inflammatory demyelinating polyneuropathy (CIDP)

34. Chronic recurrent multifocal ostomyelitis (CRMO)

35. Churg-Strauss syndrome

36. Cicatricial pemphigoid/benign mucosal pemphigoid

37. Crohn’s disease

38. Cogans syndrome

39. Cold agglutinin disease

40. Congenital heart block

41- Coxsackie myocarditis

42. CREST disease

43. Essential mixed cryoglobulinemia

44. Demyelinating neuropathies

45. Dermatitis herpetiformis

46. Dermatomyositis

47. Devic’s disease (neuromyelitis optica)

48. Discoid lupus

49. Dressier’s syndrome

50. Endometriosis

51- Eosinophilic esophagitis

52. Eosinophilic fasciitis

53. Erythema nodosum

54. Experimental allergic encephalomyelitis

55. Evans syndrome

56. Fibromyalgia

57. Fibrosing alveolitis

58. Giant cell arteritis (temporal arteritis) 59. Giant cell myocarditis

60. Glomerulonephritis

61. Goodpasture’s syndrome

62. Granulomatosis with Polyangiitis (GPA) (formerly called Wegener’s Granulomatosis)

63. Graves’ disease

64. Guillain-Barre syndrome

65. Hashimoto’s encephalitis

66. Hashimoto’s thyroiditis

67. Hemolytic anemia

68. Henoch-Schonlein purpura

69. Herpes gestationis

70. Hypogammaglobulinemia

71- Idiopathic thrombocytopenic purpura (ITP)

72. IgA nephropathy

73. IgG4-related sclerosing disease

74. Immunore gulatory lipoproteins

75. Inclusion body myositis

76. Interstitial cystitis

77. Juvenile arthritis

78. Juvenile diabetes (Type 1 diabetes)

79. Juvenile myositis

80. Kawasaki syndrome

81- Lambert-Eaton syndrome

82. Leukocytoclastic vasculitis

83. Lichen planus

84. Lichen sclerosus

85. Ligneous conjunctivitis

86. Linear IgA disease (LAD)

87. Lupus (SLE)

88. Lyme disease, chronic

89. Meniere’s disease

90. Microscopic polyangiitis

91. Mixed connective tissue disease (MCTD)

92. Mooren’s ulcer

93. Mucha-Habermann disease

94. Multiple sclerosis 95. Myasthenia gravis

96. Myositis

97. Narcolepsy

98. Neuromyelitis optica (Devic’s)

99. Neutropenia

100. Ocular cicatricial pemphigoid

101. Optic neuritis

102. Palindromic rheumatism

103. PANDAS (Pediatric Autoimmune Neuropsychiatric Disorders Associated with Streptococcus)

104. Paraneoplastic cerebellar degeneration

105. Paroxysmal nocturnal hemoglobinuria (PNH)

106. Parry Romberg syndrome

107. Parsonnage-Tumer syndrome

108. Pars planitis (peripheral uveitis)

109. Pemphigus

HO. Peripheral neuropathy

111- Perivenous encephalomyelitis

112. Pernicious anemia

113. POEMS syndrome

114. Polyarteritis nodosa

115. Type I, IL & III autoimmune polyglandular syndromes

116. Polymyalgia rheumatica

117. Polymyositis

118. Postmyocardial infarction syndrome

119. Postpericardiotomy syndrome

120. Progesterone dermatitis

121. Primary biliary cirrhosis

122. Primary sclerosing cholangitis

123. Psoriasis

124. Psoriatic arthritis

125. Idiopathic pulmonary fibrosis

126. Pyoderma gangrenosum

127. Pure red cell aplasia

128. Raynauds phenomenon

129. Reactive Arthritis 130. Reflex sympathetic dystrophy

131. Reiter’s syndrome

132. Relapsing polychondritis

133. Restless legs syndrome

134. Retroperitoneal fibrosis

135. Rheumatic fever

136. Rheumatoid arthritis

137. Sarcoidosis

138. Schmidt syndrome

139. Scleritis

140. Scleroderma

141. Sjogren’s syndrome

142. Sperm & testicular autoimmunity

143. Stiff person syndrome

144. Subacute bacterial endocarditis (SBE)

145. Susac’s syndrome

146. Sympathetic ophthalmia

147. Takayasu’s arteritis

148. Temporal arteritis/Giant cell arteritis

149. Thrombocytopenic purpura (TTP)

150. Tolosa-Hunt syndrome

151. Transverse myelitis

152. Type 1 diabetes

153. Ulcerative colitis

154. Undifferentiated connective tissue disease (UCTD)

155. Uveitis

156. Vasculitis

157. Vesiculobullous dermatosis

158. Vitiligo

159. Wegener’s granulomatosis (now termed Granulomatosis with Polyangiitis (GPA).

Inflammatory diseases for treatment or prevention

1. Alzheimer

2. ankylosing spondylitis

3. arthritis (osteoarthritis, rheumatoid arthritis (RA), psoriatic arthritis)

4. asthma 5. atherosclerosis

6. Crohn's disease

7. colitis

8. dermatitis

9. diverticulitis

10. fibromyalgia

11. hepatitis

12. irritable bowel syndrome (IBS)

13. systemic lupus erythematous (SLE)

14. nephritis

15. Parkinson's disease

16. ulcerative colitis.

For example, the composition comprising carrier cells is a human or animal food and/or beverage (eg, mixed in drinking water for livestock consumption). When supplied in a beverage, the vector may be comprised by carrier bacteria, wherein the carrier bacteria are comprised in the beverage at an amount of from 1 x 10 3 to 1 x 10 10 (eg, from 1 x 10 4 to 1 x 10 10 ; from 1 x 10 4 to 1 x 10 9 ; from 1 x 10 4 to 1 x 10 8 ; from 1 x 10 4 to 1 x 10 7 ; from 1 x 10 3 to 1 x 10 10 ; from 1 x 10 3 to 1 x 10 9 ; from 1 x 10 3 to 1 x 10 8 ; from 1 x 10 3 to 1 x 10 7 ; from 1 x 10 5 to 1 x 10 10 ; from 1 x 10 5 to 1 x 10 9 ; from 1 x 10 5 to 1 x 10 8 ; from 1 x 10 5 to 1 x 10 7 ; from 1 x 10 6 to 1 x 10 10 ; from 1 x 10 6 to 1 x 10 9 ; from 1 x 10 6 to 1 x 10 8 ; or from 1 x 10 6 to 1 x 10 7 ) cfu/ml. When supplied in a beverage, the vector may be comprised by carrier bacteria, wherein the carrier bacteria are comprised in the beverage at an amount of at least 1 x 10 8 cfu/ml, eg, wherein the subject is a human or animal (eg, a poultry bird, such as a chicken).

Optionally, the guided nuclease is any guided nuclease disclosed herein, eg, a Cas, TALEN, meganuclease or a zinc finger nuclease. In an example, the component is a crRNA or guide RNA that is operable in target cells with a cognate Cas nuclease. The Cas nuclease can be any Cas nuclease disclosed herein. The Cas nuclease may be an endogenous Cas of the target cells or may be encoded by an exogenous nucleic acid that is administered to the animal.

There is provided according to the Fifth Configuration:

A method of engineering a microbiome (eg, any microbiome disclosed herein), the method comprising contacting the microbiome with a plurality of vectors as described herein (eg, by combining the microbiome with carrier cells as described) and optionally allowing transfer of said vector nucleic acid into target cells of the microbiota. A modified microbiota obtained or obtainable by the method herein, optionally wherein the microbiota is comprised by a pharmaceutical composition for use as a medicament to treat a disease or condition in a human or animal subject.

Any of the features disclosed in the context of the First to Fourth Configurations may apply mutatis mutandis to the Fifth Configuration.

The invention also provides the following Concepts.

1. A host cell comprising nucleic acid that comprises

(a) an expressible nucleotide sequence of interest (NS 1) for producing a product of interest (P1) in the host cell; and

(b) an expressible nucleotide sequence (NS2) for producing a regulator product (P2) in the host cell, wherein P2 is operable in the host cell to regulate expression or activity of P1 ; wherein

(c) NS 1 is under the control of a first promoter (eg, a constitutive or inducible promoter) for expression of P1; and

(d) expression or activity of P2 in the host cell is regulatable by exposure of the host cell comprising the at least one vector to a regulator agent (R), thereby regulating the expression or activity of P1; wherein the host cell is a bacterial, archaeal or fungal cell.

Optionally, the nucleic acid is comprised by at least one nucleic acid vector for transfer from the host cell into a cell of a microbiota. For example the host cell and microbiota cell are bacterial cells.

For example, the fungal cell is a yeast cell. The bacterial cell may be a cell of any bacterial species or genus disclosed herein.

2. The cell of Concept 1, wherein

(a) the nucleic acid is comprised by at least one nucleic acid vector of the cell;

(b) NS1 is comprised by a nucleic acid vector of the cell and NS2 is comprised by a chromosome of the cell;

(c) NS1 is comprised by a chromosome of the cell and NS2 is comprised by a nucleic acid vector of the cell; or (d) NS1 is comprised by a chromosome of the cell and NS2 is comprised by a chromosome of the cell.

3. The cell of any one of Concept 2(a)-(c) wherein the vector or each vector is a conjugative plasmid for transfer to a cell of a microbiota comprised by a human or animal subject.

4. The cell of any preceding Concept, wherein the cell is a cell of commensal or probiotic bacterial species of a human or animal microbiota, optionally an E coli cell or a Bacteroides cell.

5. The cell of any preceding Concept, wherein NS2 is under the control of a second promoter that is regulatable for expression of P2, wherein binding of a regulator agent (R) to the vector nucleic acid regulates the second promoter, thereby regulating the expression of P2 and P1.

6. The cell of any preceding Concept, wherein P2 comprises an RNA-guided nuclease (optionally a Cas nuclease), wherein the nuclease is operable to cut the nucleic acid at a predetermined sequence motif.

7. The cell of Concept 6, wherein

(a) the sequence motif is comprised by a chromosome of the cell and the cutting kills the cell; or wherein the sequence motif is comprised by a gene (on a chromosome or vector of the cell) comprising NS1 for production of P1 and the cutting down-regulates the production of P1; or

(b) the sequence motif is comprised by a said vector comprising NS 1.

This is useful for promoting degradation of the vector, thereby reducing or inhibiting the production of P1. For example, the cut vector is degraded in the cell.

8. The cell of any preceding Concept for treating or preventing a disease or condition in a human or animal subject, wherein the cell is administered to a microbiota (optionally a gut microbiota) of the subject to produce P1 in the subject thereby treating or preventing the disease or condition in the subject.

Regulation by Xylitol or Xylose

Over half of ingested xylitol in humans is not adsorbed by human cells instead reaching the gastrointestinal tract where it is taken up by the microbiome (Livesey, 2003), making this useful to control our switches in the GI tract. As exemplified herein, we advantageously found that xylitol induces expression by lifting the repression that the transcriptional regulator exerts on the promoter. Furthermore, E. coli strains generally lack the xylose reductase and xylitol dehydrogenase typically necessary for xylitol metabolism (Ge et al., 2018). We realised that this may be useful for more precise or lasting control by xylitol when the strain cannot metabolise xylitol. Use of lower doses of xylitol may be possible when the strain cannot metabolise xylitol. Thus, in an example the cell of the invention is an E coli cell and the promoter is regulatable by xylitol.

1. A cell (optionally according to any preceding Concept), comprising a nucleic acid, wherein the nucleic acid comprises a gene encoding a product of interest (P1), the gene comprising a nucleotide sequence (NS1) encoding P1 and a regulatory region 5’ of NS 1 that comprises a promoter (Px) for controlling the expression ofNSl, wherein the combination of Px and NS1 is heterologous to the cell and Px is regulatable by xylitol or xylose.

For example, NS 1 is not found in a wild-type cell of the same species as the cell of the invention. For example, NS 1 is a non-bacterial (eg, an animal, human, mammal or plant) sequence. Preferably, NS1 is a human sequence.

For example, NS1 is an endogenous sequence of the cell and Px is heterologous to the cell.

2. The cell of Concept 1, wherein the promoter is a xylitol or xylose regulatable promoter of a Morganella species, optionally M morganii.

For example, the M morganii is Morganella morganii strain ZJG812.

3. The cell of Concept 1 or 2, wherein the promoter comprises SEQ ID NO: 3 or a nucleotide sequence that is at least 70% identical to SEQ ID NO: 3.

For example, any percent identity herein is at least 70, 80, 90, 95, 96, 97, 98 or 99%.

Optionally, the cell comprises SEQ ID NO: 4 or a nucleotide sequence encoding a xylitol regulatable promoter. For example, the sequence further encodes a repressor that is cognate to the promoter. For example, the sequence further encodes a xylitol isomerase.

4. The cell of any one of Concepts 1-3, wherein Px is homologous to a xylitol or xylose regulatable promoter of Morganella morganii. For example, the promoter is a xylitol regulatable promoter. For example, the promoter is repressible by a repressor and xylitol is capable of de-repressing the repressor.

5. The cell of any one of Concepts 1-4, wherein the cell genome encodes a repressor that is capable of repressing Px, wherein xylitol and/or xylose is capable of de-repressing the repressor.

6. The cell of Concept 5, wherein the repressor is encoded by SEQ ID NO: 1 or a nucleotide sequence that is at least 70% identical to SEQ ID NO: 1.

For example, the identity is at least 70, 80, 90, 95, 96, 97, 98 or 99%.

For example, the repressor comprises SEQ ID NO: 2 or an amino acid sequence that is at least 70% identical to SEQ ID NO: 2.

7. The cell of any one of Concepts 1-6, wherein the cell comprises a xylitol transporter, optionally a xylitol ABC transporter.

Our data surprisingly suggest that the presence of a xylitol transporter increases the sensitivity of the donor strains towards xylitol in the growth medium.

8. The cell of any one of Concepts 1-7, wherein the cell is devoid of a xylitol isomerase gene.

For example, the cell genome comprises a xylitol inducible promoter of aMorganii species and excludes a sugar isomerase gene. For example, the cell comprises a xylitol inducible promoter of a Morganii species and excludes a nucleotide sequence encoding a xylitol isomerase.

In an example, the cell genome comprises a xylABC operon from Morganella morganii. Optionally the operon is devoid of a nucleotide sequence encoding a xylitol isomerase.

9. A nucleic acid vector comprising a gene as recited in any one of Concepts 1-8.

10. The vector of Concept 9, wherein the vector is a plasmid (optionally a conjugative plasmid), transposon, phagemid or a phage.

In an example, the cell of the invention comprises any vector of the invention as described herein.

It will be understood that particular embodiments described herein are shown by way of illustration and not as limitations of the invention. The principal features of this invention can be employed in various embodiments without departing from the scope of the invention. Those skilled in the art will recognize, or be able to ascertain using no more than routine study, numerous equivalents to the specific procedures described herein. Such equivalents are considered to be within the scope of this invention and are covered by the claims. All publications and patent applications mentioned in the specification are indicative of the level of skill of those skilled in the art to which this invention pertains. All publications and patent applications and all US equivalent patent applications and patents are herein incorporated by reference to the same extent as if each individual publication or patent application was specifically and individually indicated to be incorporated by reference. The use of the word "a" or "an" when used in conjunction with the term "comprising" in the claims and/or the specification may mean "one," but it is also consistent with the meaning of "one or more," "at least one," and "one or more than one." The use of the term "or" in the claims is used to mean "and/or" unless explicitly indicated to refer to alternatives only or the alternatives are mutually exclusive, although the disclosure supports a definition that refers to only alternatives and "and/or." Throughout this application, the term "about" is used to indicate that a value includes the inherent variation of error for the device, the method being employed to determine the value, or the variation that exists among the study subjects.

As used in this specification and claim(s), the words "comprising" (and any form of comprising, such as "comprise" and "comprises"), "having" (and any form of having, such as "have" and "has"), "including" (and any form of including, such as "includes" and "include") or "containing" (and any form of containing, such as "contains" and "contain") are inclusive or open-ended and do not exclude additional, unrecited elements or method steps

The term "or combinations thereof' or similar as used herein refers to all permutations and combinations of the listed items preceding the term. For example, "A, B, C, or combinations thereof is intended to include at least one of: A, B, C, AB, AC, BC, or ABC, and if order is important in a particular context, also BA, CA, CB, CBA, BCA, ACB, BAC, or CAB. Continuing with this example, expressly included are combinations that contain repeats of one or more item or term, such as BB, AAA, MB, BBC, AAABCCCC, CBBAAA, CABABB, and so forth. The skilled artisan will understand that typically there is no limit on the number of items or terms in any combination, unless otherwise apparent from the context.

Any part of this disclosure may be read in combination with any other part of the disclosure, unless otherwise apparent from the context. All of the compositions and/or methods disclosed and claimed herein can be made and executed without undue experimentation in light of the present disclosure. While the compositions and methods of this invention have been described in terms of preferred embodiments, it will be apparent to those of skill in the art that variations may be applied to the compositions and/or methods and in the steps or in the sequence of steps of the method described herein without departing from the concept, spirit and scope of the invention. All such similar substitutes and modifications apparent to those skilled in the art are deemed to be within the spirit, scope and concept of the invention as defined by the appended claims.

EXAMPLES

EXAMPLE 1: Construction of conjugative and self-targeting production CGV

Background to Example 1

• The pBBRl based plasmid used contains an oriT that is mobilizable by the RP4 plasmid transfer machinery carried in trans in the E. coli JKE201 strain (see: Harms, A., Liesch, M., Komer, J., Quebatte, M., Engel, P. and Dehio, C., 2017. A bacterial toxin-antitoxin module is the origin of inter-bacterial and inter-kingdom effectors of Bartonella. PLOS Genetics, 13(10), p.e 1007077 ) which will harbour the plasmid.

• The genes encoding the E. coli Type I-E Cas3 were cloned under the control of a pBad promoter (which is induced by exposure to arabinose). A nucleotide sequence encoding Superfolder Green Fluorescent Protein (sfGFP) and a sfGFP -targeting CRISPR array controlled by a rhamnose inducible promoter will be inserted into the replicon. The array will encode one or more crRNAs for the formation of guide RNAs, that are cognate with the Cas3 and operable to target the sfGFP sequence to guide the Cas3 to the sequence and cut it. P1asmid Components:

• pBBRl -msc2, a mobilizable shuttle and expression vector;

• oriT from the RP4 plasmid;

• A Kanamycin antibiotic resistance marker;

• rmtB (16S rRNA methylase gene) which gives resistance to amikacin and gentamicin

• E coli CRISPR/cas system with pBAD promoter, wherein the system comprises nucleotide sequences encoding Type I-E Cas3, Cascade proteins A-E under the control of a pBAD promoter, the system also comprising the CRISPR array;

• S. pyogenes terminator after the Cascade.

• rhamnose promoter in front of the CRISPR array, which is programmed to target the sfGFP gene (contains one spacer that can hybridise to the sfGFP sequence); and constitutively expressed sfGFP gene.

The order of the components:

Kanamycin resistance gene, pBBRl-msc2, oriT from RP4 plasmid, araC, pBad promoter, E. coli Cas3 Type I-E sequence, Cascade sequence, S. pyogenes terminator, rhamnose promoter, array comprising spacer that can hybridise to the sfGFP sequence, rmtB (16S rRNA methylase gene) which gives resistance to amikacin and gentamicin, sfGFP gene

Methods, Cloning:

• Backbone pl 075 containing the pBBRl replicon and CRISPR cas system, was used as a template for cloning sfGFP gene, S. pyogenes terminator after the E. coli Type I-E Cas3 and Cascade, CRISPR array with sfGFP target and rhamnose promoter to control CRISPR array with sfGFP target (see plasmid map Fig. 1).

• Transformation, selection and sequence verification

Transformation technique: The final construct (pl 364) is electroporated to the strain of interest (NEB 10-beta cells for cloning or JKE201 strain for further conjugation). Correct colonies are selected on 3 antibiotic markers (kanamycin, gentamicin, amikacin), colonies will have a distinctive green color (due to expression of GFP) and with confirmation using colony PCR with the construct specific primers.

Sequence verification is performed by subjecting the purified plasmid to sequencing with construct specific primers to cover the whole construct.

See Figure 1 for a plasmid map highlighting the main components of the final plasmid, pl364.

EXAMPLE 2: Conjugative delivery from E. coli donor strain JKE201 to recipient E. coli strain b52

Methods:

The JKE201 strain was transformed with pl 364 and a positive clone was selected on kanamycin LB plates (supplemented with DAP) and further verified by the positive GFP signal. One colony of the transformed JKE201/pl364 donor strain as well as the b52 (strain C-la-CGSC, obtained from the Coli Genetic Stock Center) recipient strain was grown to exponential phase in liquid LB (JKE201 supplemented with DAP). The two cultures were then mixed 5: 1 (JKE201/pl364 : b52) and spotted solid LB agar to incubate overnight.

After overnight conjugation, the mix was scraped off and dissolved in liquid LB and plated on LB plates containing kanamycin but without DAP to select only b52 transconjugants. These were enumerated compared to total recipient counts on LB plates.

Results are depicted in Fig. 2.

The pl364 transfers at high frequency into the b52 model strain. Fig. 2 shows the number of transconjugants formed as a result of the RP4 oriT on pl364 transfer and the resultant absence of transconjugants when oriT is absent.

EXAMPLE 3: Production of protein of interest from mobilizable plasmid in ‘production bacteria’ strain (recipient strain) b52

Transconjugant recipient cells (b52 cells containing plasmid p 1364) were produced as described in Example 2. The transconjugants, containing the sfGFP expressing construct with a self-targeting CRISPR array, conjugative plasmid pl364, was grown in LB medium to and OD600 of 1. A plasmid- free b52 strain was grown to the same level for use as a negative/autofluorescence control. Each culture was transferred to a mictrotiter plate and measured in a Synergy™ Hl plate reader. sfGFP production was quantified using and excitation wavelength of 485nm and an emission wavelength of 510nm.

Results are depicted in Fig. 3. GFP fluorescence was determined in in b52 cells harbouring the pl364 plasmid and in b52 control cells devoid of the plasmid at OD=0.7. The GFP fluorescence was approximately five times higher than the background fluorescence of the b52 control strain, showing reliable expression of sfGFP.

EXAMPLE 4: Removal of conjugative plasmid from recpient strain b52

Transconjugant b52 cells (from Example 2) carrying the pl364 plasmid were grown with selection for pl 364 (kanamycin) for 2h and subsequently washed in LB to remove residual antibiotics.

The resuspended culture was then split in two 1ml portions and inducers (1% arabinose, lOmM of rhamnose) were added to one of the tubes to induce the self-targeting Cas mechanism. Just after induction, cells from both cultures were plated on LB plates (without inducers added, timepoint 0). Subsequently, samples from each tube (induced and non-induced) were plated on LB plates at timepoints: Ih and 24hrs after induction. All plates were incubated overnight after which the proportion of sfGFP expressing cells was determined by colony counting in a blue light transilluminator (365nm, UV benchtop transilluminator, VWR®). Results are depicted in Fig. 4 and shows the proportion of GFP positive cells over time for induced and non-induced cultures. The induction of the CRIPSR system and corresponding array on p 1364 allows for rapid curing of the self-targeting plasmid. The system allows for >70% plasmid curing within 1 h and >95% of the plasmid purged within 24 hrs.

To validate that the loss of sfGFP was indeed due to plasmid loss, PCR specific for the plasmid backbone was performed on 14 randomly selected colonies from the plate of the 24h timepoint. See the resulting gel image in Fig. 5.

EXAMPLE 5: Killing donor strain by CRISPR

A conjugative plasmid construct was made that could be introduced into host cells to produce donor cells. The plasmid will be a vector for a gene of interest (eg, GFP in the examples above) and a selftargeting CRISPR/array to target the plasmid in recipient cells after it has been transferred by conjugation from donor to recipient cells.

The plasmid was engineered so that, on induction of the CRISPR/cas system with arabinose, the plasmid was able to target the genome of the host cells. The plasmid was made using the RP4 plasmid as a starting point. The arabinose inducible E. colt Type I-E Cas3 Cascade followed by a CRISPR array for producing a guide RNA targeting the E. coli chromosomal gene IptA was integrated into the RP4 plasmid using recombineering.

The Cas system and array with the arabinose promoter was amplified along with a spectinomycin resistance gene and araC from a cloning vector containing these.

The PCR product were flanked with homology arms that allowed insertion of the CRISPR/cas-araC- Spectinomycin cassette into the tetracycline gene of RP4.

Recombineering was performed. Breifly, JKE201 cells with an IptA mutant allele, not targeted by the array, that contained apSIM5 recombineering plasmid were grown to an OD of 0.5 in LB supplemented with DAP and chloramphenicol. Then, the lambda RED system was induced at 42C degrees for 15min and the culture was placed on ice to prepare the cells for transformation by washing in cold MiliQ water. Cells were transformed with the PCR product at 18kv and recovered for 12h prior to plating on selective plates containing DAP and spectinomycin. The resulting recombinant clones were confirmed by PCR.

A positive clone as well as JKE201 with the wildtype RP4 plasmid (negative control without CRISPR/Cas) was grown in liquid LB with spectinomycin for 4hrs. Wildtype MG1655 cells were grown to the same OD and mixed with the JKE201/RP4::Cas and JKE201/RP4 to allow plasmid transfer and amplification in this strain. The mixes were resuspended in LB without DAP to remove the initial JKE201 donor strain. The resulting population of MG1655 (newly formed plasmid donor cells) containing the plasmid were now plated on LB plates with and without arabinose (arabinose for CRISPR/cas induction) to active self-targeting and killing of donor cells by cutting the IptA target comprised by the donor chromosome. The population containing the wildtype RP4 plasmid was plated on the same media as a control. After overnight incubation, the surviving cells were enumerated and presented in Figure 6.

The results depicted in Fig. 6 demonstrate that robust counterselection of a strain with one or more targets of interest can be done upon induction of CRISPR/cas. The engineered conjugative plasmid presented here, was able to discriminate between two closely related strains in a mixed culture, suggesting that it will be useful for precise donor strain removal in a microbiome context, such as in vivo in a human or animal subject. With additional targets added to the array, multiple targets can be hit simultaneously. A multi-target approach is the ideal implementation of a tight biocontainment strategy, essentially combining the two approaches of donor cell genome and plasmid targeting shown here, that will allow complete removal of the product (pathway) of interest from the microbiome in a controllable manner.

EXAMPLE 6: Illustrative pathways Employing the vectors, cells and methods of the invention

Various metabolic pathway schemes can be envisaged where the invention functions. For example, the pathway may be in any environment comprising a microbiota, such as in a human or animal subject. Examples of schemes are provided in Figure 7. The pathway scheme may function in the subject, such as confined to the microbiota (or even confined within the target cell that has received the vector nucleic acid encoding P1 and P2). Alternatively, part of the scheme (at least a part involving the target cell) takes place in the microbiota and another part takes place outside the microbiota in the environment or subject, such as where a metabolite is secreted from the target cell and is metabolized in the downstream part of the pathway scheme. In an embodiment, R may be produced as a metabolite (in the target cell or in a different cell, such as a different cell of the microbiota), thereby providing Rto regulate P2 (and thus P1) expression in a feedback loop.

R may upregulate P2 expression, P2 downregulates P1 expression and optionally P1 is a component in a metabolic pathway (eg, in the microbiota or target cell or in a subject or environment comprising the microbiota or target cell) wherein a production product (X) of the pathway downstream from P1 causes regulation of P1 or P2 expression. See, for example, Scenarios 1, 3 and 4 (Figure 7). In an embodiment, X is R or a precursor of R. In an embodiment, X regulates the first and/or second promoter. In an embodiment, X upregulates P1 or P2 expression. In an embodiment, X upregulates P1 or P2 expression.

R may upregulate P2 expression, P2 upregulates P1 expression and optionally P1 is a component in a metabolic pathway (eg, in the microbiota or target cell or in a subject or environment comprising the microbiota or target cell) wherein a production product (X) of the pathway downstream from P1 causes regulation of P1 or P2 expression. See, for example, Scenario 2 (Figure 7). In an embodiment, X is R or a precursor of R. In an embodiment, X regulates the first and/or second promoter. In an embodiment, X upregulates P1 or P2 expression. In an embodiment, X upregulates P1 or P2 expression.

EXAMPLE 7: Vector removal by CRISPR self-targeting in Bacteroides

1. Executive summary

In this study we developed and tested a plasmid comprising a CRISPR/Cas system (which we call a CRISPR-guided vector (CGV)) that could be removed from a population of Bacteroides thetaiotaomicron using vector-bome spacers targeting the CGV itself (self-targeting CGV). The CGV harbored an inducible type I-B CRISPR/Cas operon originating from Clostridioides difficile alongside five CGV-targeting spacers. We demonstrated that upon induction, the CRISPR/Cas system caused the CGV to be removed from more than 99.9% of the Bacteroides population without killing the host cells in the process.

Objectives

Objective 1

Deliver a self-targeting CGV to Bacteroides thetaiotaomicron VPI-5482 (bSNP2978) via conjugation from an Escherichia coli donor to show that the CGV can be efficiently removed upon CRISPR/Cas induction.

Objective 2

Demonstrate that self-targeting CGVs are removed from a population of Bacteroides thetaiotaomicron VPI-5482 (bSNP2978) without off-target effects.

2 Materials and methods 2.1 Bacterial strains and growth conditions

Escherichia coli strains were grown at 37°C in lysogeny broth (LB) at 250 RPM, or on solid LB-agar plates (made with 1.5% (w/v) agar). For plasmid selection, the medium was supplemented with ampicillin (Amp, 100 pg ml -1 ) or spectinomycin (Spc, 400 pg ml -1 ). For growth of auxotrophic strains, the medium was further supplemented with diaminopimelic acid (DAP, 40 pg ml -1 ).

Bacteroides thetaiotaomicron VPI-5482 (bSNP2978) only grows anaerobically but does not die under aerobic conditions. For this reason, the “M45 variable atmosphere workstation” (Don Whitley, Yorkshire, UK) was used and set to contain a mixture of gasses N2, H2, and CO2 in the ratio 92:6:2.

All solutions and media intended for Bacteroides were allowed to reduce under these conditions for ~24 hours before being used.

All Bacteroides strains were routinely grown at 37°C in Brain Heart Infusion (BHI) broth supplemented with cysteine (1 g I -1 ), hemin (5 mg I” 1 ), and NaHCCL (0.2% (w/v)). This medium, denoted as BHIpp, is based on a recent Bacteroides protocol (Bacic & Smith, 2008). When growth on solid media was required, the BHIpp broth was further supplemented with agar (1.5% (w/v)). For plasmid selection, the medium was supplemented with erythromycin (Erm, 10 pg I -1 ). For counterselection of the E. coli donor strain, the BHIpp medium was supplemented with gentamicin (Gm, 30 Pg l 1 )-

2.2 CGV design and assembly

CGV pSNP1599 is based on a shuttle plasmid, which can be delivered into the Bacteroides thetaiotaomicron VPI-5482 recipient strain (bSNP2978) via conjugation from an E. coli (bSNP3235)- based donor strain.

To construct pSNP1599, five spacers complementary to protospacers found within the plasmid itself (details found in Appendix C) were first assembled into a CRISPR array and then cloned into the shuttle plasmid. In the same cloning reaction, the type I-B CRISPR/Cas operon from the Clostridioides difficile strain 630Aerm (denoted as CdCas) was also cloned into the plasmid. Expression of the vector-bome CRISPR/Cas system was regulated by a rhamnose-inducible promoter (Prha), which allowed the plasmid removal to be controlled. Detailed information on the construction of pSNP1599 is found in Appendix B.

2.3 Conjugative transfer of CGV from E. coli to B. thetaiotaomicron First, the bSNP3235-based donor strain carrying the plasmid of interest was grown aerobically in 30 ml LB medium supplemented with DAP while the bSNP2978 recipient was grown anaerobically in 5 ml BHIpp broth. Once the donor and recipient cultures reached optical densities (ODgoo) of 0.4±0.2 and 0. 15±0.05, respectively, they were mixed, using 25 ml of the donor and 2.5 ml of the recipient. The mixture was spun down (9000 x g for 10 minutes), supernatant removed, and the pellet resuspended in 100 pl PBS. The resuspended pellet was spotted onto a BHIpp agar plate and allowed to incubate aerobically at 37°C for 18 hours to facilitate conjugation.

The mating spot was collected and resuspended in 500 pl pre-reduced BHIpp broth (from this point, the rest of the experiment was performed in the anaerobic work station). A 10-fold serial dilution was prepared in PBS (spanning dilutions 10° to 10’ 6 ) and then spotted onto selective media (BHIpp+Gm+Erm agar) with and without inducer. P1ates were packed in plastics bags and allowed to incubate anaerobically at 37°C for 2-3 days. Any transconjugant colony forming units (CFUs) emerging from the plates were enumerated. Two examples of plates after 3 days of incubation are given in Figure 10.

2.4 Plasmid removal assay

A colony of bSNP2978 containing the intact self-targeting CGV pSNP1599 (verified by sequencing) was inoculated into 5 ml BHIpp broth supplemented with gentamicin and erythromycin and allowed to grow anaerobically overnight. The culture was then washed three times (each wash consisting of a spin-down at 4500 x g for 2 min, removal of supernatant, and resuspension in 5-ml PBS) to remove all traces of erythromycin.

Two 5 -pl aliquots of the washed culture were transferred to separate tubes containing 5 ml fresh prereduced BHIpp broth. One tube was supplemented with the inducer rhamnose (10 mM) while the other tube was given an equal volume of milliQ H2O. The induced and non-induced cultures were allowed to grow anaerobically over the course of a day, with 200-pl aliquots being taken from each culture at 0-, 1-, 3-, 6- and 24 hours after induction.

10-fold serial dilutions were prepared in PBS for each collected aliquot (spanning dilutions 10° to 10" 9 ) and spotted (2.5 pl per dilution) onto BHIpp agar plates either supplemented with gentamicin (non- selective plates) or gentamicin and erythromycin (selective plates). P1ates were packed in plastic bags and allowed to incubate anaerobically for 2-3 days. CFUs were counted. Experiment was performed with biological triplicates. Two examples of plates after 3 days of incubation are given in Figure 11.

3 Results 3.1 Estimating the efficacy of CRISP R-mediated CGV removal

A self-targeting CGV, denoted as pSNP1599, was generated, comprising a type I CRISPR/Cas system from C. difficile (CdCas) and five spacers which target the CGV itself. Upon induction with rhamnose, the CdCas system is activated which lead to CRISPR targeting and, in turn, removal of the CGV.

The removal efficiency was tested by conjugating the CGV into the model strain Bacteroides thetaiotaomicron VPI-5482 (bSNP2978) via a mating with an E. coli (bSNP3235)-based donor strain and plating the mating mixture onto solid medium that selecting for the CGV with and without rhamnose. As a control, the empty plasmid pSNP1380 was conjugated via a separate mating.

Conjugation experiments showed that the CGV was efficiently removed, as 1000-fold less transconjugants emerged on the selective media with induction compared to the plates without induction (~10 4 CFU/ml and ~10 7 CFU/ml, respectively) (Figure 8). Conjugation of the empty control plasmid shows no difference in transconjugants regardless of induction.

3.2 Testing for off-target effects of self-targeting CGVs

To test if the self-targeting CGV is removed without killing the Bacteroides host, we grew bSNP2978 (pSNP1599) with and without rhamnose (Figure 9A, top). At various time points we took out aliquots, made serial dilutions, and spread them onto solid media plates with and without erythromycin (GCV antibiotic marker) (Figure 9A, bottom), allowing us to estimate the sizes of the plasmid-harboring population and the total population, respectively.

One hour after induction, around 99% of the cells in the population lost their CGVs (Figure 9B). This difference is gradually increased throughout the experiment. As expected, for the non-induced culture the number of cells was similar regardless of the selection, suggesting that our CGV was stable in Bacteroides unless we actively remove it by adding the inducer.

4 Conclusions

A new CGV carrying a self-targeting CRISPR/Cas system (CdCas) can efficiently was removed from more than 99.9% of cells in a population of B. thetaiotaomicron upon induction with rhamnose.

It was also demonstrated that survival and growth of the B. thetaiotaomicron cells was unaffected by the induced self-targeting CGV, suggesting that the CRISPR/Cas-mediated CGV removal had no off- target effects. Appendix A: Bacterial strains used in this study bSNP2978 Bacteroides thetaiotaomicron VPI-5482. Deutsche Sammlung von

Anaerobic bacterium known for being abundant in the

Mikroorganismen und human gut.

Zellkulturen GmbH

(DSM No. 2079)

Appendix B: construction and cloning of self-targeting CGV pSNP1599 Design and assembly of CRISPR array

A CRISPR array was designed by identifying five 37-bp protospacers in the backbone of the shuttle plasmid (details in Appendix C), each positioned adjacent to the consensus type I-B PAM sequence (5’-CCW-3’). A preliminary sequence similarity search (BLASTn, not shown) did not predict any off- targets for any of the five chosen spacers to the chromosome of bSNP2978. The repeat sequence (5’- GTTTTATATTAACTAAGTGGTATGTAAAT-3 ’ ) compatible with type I-B CRISPR/Cas system was chosen.

Appendix C: pSNP1599 Based on shuttle plasmid, containing a 6.6-kb type I-B Cas operon from the C. difficile strain 630Aerm (CdCas) and a 359-bp CRISPR-array containing five 37-bp spacers which are designed to target the CGV itself. The entire locus was regulated by a rhamnose-inducible promoter and the consensus RBS sequence used by

Bacteroides spp..

The five spacers are listed below:

Target gene: Spacer sequence (S’— >3’): mobB: 5’-TTCCGGGGAACTGTACCGGGACAACTGGATAGGGAAA-

3’ ampR: 5’-TGTTGTGCAAAAAAGCGGTTAGCTCCTTCGGTCCTCC-

3’ ermG: 5’-AACTATCAAATAGTAAATGATGATATACTGAAATTTA-3’

P rha: 5’-TCAAAACCAATGTAATATTCCGTCCATCCGGTCTGCC-

3’ pSClOl: 5’-GTTCTATTAGGTGTTACATGCTGTTCATCTGTTACAT-

3’

EXAMPLE 8: Evaluation of the xylitol-inducible promoter from Morganella morganii

Executive summary A predicted xylitol-inducible promoter originating from the bacterium Morganella morganii was cloned and investigated within various Escherichia coli strain backgrounds.

It was demonstrated that the promoter is repressed by a transcriptional regulator protein IZ184_04885 and that repression could be lifted either gradually by adding xylitol to the growth medium or completely by mutating the transcriptional regulator. Sensitivity of the promoter towards xylitol increased when the xylitol ABC transporter system was included as well.

Objectives

Objective 1

Demonstrate that the repression of the xylitol-inducible promoter can be lifted by adding increasing amounts of xylitol to the growth medium.

Objective 2

Demonstrate that the repression of the xylitol-inducible promoter is avoided by mutating the transcriptional regulator protein IZ184_04885. Objective 3

Investigate whether expression of the xylitol ABC transporter increases the sensitivity of the xylitol-inducible promoter towards xylitol in the medium.

Materials and methods

Bacterial strains and growth conditions for plasmid cloning

All strains of Escherichia coli used in this study are listed in Table 5, Appendix A. For cloning, E. coli strains were grown at 37°C in lysogeny broth (LB) at 250 RPM or on solid LB-agar plates (made with 1.5% (w/v) agar). For plasmid selection, the medium was supplemented with tetracycline (10 pg ml -1 ) and/or spectinomycin (Spc, 100 pg ml -1 ). P1asmid construction

GFP reporter plasmids

Based on an annotated genome sequence of Morganella morganii strain ZJG812 (Genome ID: CP064831.1), a 1213-bp DNA sequence encoding the xylitol-inducible regulatory system (excluding the sugar isomerase gene IZ184_04880) was defined and ordered as agBlock fragment from Integrated DNA technologies™ (Coralville, IA, USA). The gBlock DNA fragment was further amplified by PCR using the CloneAmp HiFi PCR Premix from Takara Bio (Mountain View, CA, USA) with primers SEM1770 and SEM1771. In parallel, the backbone of the plasmid pSNP1248 was PCR amplified using the CloneAMP HiFi PCR premix with primers SEM1282 and SEM1769, yielding a 3250-bp DNA fragment that contain the fluorescent reporter gene (gfp), a low copy-number origin of replication (cloDF13) and a spectinomycin resistance marker (Spc R ). The full list of primers used in this study is found in Table 6, Appendix B.

The amplified DNA fragments were routinely treated with Dpnl (1 hour at 37°C followed by 20 minutes at 80°C) and purified by gel extraction using the MinElute kit from Qiagen (Hilden, Germany) according to the manufacturer’s instructions. The purified regulatory system DNA was inserted into the linearized pSNP1248 backbone with the In-Fusion® HD Cloning kit from Takara Bio according to the manufacturer’s instruction. Chemically competent bSNP2480 cells were transformed with the DNA from the In-Fusion® reaction mixture. Transformed cells were spread onto LB+Spc 100 agar plates and allowed to incubate overnight. P1asmids were purified from individual colonies using the QIAprep Spin Miniprep Kit from Qiagen according to the manufacturer’s instructions and followingly verified by Sanger sequencing. The positive clone was registered as plasmid pSNP1902. In addition, a serendipitous clone containing a 595 -bp deletion located within the transcriptional regulator open reading frame (ORF) (IZ184_04885) was identified and registered as plasmid pSNP1903. xylitol ABC transporter plasmid

The 3728-bp xylABC operon from Morganella morganii strain ZJG812 (Genome ID: CP064831.1) was ordered as two gBlock DNA fragments from Integrated DNA Technologies™. The plasmid backbone was prepared by PCR amplification of plasmid pSNP639 using the CloneAmp HiFi PCR Premix with primers SEMI 814 and SEM1815. The amplified fragment was Dpnl treated and purified by gel extraction, following the same method as described in section 3.2.1.

The two gBlock DNA fragments and the linearized backbone DNA were fused together using the InFusion® HD Cloning kit from Takara Bio according to the manufacturer’s instruction. Chemically competent bSNP2522 cells were transformed with the DNA from the In-Fusion® reaction mixture. Transformed cells were spread onto LB+Tet 10 agar plates and allowed to incubate overnight. P1asmids were purified from individual colonies using the QIAprep Spin Miniprep Kit from Qiagen according to the manufacturer’s instructions and followingly verified by Sanger sequencing. The positive clone was registered as plasmid pSNP1939. The full list of plasmids used in this study is found in Table 7, Appendix C.

GFP expression assay for characterization of xylitol-inducible promoter

Preparation of model strains

The constructed GFP reporter plasmids (pSNP1902 and pSNP1903), the xylitol ABC transportercontaining plasmid (pSNP1939) as well as the control plasmids (see Table 7, Appendix C) were transformed individually or in combination into the model strains E. colt MG1655 (bSNP230) and a probiotic E. colt isolate (bSNP463) via electroporation. Transformed cells were spread onto LB+Spc 100 - , LB+Tet 10 - or LB+Spc 100 +Tet 10 agar plates depending on whether the cells were transformed with the GFP reporter plasmid, xylitol ABC transporter plasmid, or both, respectively. The plates were allowed to incubate overnight. Three colonies of each strain were picked and used as biological triplicates in the ensuing 24-hour gfp expression assay. 24-hour gfp expression assay

Triplicate colonies were inoculated into 5 -ml LB medium or 5 -ml M9 minimal medium broth (recipe found in Appendix D) supplemented with Tet (10 pg ml -1 ) and/or Spc (100 pg ml -1 ) and allowed to incubate overnight (~16 hours) at 37°C and 250 RPM. Cultures would have reached optical densities (ODgoo) of 0.8±0.3 in M9 minimal medium and 1.4±0.2 in LB medium.

Each overnight culture was then diluted lOOx in fresh medium supplemented with appropriate antibiotics (as above) and distributed into 3-6 new cultures which were given xylitol at different concentrations within the range of 0% to 5% (the exact concentrations of xylitol are listed in the related figures in section 4). The cultures were then transferred as 200-pl aliquots into a black 96-well microtiter plate with transparent bottom. To normalize the fluorescence emissions, a row of wells in the plate were dedicated to cells that harbor the control plasmids pSNP958 and pSNP1617 which do not encode gfp (to estimate autofluorescence) and another row dedicated to media without any cells (to estimate background fluorescence). The microtiter plate was sealed with a Breath-Easy® sealing membrane from Merck (Whitehouse station, NJ, USA) and placed in the Synergy Hl microplate reader from Agilent Biotek (Winooski, VT, USA) programmed to incubate for 24 hours at 37°C with a constant agitation. Both ODgoo and GFP fluorescence (excitation at 485 nm, emission at 516 nm, gain of 90) were measured every 10 minutes throughout the incubation. The recorded fluorescence emissions were normalized according to the corresponding cell densities and corrected for the background and autofluorescence levels.

Results

Criteria and choice of the inducible promoter

We have developed a biocontainment strategy using CRISPR/Cas systems as off-switches of our production circuits or strains. This requires that the CRISPR/Cas system is under tight transcriptional control from a promoter which is activated upon addition of an external inducer molecule.

Within the field of molecular microbiology, operons of genes related to the conversion/utilization of sugars may be induced by the sugars themselves as they bind and influence the transcriptional repressor proteins. For the present study, it was decided to investigate a negatively inducible promoter predicted to be induced by xylitol. This 5-carbon sugar alcohol has several advantages. First of all, xylitol is generally regarded as safe for human consumption by the FDA (Xiang et al., 2021). Furthermore, over half of ingested xylitol is not adsorbed by human cells instead reaching the gastrointestinal tract where it is taken up by the microbiome (Livesey, 2003), making this useful to control our switches in the GI tract. Finally, xylitol is a known metabolite for some bacteria: It is either taken up directly through an ABC-type transporter complex (Madigan et al., 2015) and/or generated through reduction of the corresponding sugar D-xylose. Subsequently, xylitol can be dehydrogenated and phosphorylated to xylulose-5 -phosphate which is further catabolized in the pentose phosphate pathway.

From a dated PhD thesis (Gallo, 1991), we found an operon in the genome of the bacterium Morganella morganii strain ZJG812 which contain genes related to xylitol uptake and metabolism (details summarized in Figure 8(ii)). This operon is located downstream from a ~1800-bp regulatory region which, among other elements, consist of a negatively inducible promoter and a cognate transcriptional regulator protein IZ184_04885 (details in Appendix E) which xylitol is suggested to bind.

Estimating promoter activity in response to xylitol in growth medium

For this study, two reporter plasmids were constructed consisting of a green fluorescent protein (GFP) reporter gene located downstream from the xylitol-inducible regulatory system either without (pSNP1902, Figure 8(i)A) or with (pSNP1903. Figure 8(ii)B) a loss-of-function deletion located in the ORF of the transcriptional regulator.

The reporter plasmids consist of a green fluorescent protein (GFP) reporter gene located downstream from the predicted xylitol-inducible promoter which in turn is controlled by the LacI family DNA- binding transcriptional regulator (IZ184_04885 gene). Two reporter plasmids are made, either (A) with a full-length transcriptional regulator (plasmid pSNP1902) or (B) with a 595-bp loss-of-function (LOF) deletion within the ORF of the transcriptional regulator (plasmid pSNP1903).

The GFP reporter plasmids pSNP1902 and pSNP1903 were transformed into the probiotic isolate strain bSNP463 and tested by measuring the optical density (ODgoo) and green fluorescence emitted from cultures after a 24-hour incubation in the presence of increasing amounts of xylitol. The results from these tests are summarized in Figure 9. It is observed how the fluorescence intensity per cell increases up to 6-fold as xylitol is added to the strains which harbor the GFP reporter plasmid with the full-length transcriptional regulator (Figure 9A). When the transcriptional regulator is mutated by a 595-bp deletion (Figure 9B), the basal fluorescence intensity is approximate 30-fold higher than that from the strains with the full-length transcriptional regulator and the fluorescence intensity does not change when xylitol is added. This suggest that xylitol induces expression by lifting the repression that the transcriptional regulator exerts on the promoter. The test of the GFP reporter plasmid with the full-length transcriptional regulator (pSNP1902) was repeated in a minimal medium broth using the E. coli strain MG1655 (bSNP230) which gave similar results (as shown in Figure 10). Note that the applied concentrations of xylitol were lower for these tests as it was shown that increasing concentrations of xylitol slowed down the growth rate of bacteria when grown on minimal medium supplemented with glycerol. Furthermore, when all other carbon sources than xylitol were omitted from the minimal medium, the E. coli strains did not grow at all (as illustrated from the growth profiles shown in Figure 11). This is likely because E. coli strains generally lack the xylose reductase and xylitol dehydrogenase typically necessary for xylitol metabolism (Ge et al., 2018). This may be useful for more precise or lasting control by xylitol when the strain cannot metabolise xylitol. Use of lower doses of xylitol may be possible when the strain cannot metabolise xylitol.

Testing whether the presence of the xylitol ABC transporter increases the sensitivity of the xylitolinducible promoter

The activity of a negatively inducible promoter will depend on the import of the inducer molecule into the cell. In this context, it was hypothesized that expression of the predicted xylitol ABC transporter operon (mentioned in section 4.1) would allow more xylitol to enter the cell and hence increase the sensitivity of the promoter. To test this, plasmid pSNP1939 was transformed into the bSNP230- construct already containing the GFP reporter plasmid pSNP1902 and tested by the same 24-hour assay as used previously. For these experiments, the well-defined M9 minimal media were used either with glycerol or glucose as the carbon source for growth. Glucose was chosen as it is the preferred sugar for bacterial growth, but it might also have an inhibitory effect on promoters which regulate alternative metabolic pathways such as those related to xylitol. Glycerol was chosen as it had previously been shown by us (data not shown) not to influence the expression from the xylitol-inducible promoter. The results are summarized in Figure 12.

We used bSNP230 strains containing the GFP reporter plasmid (pSNP1902) either with or without a co-resident plasmid containing the xylitol ABC transporter (pSNP1939) were grown for 24-hours in minimal medium supplemented with 0.4% (v/v) glycerol (A) or 0.4% (w/v) glucose (B) in the presence or absence of xylitol. The cell-density adjusted fluorescence emission after 24 hours of incubation are shown above. Error bars indicate standard deviations based on two (A) and three (B) biological replicates, respectively. The corresponding growth curves are shown in Figure 13.

At these low concentrations of xylitol, the strain bSNP230/pSNP1902 did not exhibit any change in fluorescence intensity (but higher concentrations of xylitol in a different experiment may lead to increased fluorescence). For the strain bSNP230/pSNP1902p+pSNP1939, however, it was observed that the fluorescence intensity increased as xylitol was added - an effect which was more pronounced when the minimal medium was supplemented with glycerol (Figure 13A) rather than glucose (Figure 13B) These data suggest that the presence of the xylitol ABC transporter increases the sensitivity of the reporter plasmid-carrying strains towards xylitol in the growth medium.

Conclusions

The xylitol-inducible promoter originating from Morganella morganii is repressed by the transcriptional regulator protein IZ184_04885 when tested in various E. colt strain backgrounds. This repression was lifted gradually by adding xylitol to the growth medium or fully by mutating IZ184_04885. The sensitivity of the promoter towards xylitol in the growth medium was further enhanced by including the xylitol ABC transporter system.

References

Gallo, M. A. (1991). Molecular characterization of xylitol catabolic pathways in the Enterobacteriaceae . Conrell University.

Ge, X., Chang, C., Zhang, L., Cui, S., Luo, X., Hu, S., Qin, Y., & Li, Y. (2018). Conversion of Lignocellulosic Biomass Into P1atform Chemicals for Biobased Polyurethane Application. In Advances in Bioenergy (Vol. 3, pp. 161-213). Elsevier. https://doi.org/10.1016/BS.AIBE.2018.03.002

Livesey, G. (2003). Health potential of polyols as sugar replacers, with emphasis on low glycaemic properties. Nutrition Research Reviews, 16(2), 163-191. https://doi.org/10.1079/nrr200371

Madigan, M. T., Martinko, J. M., Bender, K. S., Buckley, D. H., Stahl, D. A., & Brock, T. (2015).

Microbial Metabolism. In Brock Biology of Microorganisms (14th ed.). Pearson.

Xiang, S., Ye, K., Li, M., Ying, J., Wang, H., Han, J., Shi, L., Xiao, J., Shen, Y., Feng, X., Bao, X., Zheng, Y., Ge, Y., Zhang, Y., Liu, C., Chen, J., Chen, Y., Tian, S., & Zhu, X. (2021). Xylitol enhances synthesis of propionate in the colon via cross-feeding of gut microbiota. Microbiome, 9(1), 1-21. https://doi.org/10.1186/s40168-021-01029-6

Appendices

Appendix A: Bacterial strain backgrounds

Table 5. Bacterial strain backgrounds used in this study

Appendix B: Primers Table 6. Relevant primers and their sequences

Appendix C: P1asmids

Table 7. Plasmids used in this study pSNP958 P1asmid used as negative control in fluorescence Our collection analyses because it has the same resistance marker (Spc R ) and origin of replication as pSNP1248- derived plasmids but lacks the gfp reporter gene. pSNP1617 P1asmid used as a negative control because it has the Our collection same resistance marker (Tet R ) and origin of replication as pSNP1939 but lacks the xylitol ABC transporter. pSNP1903 Same as p 1902 except that the repressor protein This study ORF contains a 595-bp deletion.

Appendix D: M9 minimal medium recipe

The M9 minimal medium used in this study report consists of a medium salts solution supplemented with a carbon source (glucose or glycerol), Bl vitamin, magnesium, calcium and a source of iron. The medium salts solution is a composition of four crucial salts. We recommend preparing it as a 5x stock solution according to Table 8 below:

Table 8. 5xM9 minimal salts solution

Fill a beaker with -800 ml distilled H2O and add the specified amounts of the four salts while the solution is thoroughly mixed by a magnetic stir bar. Once the components have dissolved fill up with distilled H2O to 1000 ml. Sterilize by autoclaving the solution at 121 °C for 15 minutes. The 5x M9 salts solution can be stored at room temperature for up to a year.

All remaining stock solutions required to make the M9 medium salts solution are prepared routinely by weighing the appropriate amount of the component, dissolving it in distilled H2O under constant agitation and filter-sterilizing it (pore size = 0.2 pm). Once prepared, mix the stock solutions together according to the instructions from Table 9 below:

Table 9. Recipe forM9 minimal medium for E. coli growth Fill a beaker with -600 ml distilled H 2 O and add the specified volumes of stock solutions. The lx M9 minimal medium solution can be stored at room temperature for up to a month.

Appendix E: DNA- and amino acid sequence of the LacI family DNA-binding transcriptional regulator IZ184 04885

IZ184 04885 DNA sequence (5 ’→ 3’) (SEQ ID NO: 1) ttggcggaaccccggacacagaaagtcacactggaaaatgtggccgctattgcgaatgtc agtaagatcaccgcatcccgcgcattttcgcagccg gataaggttcacccggaaactctcaggcgtattctcgatgccgcagataaaatcggttat gtggtgaatgcggcggcacgcagcctgcgcgcgaa atcctcccgtaccatcggtattgtcagcccggacatgagcaacccgtttttcggcgggct ggccaagcgcatcaccctggaagcctataacgccgg gtatgacacgctgatgtttgactcctatgaatcacgtgaaaatgaagcgcgtatcattga taagctgattggttacaacgtggatgccatcattctgtcc gttgtctccgccgaaagggtgtatcgcccggcgtatatgaaacagctggaactgctcaat attccggtgatcctggttgaccgtgagctggatgcca aggcatgcagcggcgtttacatcgataatctcgactgcggattgcaggcagggcgctatc tgctgtcacaaaaagcggataacgtggtgattgtctc cggcccggaagattccaatgtggcccaggatcgcgtaaccggcatggtggccggtctgca cggccaggtcagcagcgtcaatgtgctgcatgcg gacttcctgatggatgaagcctttaaagtcaccgaccattacctgaaataccatcccgcg ccggattattttgtcggctgcaataaccagatctcactc gggattatcaaagcctgtatccgccacaatctgatcccgcaaaaagatgtctcgctgttc agcatcgatgaagtctctcatgccgatatttacggattca atttcccgtgcatttctcatgatttacaggaaattgcctggcaggccattaacatggcag tacggcgggcgacggatcgcagcgccccggcgagca aggtggttgtgcgcggtctcctcaaatcttaa

IZ 184_04885 amino acid sequence (SEQ ID NO: 2)

MAEPRTQKVTLENVAAIANVSKITASRAFSQPDKVHPETLRRILDAADKIGYVVNAA ARSLR AKSSRTIGIVSPDMSNPFFGGLAKRITLEAYNAGYDTLMFDSYESRENEARIIDKLIGYN VDAII LSVVSAERVYRPAYMKQLELLNIPVILVDRELDAKACSGVYIDNLDCGLQAGRYLLSQKA DN VVIVSGPEDSNVAQDRVTGMVAGLHGQVSSVNVLHADFLMDEAFKVTDHYLKYHPAPDYF VGCNNQISLGIIKACIRHNLIPQKDVSLFSIDEVSHADIYGFNFPCISHDLQEIAWQAIN MAVRR ATDRSAPASKVVVRGLLKS

Appendix F: Further Sequences

Promoter sequence (5 ’→ 3’) (SEQ ID NO: 3) cctgaatgatatcgttatcattatagccttgtcacagatagcaaagatgtgacccggaag accctttgccgaatactacatgagtagatcagactttattt aataaaagtttaaccatcatcacataattataataaatattgctctattatccgcatcat agtaatgatatcgatatcattttaatgaaaggaaatgatc Sequence comprising (in 5’ to 3’ direction) the sequence of the repressor, isomerase and promoter of

Morganella morganii strain ZJG812 (SEQ ID NO: 4)

This sequence can be used in the invention and provides the repressor and promoter. ttaagatttgaggagaccgcgcacaaccaccttgctcgccggggcgctgcgatccgtcgc ccgccgtactgccatgttaatggcctgccaggcaat ttcctgtaaatcatgagaaatgcacgggaaattgaatccgtaaatatcggcatgagagac ttcatcgatgctgaacagcgagacatctttttgcgggat cagattgtggcggatacaggctttgataatcccgagtgagatctggttattgcagccgac aaaataatccggcgcgggatggtatttcaggtaatggt cggtgactttaaaggcttcatccatcaggaagtccgcatgcagcacattgacgctgctga cctggccgtgcagaccggccaccatgccggttacgc gatcctgggccacattggaatcttccgggccggagacaatcaccacgttatccgcttttt gtgacagcagatagcgccctgcctgcaatccgcagtc gagattatcgatgtaaacgccgctgcatgccttggcatccagctcacggtcaaccaggat caccggaatattgagcagttccagctgtttcatatacg ccgggcgatacaccctttcggcggagacaacggacagaatgatggcatccacgttgtaac caatcagcttatcaatgatacgcgcttcattttcacgt gattcataggagtcaaacatcagcgtgtcatacccggcgttataggcttccagggtgatg cgcttggccagcccgccgaaaaacgggttgctcatgt ccgggctgacaataccgatggtacgggaggatttcgcgcgcaggctgcgtgccgccgcat tcaccacataaccgattttatctgcggcatcgaga atacgcctgagagtttccgggtgaaccttatccggctgcgaaaatgcgcgggatgcggtg atcttactgacattcgcaatagcggccacattttcca gtgtgactttctgtgtccggggttccgccaattttctctccttagcagcaacgtatcatt ttataccattataatatcatctaaatcgattcagtcacgtgaaa aacacgctttttattacttaaaccgacactaacaactttttaacactcgagtctttttat ccgcggatcggtaaaaatgtcgtattcatcggtgttttctgtgg agaactccgacaccaccgcgccctgcggtccggctttaaaccagtggcgggtatccggca tgatggtgtactgctctcccgggcgcagcagaata aagcggtcacaggtgtaccattcctcatccccctgcggcggacggcagaccggattgccc tgagcatcctgattcagtgtcagcgccggatcatcc acaaacagatacacctcaccccagcggcagcggaaagtttcctgcttacccggccggtct ttgtacggcggatgacggtgctccggacaggtctg gtcagcaaagagaaccagctctttggcgcagtacagcgggctgttgcagtacgtgagcag ttgcaggccagagcggggataatccggcagatta aagtgagcaatttcaatatgctgctgttccgcttctgtcagaatgatatgtgcctgttcc agaaccgccagtgattccgcgatatacggctgtttattcat ctgcctctctcctgaatgatatcgttatcattatagccttgtcacagatagcaaagatgt gacccggaagaccctttgccgaatactacatgagtagatc agactttatttaataaaagtttaaccatcatcacataattataataaatattgctctatt atccgcatcatagtaatgatatcgatatcattttaatgaaaggaa atgatc

TABLE 1: Example Cell Genera, Species & Strains

Each carrier cell may be a cell of a genus or species disclosed in this table and/or target cell may be a cell of a genus or species disclosed in this table; the genera of the carrier and target cells may be the same of different; the species of the carrier and target cells may be the same of different.

Bacillus pumilus

Acidovorax avenae subsp. cattleyae

Acidovorax avenae subsp. Brenneria

Acidovorax konjaci Brenneria alni

Acidovorax valerianellae Brenneria nigrifluens

Brenneria quercina

Agrobacterium Brenneria rubrifaciens

Agrobacterium larrymoorei Brenneria salicis

Agrobacterium radiobacter

Agrobacterium rhizogenes Burkholderia

Agrobacterium rubi

Agrobacterium tumefaciens Burkholderia andropogonis

Agrobacterium vitis Burkholderia caryophylli

Arthrobacter Burkholderia cepacia

Arthrobacter ilicis Burkholderia gladioli

Bacillus Burkholderia gladioli pv. agaricicola

Bacillus megaterium Burkholderia gladioli pv. alliicola

Bacillus megaterium pv. cere alls Burkholderia gladioli pv. gladioli

Burkholderia glumae Corynebacterium fascians

Burkholderia Corynebacterium flaccumfaciens

Corynebacterium flaccumfaciens pv. betae

Corynebacterium flaccumfaciens pv . flaccumfaciens

Clavibacter Corynebacterium flaccumfaciens pv. oortii

Clavibacter michiganensis Corynebacterium flaccumfaciens pv. poinsettiae

Clavibacter michiganensis subsp. Corynebacterium flaccumfaciens subsp.

Clavibacter michiganensis subsp. michiganensis Corynebacterium flaccumfaciens subsp. flaccumfaciens

Clavibacter michiganensis subsp. nebraskensis Corynebacterium flaccumfaciens subsp. oortii

Clavibacter michiganensis subsp. sepedonicus Corynebacterium flaccumfaciens subsp. poinsettiae

Clavibacter michiganensis subsp. tessellarius Corynebacterium ilicis

Clavibacter rathayi Corynebacterium insidiosum

Clavibacter toxicus Corynebacterium iranicum

Clavibacter tritici Corynebacterium michiganense

Clavibacter xyli Corynebacterium michiganensis pv. insidiosus

Clavibacter xyli subsp. cynodontis Corynebacterium michiganensis pv. iranicum

Clavibacter xyli subsp. xyli Corynebacterium michiganense pv. nebraskense

Corynebacterium michiganense pv. rathayi

Clostridium Corynebacterium michiganense pv. sepedonicum

Clostridium puniceum Corynebacterium michiganense pv. tritici

Corynebacterium Corynebacterium michiganense subsp. insidiosum

Corynebacterium betae Corynebacterium michiganense subsp.

Corynebacterium beticola Corynebacterium michiganense subsp. nebraskense

Corynebacterium michiganense subsp. sepedonicum Dickeya dianthicola

Corynebacterium michiganense subsp. tessellarius Dickeya dieffenbachiae

Corynebacterium oortii Dickeya paradisiaca

Dickeya zeae

Corynebacterium

Corynebacterium rathayi Enterobacter

Corynebacterium sepedonicum Enterobacter agglomerans

Enterobacter cancerogenus

Corynebacterium tritici Enterobacter cloacae

Curtobacterium Enterobacter cloacae subsp. dis solvens

Curtobacterium flaccumfaciens Enterobacter nimipressuralis

Enterobacter pyrinus

Curtobacterium flaccumfaciens pv.

Curtobacterium flaccumfaciens pv. flaccumfaciens Erwinia

Curtobacterium flaccumfaciens pv. ilicis

Curtobacterium flaccumfaciens pv. oortii Erwinia alni

Curtobacterium flaccumfaciens pv. poinsettiae Erwinia amylovora .

Erwinia amylovora pv.pyri

Dickeya Erwinia ananatis corrig.

Dickeya chrysanthemi Erwinia ananatis pv. ananatis

Dickeya chrysanthemi pv. chrysanthemi Erwinia ananas pv. uredovora

Dickeya chrysanthemi pv.parthenii Erwinia cacticida

Dickeya dadantii Erwinia cancerogena

Erwinia carnegieana Erwinia nimipressuralis

Erwinia carotovora Erwinia papayae

Erwinia carotovora pv. atroseptica Erwinia proteamaculans

Erwinia carotovora pv. carotovora Erwinia persicina

Erwinia carotovora subsp. atroseptica Enterobacter pyrinus

Erwinia carotovora subsp. carotovora Erwinia psidii

Erwinia carotovora subsp. betavasculorum Erwinia pyrifoliae

Erwinia carotovora subsp. odorifera Erwinia rhapontici

Erwinia carotovora subsp. wasabiae Erwinia rubrifaciens

Erwinia chrysanthemi Erwinia salicis

Erwinia chrysanthemi pv. chrysanthemi Erwinia stewartii

Erwinia chrysanthemi pv. Erwinia tracheiphila

Erwinia chrysanthemi pv. dieffenbachiae Erwinia uredovora

Erwinia chrysanthemi pv. paradisiaca Ewingella

Erwinia chrysanthemi pv. parthenii Ewingella americana

Erwinia chrysanthemi pv. zeae Gluconobacter Asai

Erwinia cypripedii Gluconobacter oxydans

Erwinia dissolvens Herbaspirillum

Erwinia herbicola Herbaspirillum rubrisubalbicans

Erwinia herbicola f. sp. Janthinobacterium

Erwinia herbicola pv. millettiae Janthinobacterium agaricidamnosum

Erwinia mallotivora Leifsonia

Erwinia nigrifluens Leifsonia cynodontis

Leifsonia xyli Pectobacterium

Leifs onia xyli subsp. cynodontis Pectobacterium carotovorum

Leifsonia xyli subsp. xyli

Pectobacterium carotovorum subsp. atrosepticum

Pectobacterium carotovorum subsp. betavasculorum

Nocardia Pectobacterium carotovorum subsp. brasiliensis

Pectobacterium carotovorum subsp. carotovorum

Nocardia vaccinii Pectobacterium carotovorum subsp. odoriferum

Pectobacterium carotovorum subsp. wasabiae

Pantoea Pectobacterium chrysanthemi

Pantoea agglomerans Pectobacterium chrysanthemi pv. chrysanthemi

Pantoea agglomerans pv. gypsophilae Pectobacterium chrysanthemi pv. dianthicola

Pantoea agglomerans pv. millettiae Pectobacterium chrysanthemi pv. dieffenbachiae

Pantoea ananatis Pectobacterium chrysanthemi pv. parthenii

Pantoea ananatis pv. ananatis Pectobacterium chrysanthemi pv. zeae

Pantoea ananatis pv. uredovora Pectobacterium cypripedii

Pantoea stewartii Pectobacterium rhapontici

Pantoea stewartii subsp. indologenes Pectobacterium wasabiae

Pantoea stewartii subsp. stewartii

Pectobacterium Pseudomonas

Pectobacterium

Pectobacterium Pseudomonas agarici

Pectobacterium cacticida corrig Pseudomonas amygdali

Pseudomonas andropogonis pv. andropogonis Pseudomonas fuscovaginae

Pseudomonas andropogonis pv. sojae Pseudomonas gingeri

Pseudomonas andropogonis pv. stizolobii Pseudomonas gladioli

Pseudomonas asplenii Pseudomonas gladioli pv. agaricicola

Pseudomonas avellanae Pseudomonas gladioli pv. alliicola

Pseudomonas avenae Pseudomonas gladioli pv. gladioli

Pseudomonas avenae subsp. avenae Pseudomonas glumae

Pseudomonas avenae subsp. citrulli Pseudomonas hibiscicola

Pseudomonas avenae subsp. konjaci Pseudomonas marginalis

Pseudomonas beteli corrig. Pseudomonas marginalis pv. alfalfae

Pseudomonas cannabina Pseudomonas marginalis pv. marginalis

Pseudomonas caricapapayae Pseudomonas marginalis pv. pastinacae

Pseudomonas caryophylli Pseudomonas mediterranea

Pseudomonas cattleyae Pseudomonas meliae

Pseudomonas cepacia Pseudomonas palleroniana

Pseudomonas cichorii Pseudomonas plantarii

Pseudomonas cissicola Pseudomonas pomi

Pseudomonas coronafaciens Pseudomonas pseudoalcaligenes subsp. citrulli

Pseudomonas corrugata Pseudomonas pseudoalcaligenes subsp. konjaci

Pseudomonas costantinii Pseudomonas rubrilineans

Pseudomonas dodoneae Pseudomonas rubrisubalbicans

Pseudomonas ficuserectae Pseudomonas salomonii

Pseudomonas flectens Pseudomonas savastanoi

Pseudomonas savastanoi ^N.fraxini Pseudomonas syringae pv. coriandricola

Pseudomonas savastanoi pv. glycinea Pseudomonas syringae pv. coronafaciens

Pseudomonas savastanoi pv. nerii Pseudomonas syringae pv. coryli

Pseudomonas savastanoi pv. phaseolicola Pseudomonas syringae pv. cunninghamiae

Pseudomonas savastanoi pv. retacarpa Pseudomonas syringae pv. daphniphylli

Pseudomonas savastanoi pv. savastanoi Pseudomonas syringae pv. delphinii

Pseudomonas syringae Pseudomonas syringae pv. dendropanacis

Pseudomonas syringae pv. aceris Pseudomonas syringae pv. dysoxyli

Pseudomonas syringae pv. actinidiae Pseudomonas syringae pv. eriobotryae

Pseudomonas syringae pv. aesculi Pseudomonas syringae pv. garcae

Pseudomonas syringae pv. alisalensis Pseudomonas syringae pv. glycinea

Pseudomonas syringae pv. antirrhini Pseudomonas syringae pv. helianthi

Pseudomonas syringae pv. apii Pseudomonas syringae pv.

Pseudomonas syringae pv. aptata Pseudomonas syringae pv.

Pseudomonas syringae pv. Pseudomonas syringae pv.

Pseudomonas syringae pv. atropurpurea Pseudomonas syringae pv. lapsa

Pseudomonas syringae pv. avellanae Pseudomonas syringae pv. maculicola

Pseudomonas syringae pv. avii Pseudomonas syringae pv.

Pseudomonas syringae pv. berberidis Pseudomonas syringae pv. mori

Pseudomonas syringae pv. broussonetiae Pseudomonas syringae pv. morsprunorum .

Pseudomonas syringae pv. castaneae Pseudomonas syringae pv. myricae

Pseudomonas syringae pv. cerasicola Pseudomonas syringae pv.

Pseudomonas syringae pv. ciccaronei Pseudomonas syringae pv. papulans

Pseudomonas syringae pv. passiflorae

Pseudomonas syringae pv. Ralstonia

Pseudomonas syringae pv. philadelphi Ralstonia solanacearum

Pseudomonas syringae pv. photiniae Ralstonia syzygii

Pseudomonas syringae pv. pisi Rathayibacter

Pseudomonas syringae pv. porri Rathayibacter iranicus

Pseudomonas syringae pv. primulae Rathayibacter rathayi

Pseudomonas syringae pv. rhaphiolepidis Rathayibacter

Pseudomonas syringae pv. ribicola Rathayibacter tritici

Pseudomonas syringae pv. sesami Rhizobacter

Pseudomonas syringae pv. solidagae Rhizobacter dauci corrig.

Pseudomonas syringae pv. spinaceae

Pseudomonas syringae pv. syringae Rhizobium

Pseudomonas syringae pv. tagetis Rhizobium larrymoorei

Pseudomonas syringae pv. theae Rhizobium radiobacter

Pseudomonas syringae pv. tomato Rhizobium rhizogenes

Pseudomonas syringae pv. ulmi Rhizobium rubi

Pseudomonas syringae pv. viburni Rhizobium vitis

Pseudomonas syringae pv.

Pseudomonas W” Rhodococcus

Pseudomonas tolaasii Rhodococcus fascians

Pseudomonas tremae

Pseudomonas viridiflava Samsonia

Samsonia erythrinae Streptomyces intermedins

Streptomyces ipomoeae

Serratia Streptomyces luridiscabiei

Serratia marcescens Streptomyces niveiscabiei

Serratia proteamaculans Streptomyces puniciscabiei

Streptomyces reticuliscabei

Sphingomonas Streptomyces scabiei corrig.

Sphingomonas melonis Buonaurio Streptomyces setonii

Sphingomonas suberifaciens Streptomyces steliiscabiei

Streptomyces turgidiscabies

Spiroplasma Streptomyces wedmorensis

Spiroplasma citri

Xanthomonas

Spiroplasma kunkelii Xanthomonas albilineans

Spiroplasma phoeniceum Xanthomonas alfalfae

Xanthomonas alfalfae subsp. alfalfae

Streptomyces Xanthomonas alfalfae subsp. citrumelonis

Streptomyces acidiscabies Xanthomonas arboricola

Streptomyces albidoflavus Xanthomonas axonopodis

Streptomyces candidus Xanthomonas bromi

Streptomyces caviscabies Xanthomonas campestris

Streptomyces collinus Xanthomonas cassavae

Streptomyces europaei scabiei Xanthomonas citri

Xanthomonas cucurbitae 'Candidates Liberibacter’

Xanthomonas euvesicatoria ‘'Candidates Liberibacter asiaticus’

Xanthomonas fragariae ‘Candidates Phlomobacter’

Xanthomonas fuscans ‘Candidates Phlomobacter fragariae’

Xanthomonas fuscans ‘Candidates Phytoplasma’

Xanthomonas gardneri

Xanthomonas hortorum

Xanthomonas hortorum

Xanthomonas hyacinthi

Xanthomonas oryzae

Xanthomonas populi

Xanthomonas sacchari

Xanthomonas theicola

Xanthomonas translucens

Xanthomonas vasicola

Xylella

Xylella fastidiosa

Xylophilus

Xylophilus ampelinus

‘Candidates' P1ant Pathogenic Bacteria

TABLE 2: Further Example Bacteria

Optionally, the carrier cells are selected from this Table and/or the target cells are selected from this Table (eg, wherein the carrier and target cells are of a different species; or of the same species but are a different strain or vice versa).

Abiotrophia Acidocella Actinomyces Alkalilimnicola Aquaspirillum

Abiotrophia defectiva Acidocella aminolytica Actinomyces bovis Alkalilimnicola ehrlichii Aquaspirillum polymorphum

Acidocella facilis Actinomyces denticolens Aquaspirillum

Acaricomes Actinomyces europaeus Alkaliphilus putridiconchylium

Acaricomes phytoseiuli Acidomonas Actinomyces georgiae Alkaliphilus oremlandii Aquaspirillum serpens

Acidomonas methanolica Actinomyces gerencseriae Alkaliphilus transvaalensis

Acetitomaculum

Actinomyces Aquimarina

Acetitomaculum ruminis Acidothermus Allochrom atium hordeovulneris Aquimarina latercula

Acidothermus cellulolyticus Allochromatium vinosum

Acetivibrio Actinomyces howellii

Arcanobacterium

Acetivibrio cellulolyticus Acidovorax Actinomyces hyovaginalis Alloiococcus Arcanobacterium

Acetivibrio ethanolgignens Acidovorax anthurii Actinomyces israelii Alloiococcus otitis haemolyticum

Acetivibrio multivorans Acidovorax caeni Actinomyces johnsonii

Arcanobacterium pyogenes

Acidovorax cattleyae Actinomyces meyeri Allokutzneria

Acetoanaerobium Acidovorax citrulli Actinomyces naeslundii Allokutzneria albata Archangium

Acetoanaerobium noterae Acidovorax defluvii Actinomyces neuii Archangium gephyra

Altereryth rob acter

Acidovorax delafieldii Actinomyces odontolyticus

Acetob acter Altererythrobacter

Acidovorax facilis Actinomyces oris Arcobacter

Acetobacter aceti ishigakiensis

Acidovorax konjaci Actinomyces radingae Arcobacter butzleri

Acetobacter cerevisiae

Acetobacter cibinongensis Acidovorax temperans Actinomyces slackii Arcobacter cryaerophilus

Acetobacter estunensis Acidovorax valerianellae Actinomyces turicensis Altermonas Arcobacter halophilus

Acetobacter fabarum Actinomyces viscosus Altermonas haloplanktis Arcobacter nitrofigilis

Acetobacter ghanensis Acinetobacter Altermonas macleodii Arcobacter skirrowii

Acetobacter indonesiensis Acinetobacter baumannii Actinoplanes

Acinetobacter baylyi Actinoplanes auranticolor Alysiella

Acetobacter lovaniensis Arhodomonas

Acinetobacter bo Alysiella crassa

Acetobacter malorum uvetii Actinoplanes brasiliensis Arhodomonas aquaeolei

Acinetobacter calcoaceticus Actinoplanes c Alysiella filiformis

Acetobacter nitrogenifigens onsettensis

Acinetobacter g Arsenophonus

Acetobacter oeni erneri Actinoplanes deccanensis

Aminobacter Arsenoph etobacter orientalis Acinetobacter haemol onus nasoniae

Ac yticus Actinoplanes derwentensis

Aminobacter aganoensis

Acetobacter orleanensis Acinetobacter johnsonii Actinoplanes digitatis

Aminobacter aminovorans

Acetobacter pasteurianus Acinetobacter junii Actinoplanes durhamensis

Aminobacter niigataensis

Acetobacter pornorurn Acinetobacter Iwoffi Actinoplanes ferrugineus Arthrobacter

Acetobacter senegalensis Acinetobacter parvus Actinoplanes globisporus Aminobacterium Arthrobacter agilis

Acetobacter xylinus Acinetobacter radioresistens Actinoplanes humidus Aminobacterium mobile Arthrobacter albus

Acinetobacter schindleri Actinoplanes italicus Arthrobacter aurescens

Acetobacterium Acinetobacter soli Actinoplanes liguriensis Aminomonas Arthrobacter

Acetobacterium bakii Acinetobacter tandoii Actinoplanes lobatus Aminomonas paucivorans chlor ophenolicus

Acetobacterium carbinolicum Acinetobacter tjernbergiae Actinoplanes missouriensis Arthrobacter citreus

Ammoniphilus

Acetobacterium dehalogenans Acinetobacter towneri Actinoplanes palleronii Arthrobacter crystallopoietes

Ammoniphilus oxalaticus

Acetobacterium fimetarium Acinetobacter ursingii Actinoplanes philippinensis Arthrobacter cumminsii

Ammoniphilus oxalivorans

Acetobacterium malicum Acinetobacter venetianus Actinoplanes rectilineatus Arthrobacter globiformis

Acetobacterium paludosum Actinoplanes regularis

Acetobacterium tundrae Actinoplanes Arthrobacter

Acetobacterium wieringae Acrocarpospora teichomyceticus Amphibacillus histidinolovorans

Acetobacterium woodii Acrocarpospora corrugata Actinoplanes utahensis Amphibacillus xylanus Arthrobacter ilicis

Acrocarpospora Arthrobacter luteus

Acetofilamentum macrocephala Actinopolyspora Amphritea

Arthrobacter methylotrophus

Acetofilamentum rigidum Acrocarpospora Actinopolyspora halophila Amphritea balenae

Arthrobacter mysorens pleiomorpha Actinopolyspora Amphritea japonica

Arthrobacter nicotianae

Acetohalobium mortivallis Arthrobacter nicotin lobium arabaticum Actibacter Amycol ovorans

Acetoha atopsis

Amycolato Arthrobacter oxydans

Actibacter sediminis psis alba

Actinosynnema

Acetomicrobium Amycolato Arthrobacter pascens

Actinosynnema mirum psis albidoflavus

Acetomicrobium faecale Actinoalloteichus Amycolatopsis azurea Arthrobacter

Acetomicrobium flavidum Actinoallot eichus Actinotalea Amycolatopsis coloradensis phenanthrenivorans cyanogriseus Actinotalea fermentans Amycolatopsis lurida Arthrobacter

Acetonema Actinoalloteichus Amycolatopsis mediterranei poly chromogenes

Acetonema longum hymeniacidonis Aerococcus Amycolatopsis rifamycinica Atrhrobacter protophormiae

Actinoalloteichus spitiensis Aerococcus sanguinicola Amycolatopsis rubida Arthrobacter

Acetothermus Aerococcus urinae Amycolatopsis sulphurea psychrolactophilus

Acetothermus paucivorans

Actinobaccillus Aerococcus urinaeequi Amycolatopsis tolypomycina Arthrobacter ramosus

Actinobacillus capsulatus Aerococcus urinaehominis Arthrobacter sulfonivorans

Acholeplasma Actinobacillus delphinicola Aerococcus viridans Anabaena Arthrobacter sulfureus

Acholeplasma axanthum Actinobacillus hominis Anabaena cylindrica Arthrobacter uratoxydans

Acholeplasma brassicae Actinobacillus indolicus Aeromicrobium

Arthrobacter ureafaciens

Acholeplasma cavigenitalium Aeromicrobium erythreum

Acholeplasma equifetale Actinobacillus lignieresii Anabaena flos-aquae Arthrobacter viscosus

Acholeplasma granularum Actinobacillus minor Aeromonas Anabaena variabilis Arthrobacter woluwensis

Acholeplasma hippikon Actinobacillus muris Aeromonas

Acholeplasma laidlawii Actinobacillus allosaccharophila Anaeroarcus Asaia

Acholeplasma modicum pleuropneumoniae Aeromonas bestiarum Anaeroarcus burkinensis Asaia bogorensis

Acholeplasma morum Actinobacillus porcinus Aeromonas caviae

Aeromonas enchel Anaerobaculum Asanoa

Acholeplasma multilocale Actinobacillus rossii eia

Anaerobaculum mobile Asanoa ferrugi choleplasma oculi Actinobacillus scotiae Aer nea

A omonas

Acholeplasma palmae Actinobacillus seminis enteropelogenes

Anaerobiospirillum Asticcacaulis

Acholeplasma parvum Actinobacillus succinogenes Aeromonas eucrenophila

Anaerobiospirillum Asticcacaulis biprosthecium

Acholeplasma pleciae Actinobaccillus suis Aeromonas ichthiosmia succiniciproducens Asticcacaulis excentricus

Acholeplasma vituli Actinobacillus ureae Aeromonas jandaei

Anaerobiospirillum thomasii

Aeromonas media Atopobacter

Achromobacter Actinobaculum Aeromonas popoffii Anaerococcus Atopobacter phocae

Achromohacter denitrificans Actinobaculum massiliense Aeromonas sobria Anaerococcus hydrogenalis

Achromobacter insolitus Actinobaculum schaalii Aeromonas veronii Anaerococcus lactolyticus Atopobium

Achromobacter piechaudii Actinobaculum suis Anaerococcus prevotii Atopobium fossor

Achromobacter ruhlandii Actinomyces urinale Agrobacterium

Anaerococcus tetradius Atopobium minutum

Achromobacter spanius Agrobacterium

Anaerococcus vaginalis Atopobium parvulum

Actinocatenispora gelatinovorum Atopobium rimae

Acidaminobacter Actinocat enispor a rupis Anaerofustis Atopobium vaginae

Acidaminobacter Actinocatenispora Anaerofustis stercorihominis hydrogenoformans

thailandica

Acidaminococcus Actinocatenispora sera Agrococcus Anaeromusa Aureobacterium

Acidaminococcus fermentans Agrococcus citreus Anaeromusa acidaminophila Aureobacterium barkeri

Acidaminococcus intestini Actinocorallia Agrococcus jenensis

Actinocorallia aurantiaca Anaeromyxobacter Aurobacterium

Acidicaldus Actinocorallia aurea Agromonas Anaeromyxobacter Aurobacterium liquefaciens

Acidicaldus organivorans Actinocorallia cavernae Agromonas oligotrophica dehalogenans

Avibacterium

Actinocorallia glomerata

Acidimicrobium Agromyces Anaerorhabdus Avibacterium avium

Actinocorallia herbida

Acidimicrobium ferrooxidans Agromyces fucosus Anaerorhabdus furcosa Avibacterium gallinarum

Actinocorallia libanotica

Agromyces hippuratus Avibacterium paragallinarum

Acidiphilium Actinocorallia longicatena

Agromyces luteolus Anaerosinus Avibacterium volantium

Acidiphilium acidophilum Agromyces mediolanus Anaerosinus glycerini

Actinomadura

Acidiphilium angustum Agro Azoarcus

Actinomadura alba myces ramosus

Acidiphilium cryptum Anaerovirgula tinomadura atramentaria Agromyces r Azoarcus indigens

Ac hizospherae

Acidiphilium multivorum Anaerovirgula multivorans Azoarcus tolulyticus

Actinomadura

Acidiphilium organovorum Akkermansia Azoarcus toluvorans banglade shensi s Ancalomicrobium

Acidiphilium rubrum Akkermansia muciniphila

Actinomadura catellatispora Ancalomicrobium adetum Azohydromonas

Acidisoma Actinomadura chibensis Albidiferax Azohydromonas australica

Ancylobacter

Acidisoma sibiricum Actinomadura chokoriensis Albidiferax ferrireducens Azohydromonas lata

Ancylobacter aquaticus

Acidisoma tundrae Actinomadura citrea

Actinomadura coerulea Albidovulum Azomonas

Actinomadura echinospora Albidovulum inexpectatum Azomonas agilis

Actinomadura fibrosa Azomonas insignis

Acidisphaera Actinomadura formosensis Alcaligenes Aneurinibacillus Azomonas macrocytogenes

Acidisphaera rubrifaciens Actinomadura hibisca Alcaligenes denitrificans Aneurinibacillus

Actinomadura kijaniata Alcaligenes faecalis aneurinilyticus Azorhizobium

Acidithiobacillus

Actinomadura latina Aneurinibacillus migulanus Azorhizobium caulinodans

Acidithiobacillus albertensis Alcanivorax

Actinomadura livida Aneurinibacillus

Acidithiobacillus caldus Alcanivorax borkumensis thermoaeroph Azorhizophilus

Actinomadura ilus

Acidithiobacillus ferrooxidans Alcanivorax jadensis Azorhizophilus paspali luteofluorescens

Acidithiobacillus thiooxidans Angiococcus

Actinomadura macra Algicola Angiococcus disciformis Azospirillum

Acidobacterium Actinomadura madurae Algicola bacteriolytica Azospirillum brasilense

Acidobacterium capsulatum Actinomadura oligospora Angulomicrobium Azospirillum halopraeferens

Actinomadura pelletieri Alicyclobacillus Angulomicrobium tetraedrale Azospirillum irakense

Actinomadura rubrobrunea Alicyclobacillus

Actinomadura rugatobispora disulfidooxidans Anoxybacillus Azotobacter

Actinomadura umbrina Alicyclobacillus Anoxybacillus pushchinoensis Azotobacter beijerinckii

Actinomadura sendaiensis Azotobacter chroococcum verrucosospora Alicyclobacillus vulcanalis Aquabacterium Azotobacter nigricans

Aquabacterium commune

Actinomadura vinacea Azotobacter salinestris

Alishewanella Aquabacterium parvum

Actinomadura viridilutea Azotobacter vinelandii

Alishewanella fetalis

Actinomadura viridis

Actinomadura yumaensis

Alkalibacillus

Alkalibacillus haloalkaliphilus

Bacillus Bacteroides Bibersteinia Borrelia Brevinema

[see below] Bacteroides caccae Bibersteinia trehalosi Borrelia afzelii Brevinema andersonii

Bacteroides coagulans Borrelia americana

Bacteroides eggerthii Bifidobacterium Borrelia burgdorferi Brevundimonas

Bacteroides fragilis Bifidobacterium adolescentis Borrelia carolinensis Brevundimonas alba

Bacteriovorax

Bacteroides galacturonicus Bifidobacterium angulatum Borrelia coriaceae Brevundimonas aurantiaca

Bacteriovorax stolpii

Bacteroides helcogenes Bifidobacterium animalis Borrelia garinii Brevundimonas diminuta

Bacteroides ovatus Bifidobacterium asteroides Borrelia japonica Brevundimonas intermedia

Bacteroides pectinophilus Bifidobacterium bifidum Brevundimonas subvibrioides

Bacteroides pyogenes Bifidobacterium bourn Bosea Brevundimonas vancanneytii

Bacteroides salyersiae Bifidobacterium breve Bosea minatitlanensis Brevundimonas variabilis

Bacteroides stercoris Bifidobacterium catenulatum Bosea thiooxidans Brevundimonas vesicularis

Bacteroides suis Bifidobacterium choerinum

Bifidobacterium coryneform Brachybacterium Brochothrix

Bacteroides tectus e eroides thetaiotaomicron Bifido Brachybacterium Brochothrix campestris

Bact bacterium cuniculi

Bifidobacterium dentiu alimentarium Brochothrix thermosphacta

Bacteroides uniformis m

Bifidobacterium gallicum Brachybacterium faecium

Brachybacterium

Bacteroides ureolyticus Bifidobacterium gallinarum paraconglomeratum

Bacteroides vulgatus Bifidobacterium indicum Brachybacterium rhamnosum Brucella

Bifidobacterium longum Brachybacterium Brucella canis

Balnearium Bifidobacterium tyrofermentans Brucella neotomae

Balnearium lithotrophicum magnumBifidobacterium

Bryobacter merycicum Brachyspira

Balneatrix B Bryobacter aggregatus

Bifidobacterium minimum rachyspira alvinipulli

Balneatrix alpica

Bifidobacterium Brachyspira hyodysenteriae

Burkholderia

Brachyspira innocens

Balneola pseudocatenulatum Burkholderia ambifaria la vulgaris Bifidobact Brachyspira murdochii

Balneo erium Burkholderia andropogonis pseudoIongum Brachyspira pilosicoli

Burkholderia anthina

Barnesiella Bifidobacterium pullorum Burkholderia caledonica

Barnesiella viscericola Bifidobacterium ruminantium Burkholderia caryophylli

Bifidobacterium saeculare Bradyrhizobium Burkholderia cenocepacia

Bartonella

Bifidobacterium subtile Bradyrhizobium canariense Burkholderia cepacia

Bartonella alsatica

Bifidobacterium Bradyrhizobium elkanii Burkholderia cocovenenans

Bartonella bacilliformis thermophilum Bradyrhizobium japonicum Burkholderia dolosa

Bartonella clarridgeiae

Bradyrhizobium liaoningense Burkholderia fungorum

Bartonella doshiae Bilophila

Burkholderia glathei

Bartonella elizabethae Bi lophila wadsworthia Brenneria Burkholderia glumae

Bartonella grahamii Brenneria alni Burkholderia graminis

Bartonella henselae Biostraticola

Brenneria nigrifluens

Biostraticola tofi Burkholderia kururiensis

Bartonella rochalimae Brenneria quercina Burkholderia multivorans

Bartonella vinsonii Bizionia Brenneria quercina Burkholderia phenazinium

Bizionia argentinensis Brenneria salicis Burkholderia plantarii

Bavariicoccus Burkholderia pyrrocinia

Bavariicoccus seileri Blastobacter Brevibacillus Burkholderia silvatlantica

Blastobacter capsulatus Brevibacillus agri Burkholderia stabilis

Bdellovibrio Blastobacter denitrificans Brevibacillus borstelensis Burkholderia thailandensis

Bdellovibrio bacteriovorus Brevibacillus brevis s Blastococ Burkholderia tropica

Bdellovibrio exovoru cus

Brevibacillus centrosporus

Blastococcus aggregatus Burkholderia unamae

Brevibacillus choshinensis

Beggiatoa Blastococcus saxobsidens Burkholderia vietnamiensis

Brevibacillus invocatus

Beggiatoa alba

Blastochloris Brevibacillus laterosporus Buttiauxella

Beijerinckia Blastochloris viridis Brevibacillus parabrevis Buttiauxella agrestis

Beijerinckia derxii Brevibacillus reuszeri Buttiauxella brennerae

Beijerinckia fluminensis Blastomonas Buttiauxella ferragutiae

Bl Brevibacterium

Beijerinckia indica astomonas natatoria Buttiauxella gaviniae

Brevibacterium abidum

Beijerinckia mobilis Buttiauxella izardii

Blastopirellula Brevibacterium album Buttiauxella noackiae

Belliella Blastopirellula marina Brevibacterium aurantiacum Buttiauxella warmboldiae

Belliella baltica Brevibacterium celere

Blautia Brevibacterium epidermidis Butyrivibrio

Bellilinea Blautia coccoides Brevibacterium Butyrivibrio fibrisolvens

Bellilinea caldifistulae Blautia hansenii frigoriTolerans

Blautia producta Brevibacterium halotolerans Butyrivibrio hungatei

Belnapia Blautia wexlerae Brevibacterium iodinum Butyrivibrio proteoclasticus

Belnapia moabensis Brevibacterium linens

Bogoriella Brevibacterium lyticum

Bergeriella Bogoriella caseilytica Brevibacterium mcbrellneri

Bergeriella denitrificans

Brevibacterium otitidis

Bordetella

Beutenbergia Brevibacterium oxydans

Bordetella avium

Beutenbergia cavernae Brevibacterium paucivorans

Bordetella bronchiseptica

Brevibacterium stationis

Bordetella hinzii

Bordetella holmesii

Bordetella parapertussis

Bordetella pertussis

Bordetella petrii

Bordetella trematum

Bacillus

B. acidiceler B. aminovorans B. glucanolyticus B. taeanensis B. lautus

B. acidicola B. amylolyticus B. gordonae B. tequilensis B. lehensis

B. acidiproducens B. andreesenii B. gottheilii B. thermantarcticus B. lentimorbus

B. acidocaldarius B. aneurinilyticus B. graminis B. thermoaerophilus B. lentus

B. acidoterrestris B. anthracis B. halmapalus B. thermoamylovorans B. licheniformis

B. aeolius B. aquimaris B. haloalkaliphilus B. thermocatenulatus B. ligniniphilus

B. aerius B. arenosi B. halochares B. thermocloacae B. litoralis

B. aerophilus B. arseniciselenatis B. halodenitrificans B. thermocopriae B. locisalis

B. agaradhaerens B. arsenicus B. halodurans B. thermodenitrificans B. luciferensis

B. agri B. aurantiacus B. halophilus B. thermoglucosidasius B. luteolus

B. aidingensis B. arvi B. halosaccharovorans B. thermolactis B. luteus

B. akibai B. aryabhattai B. hemicellulosilyticus B. thermoleovorans B. macauensis

B. alcalophilus B. asahii B. hemicentroti B. thermophilus B. macerans

B. algicola B. atrophaeus B. herbersteinensis B. thermoruber B. macquariensis

B. alginolyticus B. axarquiensis B. horikoshii B. thermosphaericus B. macyae

B. alkalidiazotrophicus B. azotofixans B. horneckiae B. thiaminolyticus B. malacitensis

B. alkalinitrilicus B. azotoformans B. horti B. thioparans B. mannanilyticus

B. alkali sediminis B. badius B. huizhouensis B. thuringiensis B. marisflavi

B. alkalitelluris B. barbaricus B. humi B. tianshenii B. marismortui

B. altitudinis B. bataviensis B. hwajinpoensis B. trypoxylicola B. marmarensis

B. alveayuensis B. beijingensis B. idriensis B. tusciae B. massiliensis

B. alvei B. benzoevorans B. indicus B. validus B. megaterium

B. amyloliquefaciens B. beringensis B. infantis B. vallismortis B. mesonae

B. berkeleyi B. infernus B. vedderi B. methanolicus

• B. B. beveridgei B. insolitus B. velezensis B. methylotrophicus a. subsp. amyloliquefaciens B. bogoriensis B. invictae B. vietnamensis B. migulanus

• B. a. subsp. plantarum B. boroniphilus B. iranensis B. vireti B. mojavensis

B. borstelensis B. isabeliae B. vulcani B. mucilaginosus

B. brevis Migula B. isronensis B. wakoensis B. muralis

B. dipsosauri B. butanolivorans B. jeotgali B. weihenstephanensis B. murimardni

B. drentensis B. canaveralius B. kaustophilus B. xiamenensis B. mycoides

B. edaphicus B. carboniphilus B. kobensis B. xiaoxiensis B. naganoensis

B. ehimensis B. cecembensis B. kochii B. zhanjiangensis B. nanhaiensis

B. eiseniae B. cellulosilyticus B. kokeshiiformis B. nanhaii sediminis

B. enclensis B. centrosporus B. koreensis B. peoriae B. nealsonii

B. endophyticus B. cereus B. korlensis B. persepolensis B. neidei

B. endoradicis B. chagannorensis B. kribbensis B. persicus B. neizhouensis

B. farraginis B. chitinolyticus B. krulwi chiae B. pervagus B. niabensis

B. fastidiosus B. chondroitinus B. laevolacdcus B. plakortidis B. niacini

B. fengqiuensis B. choshinensis B. larvae B. pocheonensis B. novalis

B. firmus B. chungangensis B. laterosporus B. polygoni B. oceanisediminis

B. flexus B. cibi B. salexigens B. polymyxa B. odysseyi

B. foraminis B. circulans B. saliphilus B. popilliae B. okhensis

B. fordii B. clarkii B. schlegelii B. pseudalcalophilus B. okuhidensis

B. formosus B. clausii B. sediminis B. pseudofirmus B. oleronius

B. fords B. coagulans B. selenatarsenatis B. pseudomycoides B. oryzaecorticis

B. fumarioli B. coahuilensis B. selenitireducens B. psychrodurans B. oshimensis

B. funiculus B. cohnii B. seohaeanensis B. psychrophilus B. pabuli

B. fusiformis B. composti B. shacheensis B. psychrosaccharolyticus B. pakistanensis

B. galactophilus B. psychrotolerans

B. galactosidilyticus B. curdlanolyticus B. shackletonii B. pulvifaciens B. pallidus

B. galliciensis B. cycloheptanicus B. siamensis B. pumilus B. pallidus

B. gelatini B. cytotoxicus B. silvestris B. purgationiresistens B. panacisoli

B. gibsonii B. daliensis B. simplex B. pycnus B. panaciterrae

B. ginsengi B. decisifrondis B. siralis B. qingdaonensis B. pantothenticus

B. ginsengihumi B. decolorationis B. smithii B. qingshengii B. parabrevis

B. ginsengisoli B. deserti B. soli B. reuszeri B. paraflexus

B. globisporus (eg, B. B. solimangrovi B. rhizosphaerae B. pasteurii g. subsp. Globisporus; or B. B. solisalsi B. rigui B. patagoniensis g. subsp . Marinus) B. songklensis B. ruris

B. sonorensis B. safensis

B. sphaericus B. salarius

B. sporothermodurans

B. stearothermophilus

B. stratosphericus

B. subterraneus

B. subtilis (eg, B. s. subsp. Inaquosorum; or B. s. subsp. Spizizeni; or B. s. subsp. Subtilis)'

Caenimonas Campylobacter Cardiobacterium Catenuloplanes Curtobacterium

Caenimonas koreensis Campylobacter coli Cardiobacterium hominis Catenuloplanes atrovinosus Curtobacterium

Campylobacter concisus Catenuloplanes castaneus albidum

Caldalkalibacillus Campylobacter curvus Carnimonas Catenuloplanes crispus Curtobacterium citreus

Caldalkalibacillus uzonensis Campylobacter fetus Carnimonas nigrificans Catenuloplanes indicus

Campylobacter gracilis Catenuloplanes japonicus

Caldanaerobacter Carnobacterium

Campylobacter helveticus Catenuloplanes nepalensis

Caldanaerobacter subterraneus Carnob acterium

Campylobacter hominis Catenuloplanes niger alterfunditum

Caldanaerobius Campylobacter hyointestinalis

Carnobacterium divergens

Campylobacter jeju Chryseobacterium

Caldanaerobius ftjiensis ni

Carnobacterium funditum bius Camp Chryseobacterium

Caldanaero ylobacter lari

Carnobacterium gallinarum

Campylo balustinum polys accharolyticus bacter mucosalis

Carnobacterium

Caldanaerobius zeae Campylobacter rectus maltaromaticum Citrobacter

Campylobacter showae

Carnobacterium mobile C. amalonaticus

Caldanaerovirga Campylobacter sputorum

Carnobacterium viridans C. braakii

Caldanaerovirga acetigignens Campylobacter upsaliensis

C. diversus

Caryophanon

C aldicellulosiruptor Capnocytophaga C. farmeri

Caryophanon latum

Caldicellulosiruptor bescii Capnocytophaga canimorsus C. freundii

Caryophanon tenue

Caldicellulosiruptor kristjanssonii Capnocytophaga cynodegmi C. gillenii

Caldicellulosiruptor owensensis Capnocytophaga gingivalis C. koseri

Catellatospora

Capnocytophaga granulosa C. murliniae

Catellatospora citrea

Capnocytophaga haemolytica C. pasteurii

Catellatospora

Capnocytophaga ochracea C. rodentium methionotrophica

Capnocytophaga sputigena C. sedlakii

C. werkmanii

Catenococcus C. youngae

Catenococcus thiocycli

Clostridium

(see below)

Coccochloris

Coccochloris elabens

Corynebacterium

Corynebacterium flavescens

Corynebacterium variabile

Clostridium

Clostridium absonum, Clostridium aceticum, Clostridium acetireducens, Clostridium acetobutylicum, Clostridium acidisoli, Clostridium aciditolerans, Clostridium acidurici, Clostridium aerotolerans, Clostridium aestuarii, Clostridium akagii, Clostridium aldenense, Clostridium aldrichii, Clostridium algidicarni, Clostridium algidixylanolyticum, Clostridium algifaecis, Clostridium algoriphilum, Clostridium alkalicellulosi, Clostridium aminophilum, Clostridium aminovalericum, Clostridium amygdalinum, Clostridium amylolyticum, Clostridium arbusti, Clostridium arcticum, Clostridium argentinense, Clostridium asparagi forme, Clostridium aurantibutyricum, Clostridium autoethanogenum, Clostridium baratii, Clostridium barkeri, Clostridium bartlettii, Clostridium beijerinckii, Clostridium bifermentans, Clostridium bolteae, Clostridium bornimense, Clostridium botulinum, Clostridium bowmanii, Clostridium bryantii, Clostridium butyricum, Clostridium cadaveris, Clostridium caenicola, Clostridium camini thermale, Clostridium carboxidivorans, Clostridium carnis, Clostridium cavendishii, Clostridium celatum, Clostridium celerecrescens, Clostridium cellobioparum, Clostridium cellulofermentans, Clostridium cellulolyticum, Clostridium cellulosi, Clostridium cellulovorans, Clostridium chartatabidum, Clostridium chauvoei, Clostridium chromiireducens, Clostridium citroniae, Clostridium clariflavum, Clostridium clostridioforme, Clostridium coccoides, Clostridium cochlearium, Clostridium colletant, Clostridium colicanis,

Clostridium colinum, Clostridium collagenovorans, Clostridium cylindrosporum, Clostridium difficile, Clostridium diolis, Clostridium disporicum, Clostridium drakei, Clostridium durum, Clostridium estertheticum, Clostridium estertheticum estertheticum, Clostridium estertheticum laramiense, Clostridium fallax, Clostridium felsineum, Clostridium fervidum, Clostridium fimetarium, Clostridium formicaceticum, Clostridium frigidicarnis, Clostridium frigoris, Clostridium ganghwense, Clostridium gasigenes, Clostridium ghonii, Clostridium glycolicum, Clostridium glycyrrhizinilyticum, Clostridium grantii, Clostridium haemolyticum, Clostridium halophilum, Clostridium hastiforme, Clostridium hathewayi, Clostridium herbivorans, Clostridium hiranonis, Clostridium histolyticum, Clostridium homopropionicum, Clostridium huakuii, Clostridium hungatei, Clostridium hydrogenif ormans, Clostridium hydroxybenzoicum, Clostridium hylemonae, Clostridium jejuense, Clostridium indolis, Clostridium innocuum, Clostridium intestinale, Clostridium irregulare, Clostridium isatidis, Clostridium josui, Clostridium kluyveri, Clostridium lactatifermentans, Clostridium lacusfryxellense, Clostridium laramiense, Clostridium lavalense, Clostridium lentocellum, Clostridium lentoputrescens, Clostridium leptum, Clostridium limosum, Clostridium litorale, Clostridium lituseburense, Clostridium ljungdahlii, Clostridium lortetii, Clostridium lundense, Clostridium magnum, Clostridium malenominatum, Clostridium mangenotii, Clostridium mayombei, Clostridium methoxybenzovorans, Clostridium methylpentosum, Clostridium neopropionicum, Clostridium nexile, Clostridium nitrophenolicum, Clostridium novyi, Clostridium oceanicum, Clostridium orbiscindens, Clostridium oroticum, Clostridium oxalicum, Clostridium papyrosolvens, Clostridium paradoxum, Clostridium paraperfringens (Alias: C. welchii), Clostridium paraputrificum, Clostridium pascui, Clostridium pasteurianum, Clostridium peptidivorans, Clostridium perenne, Clostridium perfringens, Clostridium pfennigii, Clostridium phytofermentans, Clostridium piliforme, Clostridium polysaccharolyticum, Clostridium populeti, Clostridium propionicum, Clostridium proteoclasticum, Clostridium proteolyticum, Clostridium psychrophilum, Clostridium puniceum, Clostridium purinilyticum, Clostridium putrefaciens, Clostridium putrificum, Clostridium quercicolum, Clostridium quinii, Clostridium ramosum, Clostridium rectum, Clostridium roseum, Clostridium saccharobutylicum, Clostridium saccharogumia, Clostridium saccharolyticum, Clostridium saccharoperbutylacetonicum, Clostridium sardiniense, Clostridium sartagoforme, Clostridium scatologenes, Clostridium schirmacherense, Clostridium scindens, Clostridium septicum, Clostridium sordellii, Clostridium sphenoides, Clostridium spiroforme, Clostridium sporogenes, Clostridium sporosphaeroides, Clostridium stercorarium, Clostridium stercorarium leptospartum, Clostridium stercorarium stercorarium, Clostridium stercorarium thermolacticum, Clostridium sticklandii, Clostridium straminisolvens, Clostridium subterminale, Clostridium sufflavum, Clostridium sulfidigenes, Clostridium symbiosum, Clostridium tagluense, Clostridium tepidiprofundi, Clostridium termitidis, Clostridium tertium, Clostridium tetani, Clostridium tetanomorphum,

Clostridium thermaceticum, Clostridium thermautotrophicum, Clostridium thermoalcaliphilum, Clostridium thermobutyricum, Clostridium thermocellum,

Clostridium thermocopriae, Clostridium thermohydrosulfuricum, Clostridium thermolacticum, Clostridium thermopalmarium, Clostridium thermopapyrolyticum, Clostridium thermosaccharolyticum, Clostridium thermosuccinogenes, Clostridium thermosulfurigenes, Clostridium thiosulfatireducens, Clostridium tyrobutyricum, Clostridium uliginosum, Clostridium ultunense, Clostridium villosum, Clostridium vincentii, Clostridium viride, Clostridium xylanolyticum, Clostridium xylanovorans

Dactylosporangium Deinococcus Delftia Echinicola

Dactylosporangium aurantiacum Deinococcus aerius Delftia acidovorans Echinicola pacifica

Dactylosporangium fulvum Deinococcus apachensis Desulfovibrio Echinicola vietnamensis

Dactylosporangium matsuzakiense Deinococcus aquaticus Desulfovibrio desulfuricans

Dactylosporangium roseum Deinococcus aquatilis Diplococcus

Dactylosporangium thailandense Deinococcus caeni Diplococcus pneumoniae

Dactylosporangium vinaceum Deinococcus radiodurans

Deinococcus radiophilus

Enterobacter Enterobacter kobei Faecalibacterium Flavobacterium

E. aerogenes E. ludwigii Faecalibacterium prausnitzii Flavobacterium antarcticum

E. amnigenus E. mori Fangia Flavobacterium aquatile

E. agglomerans E. nimipressuralis Fangia hongkongensis Flavobacterium

E. arachidis E. oryzae Fastidiosipila aquidurense

E. asburiae E. pulveris Fastidiosipila sanguinis Flavobacterium balustinum

E. cancerogenous E. pyrinus Fusobacterium Flavobacterium croceum

E. cloacae E. radicincitans Fusobacterium nucleatum Flavobacterium cucumis

E. cowanii E. taylorae Flavobacterium

E. dissolvens E. turicensis daejeonense

E. gergoviae E. sakazakii Enterobacter soli Flavobacterium defluvii

E. helveticus Enterococcus Flavobacterium degerlachei

E. hormaechei Enterococcus durans Flavobacterium

E. intermedins Enterococcus faecalis denitrificans

Enterococcus faecium Flavobacterium filum

Erwinia Flavobacterium flevense

Erwinia hapontici Flavobacterium frigidarium

Escherichia Flavobacterium mizutaii

Escherichia coli Flavobacterium okeanokoites

Gaetbulibacter Haemophilus Ideonella Janibacter

Gaetbulibacter saemankumensis Haemophilus aegyptius Ideonella azotifigens Janibacter anophelis

Gallibacterium Haemophilus aphrophilus Idiom arina Janibacter corallicola

Gallibacterium anatis Haemophilus felis Idiomarina abyssalis Janibacter limosus

Gallicola Haemophilus gallinarum Idiomarina baltica Janibacter melonis

Gallicola barnesae Haemophilus haemolyticus Idiomarina fontislapidosi Janibacter terrae

Garciella Haemophilus influenzae Idiomarina loihiensis Jannaschia

Garciella nitratireducens Haemophilus paracuniculus Idiomarina ramblicola Jannaschia cystaugens

Geobacillus Haemophilus parahaemolyticus Idiomarina seosinensis Jannaschia helgolandensis

Geobacillus thermoglucosidasius Haemophilus parainfluenzae Idiomarina zobellii Jannaschia pohangensis

Geobacillus stearothermophilus Haemophilus Ignatzschineria Jannaschia rubra

Geobacter paraphrohaemolyticus Ignatzschineria larvae

Geobacter bemidjiensis Haemophilus parasuis Janthinobacterium

Geobacter bremensis Haemophilus pittmaniae Ignavigranum Janthinobacterium

Geobacter chapellei Hafnia Ignavigranum ruoffiae agaricidamnosum

Geobacter grbiciae Haftiia alvei Ilumatobacter Janthinobacterium lividum

Geobacter hydrogenophilus Hahella Ilumatobacter fluminis Jejuia

Geobacter lovleyi Hahella ganghwensis Ilyobacter Jejuia pallidilutea

Geobacter metallireducens Halalkalibacillus Ilyobacter delafieldii Jeotgalibacillus

Geobacter pelophilus Halalkalibacillus halophilus Ilyobacter insuetus Jeotgalibacillus

Geobacter pickeringii Helicobacter Ilyobacter polytropus alimentarius

Geobacter sulfurreducens Helicobacter pylori Ilyobacter tartaricus Jeotgalicoccus

Geodermatophilus Jeotgalicoccus halotolerans

Geodermatophilus obscurus

Gluconacetobacter

Gluconacetobacter xylinus

Gordonia

Gordonia rubripertincta

Kaistia Labedella Listeria ivanovii Micrococcus Nesterenkonia

Kaistia adipata Labedella gwakjiensis L. marthii Micrococcus luteus Nesterenkonia holobia

Kaistia soli Labrenzia L. monocytogenes Micrococcus lylae Nocardia

Kangiella Labrenzia aggregata L. newyorkensis Moraxella Nocardia argentinensis

Kangiella aquimarina Labrenzia alba L. riparia Moraxella bovis Nocardia corallina

Kangiella koreensis Labrenzia alexandrii L. rocourtiae Moraxella nonliquefaciens Nocardia

Labrenzia marina L. seeligeri Moraxella osloensis otitidiscaviarum

Kerstersia Labrys L. weihenstephanensis Nakamurella

Kerstersia gyiorum Labrys methylaminiphilus L. welshimeri Nakamurella multipartita

Kiloniella Labrys miyagiensis Listonella Nannocystis

Kiloniella laminariae Labrys monachus Listonella anguillarum Nannocystis pusilia

Klebsiella Labrys okinawensis Macrococcus Natranaerobius

K. granulomatis Labrys portucalensis Macrococcus bovicus Natranaerobius

K. oxytoca Marinobacter thermophilus

K. pneumoniae Lactobacillus Marinobacter algicola Natranaerobius trueperi

K. terrigena [see below] Marinobacter bryozoorum Naxibacter

K. variicola Laceyella Marinobacter flavimaris Naxibacter alkalitolerans

Kluyvera Laceyella putida Meiothermus Neisseria

Kluyvera ascorbata Lechevalieria Meiothermus ruber Neisseria cinerea

Kocuria Lechevalieria aerocolonigenes Methylophilus Neisseria denitrificans

Kocuria roasea Legionella Methylophilus Neisseria gonorrhoeae

Kocuria varians [see below] methylotrophus Neisseria lactamica

Kurthia Listeria Microbacterium Neisseria mucosa

Kurthia zopfii L. aquatica Microbacterium Neisseria sicca

L. booriae ammoniaphilum Neisseria subflava

L. cornellensis Microbacterium arborescens Neptunomonas

L. fleischmannii Microbacterium liquefaciens Neptunomonas japonica

L. floridensis Microbacterium oxydans

L. grandensis

L. grayi

L. innocua

Lactobacillus

L. acetotolerans L. catenaformis L. mali L. parakefiri L. sakei

L. acidifcirinae L. ceti L. manihotivorans L. paralimentarius L. salivarius

L. acidipiscis L. coleohominis L. mindensis L. paraplantarum L. sanfranciscensis

L. acidophilus L. collinoides L. mucosae L. pentosus L. satsumensis

Lactobacillus agilis L. composti L. murinus L. perolens L. secaliphilus

L. algidus L. concavus L. nagelii L. plantarum L. sharpeae

L. alimentarius L. coryniformis L. namurensis L. pontis L. siliginis

L. amylolyticus L. crispatus L. nantensis L. protectus L. spicheri

L. amylophilus L. crustorum L. oligofermentans L. psittaci L. suebicus

L. amylotrophicus L. curvatus L. oris L. rennini L. thailandensis

L. amylovorus L. panis L. reuteri L. ultunensis

L. animalis L. delbrueckii subsp. L. pantheris L. rhamnosus L. vaccinostercus

L. antri bulgaricus L. parabrevis L. rimae L. vaginalis

L. apodemi L. delbrueckii subsp. L. parabuchneri L. rogosae L. versmoldensis

L. aviarius delbrueckii L. paracasei L. rossiae L. vini

L. bifermentans L. delbrueckii subsp. lactis L. paracollinoides L. ruminis L. vitulinus

L. brevis L. dextrinicus L. parafarraginis L. saerimneri L. zeae

L. buchneri L. diolivorans L. homohiochii L. jensenii L. zymae

L. camelliae L. equi L. iners L. johnsonii L. gastricus

L. casei L. equigenerosi L. ingluviei L. kalixensis L. ghanensis

L. kitasatonis L. farraginis L. intestinalis L. kefiranofaciens L. graminis

L. kunkeei L. farciminis L. fuchuensis L. kefiri L. hammesii

L. leichmannii L. fermentum L. gallinarum L. kimchii L. hamsteri

L. lindneri L. fornicalis L. gasseri L. helveticus L. harbinensis

L. malefermentans L. fructivorans L. hilgardii L. hayakitensis

L. frumenti

Legionella

Legionella adelaidensis Legionella drancourtii Candidatus Legionella jeonii Legionella quinlivanii

Legionella anisa Legionella dresdenensis Legionella jordanis Legionella rowbothamii

Legionella beliardensis Legionella drozanskii Legionella lansingensis Legionella rubrilucens

Legionella birminghamensis Legionella dumoffii Legionella londiniensis Legionella sainthelensi

Legionella bozemanae Legionella erythra Legionella longbeachae Legionella santicrucis

Legionella brunensis Legionella fairfieldensis Legionella lytica Legionella shakespearei

Legionella busanensis Legionella fallonii Legionella maceachernii Legionella spiritensis

Legionella cardiaca Legionella feeleii Legionella massiliensis Legionella steelei

Legionella cherrii Legionella geestiana Legionella micdadei Legionella steigerwaltii

Legionella Cincinnati ensis Legionella genomospecies Legionella monrovica Legionella taurinensis

Legionella clemsonensis Legionella gormanii Legionella moravica Legionella tucsonensis

Legionella donaldsonii Legionella gratiana Legionella nagasakiensis Legionella tunisiensis

Legionella gresilensis Legionella nautarum Legionella wadsworthii

Legionella hackeliae Legionella norrlandica Legionella waltersii

Legionella impletisoli Legionella oakridgensis Legionella worsleiensis

Legionella israelensis Legionella parisiensis Legionella yabuuchiae

Legionella jamestowniensis Legionella pittsburghensis

Legionella pneumophila

Legionella quateirensis

Oceanibulbus Paenibacillus Prevotella Quadrisphaera

Oceanibulbus indolifex Paenibacillus thiaminolyticus Prevotella albensis Quadrisphaera granulorum

Oceanicaulis Pantoea Prevotella amnii Quatrionicoccus

Oceanicaulis alexandrii Pantoea agglomerans Prevotella bergensis Quatrioni coccus

Oceanicola Prevotella bivia australiensis

Oceanicola batsensis Paracoccus Prevotella brevis

Paracoccus alcaliphilus Prevotella bryantii

Oceanicola granulosus Paucimonas Prevotella buccae Quinella

Oceanicola nanhaiensis Paucimonas lemoignei Prevotella buccalis Quinella ovalis

Oceanimonas Pectobacterium Prevotella copri

Oceanimonas baumannii Pectobacterium aroidearum Prevotella dentalis Ralstonia

Oceaniserpentilla Pectobacterium atrosepticum Prevotella denticola Ralstonia eutropha

Oceaniserpentilla haliotis Pectobacterium Prevotella disiens Ralstonia insidiosa

Oceanisphaera betavasculorum Prevotella histicola Ralstonia mannitolilytica

Oceanisphaera donghaensis Pectobacterium cacticida Prevotella intermedia Ralstonia pickettii

Oceanisphaera litoralis Pectobacterium carnegieana Prevotella maculosa Ralstonia

Oceanithermus Pectobacterium carotovorum Prevotella marshii pseudosolanacearum

Oceanithermus desulfurans Pectobacterium chrysanthemi Prevotella melaninogenica Ralstonia syzygii

Oceanithermus profundus Pectobacterium cypripedii Prevotella micans Ralstonia solanacearum

Oceanobacillus Pectobacterium rhapontici Prevotella multiformis Ramlibacter

Oceanobacillus caeni Pectobacterium wasabiae Prevotella nigrescens Ramlibacter henchirensis

Oceanospirillum Pianococcus Prevotella oralis Ramlibacter tataouinensis

Oceanospirillum linum Pianococcus citreus Prevotella oris P1anomicrobium Prevotella oulorum Raoultella

Planomicrobium okeanokoites Prevotella pallens Raoultella ornithinolytica P1esiomonas Prevotella salivae Raoultella planticola

Plesiomonas shigelloides Prevotella stercorea Raoultella terrigena

Proteus Prevotella tannerae Rathayibacter

Proteus vulgaris Prevotella timonensis Rathayibacter caricis

Prevotella veroralis Rathayibacter festucae

Providencia Rathayibacter iranicus

Providencia stuartii Rathayibacter rathayi

Pseudomonas Rathayibacter toxicus

Pseudomonas aeruginosa Rathayibacter tritici

Pseudomonas alcaligenes Rhodobacter

Pseudomonas anguillispetica Rhodobacter sphaeroides

Pseudomonas fluorescens Ruegeria

Pseudoalteromonas Ruegeria gelatinovorans haloplanktis

Pseudomonas mendocina

Pseudomonas pseudoalcali genes

Pseudomonas putida

Pseudomonas tutzeri

Pseudomonas syringae

Psychrobacter

Psychrobacter faecalis

Psychrobacter phenylpyruvicus

Saccharococcus Sagittula Sanguibacter Stenotrophomonas Tatlockia

Saccharococcus thermophilus Sagittula stellata Sanguihacter keddieii Stenotrophomonas Tatlockia maceachemii

Sanguibacter suarezii maltophilia Tatlockia micdadei

Saccharomonospora Salegentibacter Streptococcus Tenacibaculum

Saccharomonospora azurea Salegentibacter salegens Saprospira Tenacibaculum

Saccharomonospora cyanea Saprospira grandis [also see below] amylolyticum

Saccharomonospora viridis Salimicrobium

Tenacibaculum discolor

Salimicrobium album Sarcina Streptomyces Tenacibaculum

Saccharophagus Sarcina maxima Streptomyces charophagus degradans Salinib gallaicum

Sac acter Sarcina ventriculi achromogenes

Salinibacter ruber Tenacibaculum

Streptomyces cesalbus

Saccharopolyspora Sebaldella lutimaris olyspora erythraea Sali Streptomyces cescaepitosus

Saccharop nicoccus Sebaldella termitidis Tenacibaculum orii Salini Streptomyces cesdiastaticus

Saccharopolyspora greg coccus alkaliphilus mesophilum

Salinicoccus hispanic Streptomyces cesexfoliatus

Saccharopolyspora hirsuta us Tenacibaculum

Streptomyc ropolyspora hordei Salinicoccus es fimbriatus

Saccha roseus skagerrakense

Serratia Streptomyces fradiae

Saccharopolyspora rectivirgula Tepidanaerobacter

Salinispora Serratia fonticola Streptomyces fulvissimus

Saccharopolyspora spinosa Tepidanaerobacter

Salinispora arenicola Serratia marcescens Streptomyces griseoruber

Saccharopolyspora taheri syntrophicus

Salinispora tropica Streptomyces griseus Tepidibacter

Sphaerotilus

Saccharothrix Streptomyces lavendulae Tepidibacter

Salinivibrio Sphaerotilus natans

Saccharothrix australiensis Streptomyces formicigenes

Salinivibrio costicola

Saccharothrix coeruleofusca phaeochromogenes

Streptomyces

Saccharothrix espanaensis thermodiastaticus Tepidibacter

Saccharothrix longispora Salmonella Sphingobacterium Streptomyces tubercidicus thalassicus

Saccharothrix mutabilis Salmonella bongori Sphingobacterium multivorum Thermus

Saccharothrix syringae Salmonella enterica Thermus aquaticus

Salmonella subterran Staphylococcus

Saccharothrix tangerinus ea Thermus flliformis

Salmonel [see below]

Saccharothrix texasensis la typhi Thermus thermophilus

Staphylococcus

S. arlettae S. microti

S. agnetis S. equorum S. muscae S. schleiferi

S. aureus S. felis S. nepalensis S. sciuri

S. auricularis S. fleurettii S. pasteuri S. simiae

S. capitis S. gallinarum S. petrasii S. simulans

S. caprae S. haemolyticus S. pettenkoferi S. Stepanovich

S. carnosus S. hominis S. piscifermentans S. succinus

S. caseolyticus S. hyicus S. pseudintermedius S. vitulinus

S. chromogenes S. intermedius S. pseudolugdunensis S. warneri

S. cohnii S. kloosii S. pulvereri S. xylosus

S. condimenti S. leei S. rostri

S. delphini S. lentus S. saccharolyticus

S. devriesei S. lugdunensis S. saprophyticus

S. epidermidis S. lutrae

S. lyticans

S. massiliensis

Streptococcus

Streptococcus agalactiae Streptococcus infantarius Streptococcus orisratti Streptococcus thermophilus

Streptococcus anginosus Streptococcus iniae Streptococcus parasanguinis Streptococcus sanguinis

Streptococcus bovis Streptococcus intermedius Streptococcus peroris Streptococcus sohrinus

Streptococcus canis Streptococcus lactarius Streptococcus pneumoniae Streptococcus suis

Streptococcus constellatus Streptococcus milleri Streptococcus Streptococcus uheris

Streptococcus downei Streptococcus mitis pseudopneumoniae Streptococcus vestibularis

Streptococcus dysgalactiae Streptococcus mutans Streptococcus pyogenes Streptococcus viridans

Streptococcus equines Streptococcus oralis Streptococcus ratti Streptococcus

Streptococcus faecalis Streptococcus tigurinus Streptococcus salivariu zooepidemicus

Streptococcus ferus

Uliginosibacterium Vagococcus Vibrio Virgibacillus Xanthobacter

Vagococcus carniphilus Vibrio aerogenes Virgibacillus Xanthobacter agilis

Uliginosibacterium gangwonense Vagococcus elongatus Vibrio aestuarianus halodenitrificans Xanthobacter

Ulvibacter Vagococcus fessus Vibrio albensis Virgibacillus aminoxidans

Ulvibacter litoralis Vagococcus fluvialis Vibrio alginolyticus pantothenticus Xanthobacter

Vibrio campbellii autotrophicus

Umezawaea Vagococcus lutrae Vibrio cholerae Xanthobacter flavus

Umezawaea tangerina Vagococcus salmoninarum Vibrio Cincinnati ensis Weissella Xanthobacter tagetidis

Undibacterium Vibrio coralliilyticus Weissella cibaria Xanthobacter viscosus igrum Variovo Weissella confusa

Undibacterium p rax Vibrio cyclitrophicus Xanthomonas

Variovorax boronicumula Weissella halotolerans

Ureaplasma ns Vibrio diazotrophicus Xanthomonas

Weisse

Ureaplasma urealyticum Variovorax dokdonensis Vibrio fluvialis lla hellenica albilineans

Variovorax paradoxus Vibrio furnissii Weissella kandleri Xanthomonas alfalfae

Ureibacillus Variovorax soli Vibrio gazogenes Weissella koreensis Xanthomonas

Weissella minor

Ureibacillus composti Vibrio halioticoli arboricola

Veillonella

Ureibacillus suwonensis Vibrio harveyi Weissella Xanthomonas

Veillonella atypica paramesenteroides

Ureibacillus terrenus Vibrio ichthyoenteri axonopodis

Veillonella caviae

Ureibacillus thermophilus Vibrio mediterranei Weissella soli Xanthomonas

Veillonella criceti

Ureibacillus thermosphaericus Vibrio metschnikovii Weissella thailandensis campestris

Veillonella dispar

Vibrio mytili Weissella viridescens Xanthomonas citri

Veillonella montpellierensis

Vibrio natriegens Xanthomonas codiaei

Veillonella parvula Williamsia

Vibrio navarrensis Xanthomonas

Veillonella ratti Williamsia marianensis

Vibrio nereis cucurbitae

Veillonella rodentium Williamsia marts

Vibrio nigripulchritudo Xanthomonas

Williamsia serinedens

Venenivibrio Vibrio ordalii euvesicatoria

Venenivibrio stagnispumantis Vibrio orientalis Winogradskyella Xanthomonas fragariae

Vibrio parahaemolyticus Winogradskyella Xanthomonas fuscans

Vibrio pectenicida thalassocola Xanthomonas gardneri

Vibrio penaeicida Xanthomonas hortorum

Verminephrobacter Vibrio proteolyticus Wolbachia Xanthomonas hyacinthi

Verminephrobacter eiseniae Vibrio shilonii Wolbachia persica Xanthomonas perforans

Vibrio splendidus Xanthomonas phaseoli

Vibrio tubiashii Xanthomonas pisi

Verrucomicrobium Vibrio vulnificus Wolinella Xanthomonas populi

Verrucomicrobium spinosum Wolinella succinogenes Xanthomonas theicola

Xanthomonas translucens

Zobellia Xanthomonas

Zobellia galactanivorans vesicatoria

Zobellia uliginosa

Xylella

Zoogloea Xylella fastidiosa

Zoogloea ramigera

Xylophilus

Zoogloea resiniphila

Xylophilus ampelinus

Xenophilus Yangia Yersinia mollaretii Zooshikella Zobellella

Xenophilus azovorans Yangia pacifica Yersinia philomiragia Zooshikella ganghwensis Zobellella denitrificans

Xenorhabdus Yersinia pestis Zobellella taiwanensis

Xenorhabdus beddingii

Xenorhabdus bovienii Yersinia pseudotuberculosis

Xenorhabdus cabanillasii Yaniella Yersinia rohdei Zunongwangia

Xenorhabdus douce tiae Yaniella flava Yersinia ruckeri Zunongwangia profunda

Yaniella halotolera Zeaxanthinibacter

Xenorhabdus griffiniae ns

Y Zymobacter Zeaxanthinibacter

Xenorhabdus hominickii okenella

Yeosuana Zymobacter enoshimensis nhoeferi Yokenella re palmae

Xenorhabdus koppe gensburgei

Yeosuana aromativorans

Xenorhabdus nematophila

Yonghaparkia Zymomonas Zhihengliuella

Xenorhabdus poinarii Yersinia Yonghaparkia alkaliphila Zymomonas mobilis Zhihengliuella

Xylanibacter Yersinia aldovae halotolerans

Xylanibacter oryzae Yersinia bercovieri Zavarzinia Zymophilus Xylanibacterium

Yersinia enterocolitica Zavarzinia compransoris Zymophilus paucivorans Xylanibacterium ulmi

Yersinia entomophaga Zymophilus raffinosivorans

Yersinia frederiksenii

Yersinia intermedia

Yersinia kristensenii

TABLE 3: Medicaments

TABLE 4: Example Conjugation Genes