Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
GENE EXPRESSION IN BACTEROIDES
Document Type and Number:
WIPO Patent Application WO/2016/201174
Kind Code:
A2
Abstract:
Provided herein, in some aspects, are tools (e.g., methods, compositions and nucleic acids) for building genetic circuits in Bacteroides and Parabacteroides bacteria, as well as the bacteria containing the genetic circuits.

Inventors:
LU TIMOTHY KUAN-TA (US)
MIMEE MARK K (US)
VOIGT CHRISTOPHER (US)
TUCKER ALEX C (US)
Application Number:
PCT/US2016/036811
Publication Date:
December 15, 2016
Filing Date:
June 10, 2016
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
MASSACHUSETTS INST TECHNOLOGY (US)
International Classes:
A61K39/02; C12N1/21
Other References:
See references of EP 3307306A4
Attorney, Agent or Firm:
DIPIETRANTONIO, Heather, J. (Greenfield & Sacks P.C.,600 Atlantic Avenu, Boston Massachusetts, US)
Download PDF:
Claims:
What is claimed is:

CLAIMS

1. A Bacteroides bacterium comprising:

(a) an engineered nucleic acid comprising a region containing a promoter and a nucleotide sequence encoding a ribosomal binding site (RBS) operably linked to a nucleotide sequence encoding a recombinase; and

(b) an engineered nucleic acid comprising a promoter and a nucleotide sequence encoding a RBS operably linked to a nucleotide sequence encoding a molecule of interest, wherein the nucleotide sequence encoding the molecule of interest is flanked by a pair of cognate recombinase recognition sequences.

2. The Bacteroides bacterium of claim 1, wherein the nucleotide sequence encoding a RBS comprises a sequence selected from the group consisting SEQ ID NO: 1 - SEQ ID NO: 143 and SEQ ID NO: 168 - SEQ ID NO: 172.

3. The Bacteroides bacterium of claim 1, wherein the promoter of (a) and/or (b) is constitutive. 4. The Bacteroides bacterium of claim 3, wherein the region containing a promoter and a nucleotide sequence encoding a RBS comprises a sequence selected from the group consisting SEQ ID NO: 151 - SEQ ID NO: 155 and SEQ ID NO: 160 - SEQ ID NO: 163.

5. The Bacteroides bacterium of claim 1, wherein the promoter of (a) and/or (b) is inducible.

6. The Bacteroides bacterium of claim 5, wherein the region containing a promoter and a nucleotide sequence encoding a RBS comprises a sequence selected from the group consisting SEQ ID NO: 144 - SEQ ID NO: 149.

7. The Bacteroides bacterium of any one of claims 1-6, wherein the recombinase is a serine recombinase or a tyrosine recombinase.

8. The Bacteroides bacterium of claim 7, wherein the recombinase is a serine recombinase.

9. The Bacteroides bacterium of claim 8, wherein the serine recombinase is selected from the group consisting of Int7, Int8, Int9, Intl2, Bxbl, (|)C31, TP901, TGI, φΒΤΙ, R4, cpRVl, (pFCl, MRll, A118, U153 and gp29.

10. The Bacteroides bacterium of claim 9, wherein the serine recombinase is Int7 encoded by a nucleotide sequence of SEQ ID NO: 164.

11. The Bacteroides bacterium of claim 9, wherein the serine recombinase is Int8 encoded by a nucleotide sequence of SEQ ID NO: 165.

12. The Bacteroides bacterium of claim 9, wherein the serine recombinase is Int9 encoded by a nucleotide sequence of SEQ ID NO: 166.

13. The Bacteroides bacterium of claim 9, wherein the serine recombinase is Intl2 encoded by a nucleotide sequence of SEQ ID NO: 167. 14. The Bacteroides bacterium of any one of claims 1-13, wherein the engineered nucleic acid of (a) is on the same vector as the engineered nucleic acid of (b).

15. The Bacteroides bacterium of claim 14, wherein the vector further comprises a promoter operably linked to a nucleic acid encoding a recombinase derived from a conjugated transposon.

16. The Bacteroides bacterium of claim 15, wherein the recombinase is IntNl.

17. The Bacteroides bacterium of claim 15, wherein the recombinase is IntN2.

18. The Bacteroides bacterium of any one of claims 1-17, wherein the engineered nucleic acid of (a) and the engineered nucleic acid of (b) are integrated into a chromosome of the Bacteroides bacterium.

19. The Bacteroides bacterium of any one of claims 1-17, wherein the vector is a plasmid.

20. A Bacteroides bacterium comprising an engineered nucleic acid comprising a promoter and a nucleotide sequence encoding a ribosomal binding site (RBS) operably linked to a nucleotide sequence encoding a molecule of interest, wherein the RBS comprises a sequence selected from the group consisting SEQ ID NO: 1 - SEQ ID NO: 143 and SEQ ID NO: 168 - SEQ ID NO: 172. 21. A Bacteroides bacterium comprising an engineered nucleic acid comprising a region containing a constitutive promoter and a nucleotide sequence encoding a ribosomal binding site (RBS) operably linked to a nucleotide sequence encoding a molecule of interest, wherein the region containing a constitutive promoter and a RBS comprises a sequence selected from the group consisting SEQ ID NO: 151 - SEQ ID NO: 155 and SEQ ID NO: 160 - SEQ ID NO: 163.

22. A Bacteroides bacterium comprising an engineered nucleic acid comprising a region containing an inducible promoter and a nucleotide sequence encoding a ribosomal binding site (RBS) operably linked to a nucleotide sequence encoding a molecule of interest, wherein the region containing an inducible promoter and a RBS comprises a sequence selected from the group consisting SEQ ID NO: 144 - SEQ ID NO: 149.

23. A Bacteroides bacterium comprising an engineered nucleic acid comprising a region containing an inducible promoter and a nucleotide sequence encoding a ribosomal binding site (RBS) operably linked to a nucleotide sequence encoding a molecule of interest, wherein the nucleotide sequence encoding a RBS is immediately downstream from a 10- to 20- nucleotide region, wherein at least 80% of the nucleotides in the 10- to 20-nucleotide region are adenine or thymine. 24. The Bacteroides bacterium of any one of claims 1-19, wherein the molecule of interest is a therapeutic molecule, a prophylactic molecule, or a diagnostic molecule.

25. A method of expressing a molecule of interest in a Bacteroides bacterium, the method comprising culturing a Bacteroides bacterium of any one of claims 1-19 under conditions that result in expression of the molecule of interest. 26. A method of treating a condition in a subject, the method comprising administering to the subject the Bacteroides bacterium of any one of claims 1-19, wherein the molecule of interest is a therapeutic molecule.

27. A method of preventing a condition in a subject, the method comprising administering to the subject the Bacteroides bacterium of any one of claims 1-19, wherein the molecule of interest is a prophylactic molecule.

28. A Bacteroides bacterium comprising:

(a) an engineered nucleic acid comprising a region containing a promoter and a nucleotide sequence encoding a ribosomal binding site (RBS) operably linked to a nucleotide sequence encoding a catalytically-inactive Cas9 nuclease; and

(b) an engineered nucleic acid comprising a promoter and a nucleotide sequence encoding a RBS operably linked to a nucleotide sequence encoding a guide RNA, wherein the guide RNA targets a nucleotide sequence encoding a molecule of interest.

29. The Bacteroides bacterium of claim 28, wherein the catalytically-inactive Cas9 nuclease is encoded by the nucleotide sequence of SEQ ID NO: 157.

30. An engineered nucleic acid comprising a nucleotide sequence selected from the group consisting of SEQ ID NO: 1 - SEQ ID NO: 180.

31. A cell comprising the engineered nucleic acid of claim 30.

32. An engineered nucleic acid comprising the nucleotide sequence of SEQ ID NO: 209.

Description:
GENE EXPRESSION IN BACTEROIDES

RELATED APPLICATIONS

This application claims the benefit under 35 U.S.C. § 119(e) of U.S. provisional application number 62/173,481, filed June 10, 2015, which is incorporated by reference herein in its entirety.

FIELD OF THE INVENTION

Aspects of the present disclosure relate to the general field of biotechnology and, particularly, to the fields of genetic engineering and microbiology.

BACKGROUND OF THE INVENTION

Bacteroides species are prominent Gram-negative anaerobic symbionts of the mammalian gut microbiome, comprising 25% of culturable anaerobes in the human gastrointestinal tract. Of the Bacteroides genus, Bacteroides thetaiotaomicron is both prevalent (present in 46% of humans) and abundant (up to 10 10 per gram stool). Stable and robust colonization of the densely populated gut environment is facilitated by the metabolic diversity of Bacteroides. Specifically, B. thetaiotaomicron and its relatives are equipped with an extensive repertoire of saccharolytic enzymes and serve as primary fermenters of host-, diet- or microbially-derived polysaccharides.

SUMMARY OF THE INVENTION

Bacteroides thetaiotaomicron, a commensal bacterium, forms stable interactions with the gastrointestinal tract and is a candidate for modulating the gut ecosystem. However, there are few genetic parts and circuits available to control expression in this Bacteroides species as well as other Bacteroides and Parabacteroides species. Provided herein is a library of constitutive promoters and ribosome-binding sites that may be used, in some embodiments, to achieve a 10,000-fold range in gene expression. For inducible control, a series of promoters, able to elicit up to 100-fold regulation in gene expression, were constructed. Further provided herein are vector systems that maybe used to manipulate gene expression in a variety of Bacteroides and Parabacteroides species. These tools were used as a platform to build recombinase-based memory gates that permanently record DNA-encoded information in the genome. CRISPR interference (CRISPRi) was used to enable the regulated

knockdown of recombinant and endogenous gene expression. Finally, the function of the inducible systems, CRISPRi, and memory switch were validated in B. thetaiotaomicron colonizing the mouse gut. Collectively, these tools provide a resource to engineer

Bacteroides and Parabacteroides to respond to environmental stimuli, record this information, and control genetic pathways as a means of surveillance of or therapeutic delivery to the human microbiome.

Some aspects of the present disclosure are directed to Bacteroides (or

Parabacteroides) bacteria comprising (a) an engineered nucleic acid comprising a region containing a promoter and a nucleotide sequence encoding a ribosomal binding site (RBS) operably linked to a nucleotide sequence encoding a recombinase, and (b) an engineered nucleic acid comprising a promoter and a nucleotide sequence encoding a RBS operably linked to a nucleotide sequence encoding a molecule of interest, wherein the nucleotide sequence encoding the molecule of interest is flanked by a pair of cognate recombinase recognition sequences.

In some embodiments, wherein the nucleotide sequence encoding a RBS comprises a sequence selected from the group consisting SEQ ID NO: 1 - SEQ ID NO: 143 and SEQ ID NO: 168 - SEQ ID NO: 172.

In some embodiments, the promoter is constitutive. Thus, the region containing a promoter and a nucleotide sequence encoding a RBS may comprise a sequence selected from the group consisting SEQ ID NO: 151 - SEQ ID NO: 155 and SEQ ID NO: 160 - SEQ ID NO: 163. Other constitutive promoters are encompassed by the present disclosure.

In some embodiments, the promoter is inducible. Thus, the region containing a promoter and a nucleotide sequence encoding a RBS may comprise a sequence selected from the group consisting SEQ ID NO: 144 - SEQ ID NO: 149. Other inducible promoters are encompassed by the present disclosure.

In some embodiments, the recombinase is a serine recombinase or a tyrosine recombinase. For example, the recombinase may be a serine recombinase. In some embodiments, the serine recombinase is selected from the group consisting of Int7 (SEQ ID NO: 164), Int8 (SEQ ID NOT: 165), Int9 (SEQ ID NO: 166) and Intl2 (SEQ ID NO: 167). Other serine recombinases and tyrosine recombinases are encompassed by the present disclosure.

Also provided herein are Bacteroides (or Parabacteroides) bacteria comprising an engineered nucleic acid comprising a promoter and a nucleotide sequence encoding a ribosomal binding site (RBS) operably linked to a nucleotide sequence encoding a molecule of interest, wherein the RBS comprises a sequence selected from the group consisting SEQ ID NO: 1 - SEQ ID NO: 143 and SEQ ID NO: 168 - SEQ ID NO: 172.

Further provided herein are Bacteroides (or Parabacteroides) bacteria comprising an engineered nucleic acid comprising a region containing a constitutive promoter and a nucleotide sequence encoding a ribosomal binding site (RBS) operably linked to a nucleotide sequence encoding a molecule of interest, wherein the region containing a constitutive promoter and a RBS comprises a sequence selected from the group consisting SEQ ID NO: 151 - SEQ ID NO: 155 and SEQ ID NO: 160 - SEQ ID NO: 163.

Also provided herein are Bacteroides bacteria comprising an engineered nucleic acid comprising a region containing an inducible promoter and a nucleotide sequence encoding a ribosomal binding site (RBS) operably linked to a nucleotide sequence encoding a molecule of interest, wherein the region containing an inducible promoter and a RBS comprises a sequence selected from the group consisting SEQ ID NO: 144 - SEQ ID NO: 149.

Further provided herein are Bacteroides (or Parabacteroides) bacteria comprising an engineered nucleic acid comprising a region containing an inducible promoter and a nucleotide sequence encoding a ribosomal binding site (RBS) operably linked to a nucleotide sequence encoding a molecule of interest, wherein the nucleotide sequence encoding a RBS is immediately downstream from (3' from) a 10-nucleotide to 20-nucleotide region, wherein at least 80% of the nucleotides in the 10-nucleotide to 20-nucleotide region are adenine or thymine, or a combination of adenine and thymine.

In some embodiments, the molecule of interest is a therapeutic molecule, a prophylactic molecule, or a diagnostic molecule.

Some aspects of the present disclosure provide methods of expressing a molecule of interest in a Bacteroides (or Parabacteroides) bacterium, the method comprising culturing a Bacteroides (or Parabacteroides) bacterium (or a population of Bacteroides bacteria), as described herein, under conditions that result in expression of the molecule of interest.

Some aspects of the present disclosure provide methods of treating a condition in a subject, the method comprising administering to the subject a Bacteroides (or

Parabacteroides) bacterium, as described herein, wherein the molecule of interest is a therapeutic molecule. Some aspects of the present disclosure provide methods of preventing a condition in a subject, the method comprising administering to the subject a Bacteroides (or Parabacteroides) bacterium, as described herein, wherein the molecule of interest is a prophylactic molecule. Also provided herein are Bacteroides (or Parabacteroides)bact^a comprising (a) an engineered nucleic acid comprising a region containing a promoter and a nucleotide sequence encoding a ribosomal binding site (RBS) operably linked to a nucleotide sequence encoding a catalytically-inactive Cas9 nuclease, and (b) an engineered nucleic acid comprising a promoter and a nucleotide sequence encoding a RBS operably linked to a nucleotide sequence encoding a guide RNA, wherein the guide RNA targets a nucleotide sequence encoding a molecule of interest.

In some embodiments, the catalytically-inactive Cas9 nuclease is encoded by the nucleotide sequence of SEQ ID NO: 157.

Some aspects of the present disclosure provide engineered nucleic acids comprising a nucleotide sequence selected from the group consisting of SEQ ID NO: 1 - SEQ ID NO: 180.

In some embodiments, the present disclosure provides a vector comprising the genetic elements depicted in Fig. 10, including a nucleotide sequence encoding an IntNl integrase (e.g., obtained from B. uniformis), capable of facilitating integration of the vector in a variety of Bacteroides and Parabacteroides species. Thus, in some embodiments, the present disclosure provides an engineered nucleic acid comprising the nucleotide sequence of SEQ ID NO: 206, or a nucleotide sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 98% or at least 99% identity with the nucleotide sequence of SEQ ID NO: 206.

Also provided herein are cells comprising engineered nucleic acid(s), as described herein (e.g., engineered nucleic acids comprising a nucleotide sequence selected from the group consisting of SEQ ID NO: 1 - SEQ ID NO: 180).

BRIEF DESCRIPTION OF THE DRAWINGS

The accompanying drawings are not intended to be drawn to scale. For purposes of clarity, not every component may be labeled in every drawing.

Figs. 1A-1G. Genetic parts to control expression in B. thetaiotaomicron. (Fig. 1A) The ranges of gene expression are shown for the different gene regulation systems provided herein. (Fig. IB) IntN2 catalyzes stable integration of pNBU2 -based expression constructs into one of two attBT2 sites in the B. thetaiotaomicron genome. The two attBT2 sites (attBT2-l at nucleotide (nt) 6,217,227 and attBT2-2 at nt 6,138,320) are in the 3' ends of tRNA Ser genes (BT_t71 and BT_t70, respectively). (Fig. 1C) Constitutive promoters and ribosome binding sites for the construction of gene expression libraries. The putative -33 and -7 regions of the P BT II promoter, the Shine-Delgarno sequence, and the start codon are indicated by black boxes. Numbers below the black boxes represent nucleotide locations relative the PBTBII transcription start site. The 26 nt sequences introduced in the PAM promoters are shown (see also Figs. 6A-6B). Numbers at the edges of the boxes indicate the PBTBII nucleotides replaced or the insertion site within the promoter. The location of residues randomized in the rpiL* RBS library are indicated with gray arrows (for library A: nt -14, - 13, -12; for library B: nt -21, -18, -15; and for library C: nt -17, -16, -11; nt numbering is relative to the translation start site). (Fig. ID) Activity was measured for a set of constitutive promoters and their cognate RBSs. Furthermore, a set of constitutive promoters (PBTBII, PAMI, PAM2, PAM3, PAM4) was combined with RBSs of varying strengths. Gene expression was measured using a luciferase reporter (NanoLuc). (Fig. IE) Three large RBS libraries were constructed and combined with promoter PBTBII to span 10 -fold in gene expression. For reference, the parent rpiL* RBS is indicated with a black arrow. The sequences of the RBSs are provided in Table 1. For D and E, error bars represent the standard deviation of three independent biological replicates made on separate days. (Fig. IF) The strength of each RBS was compared to the predicted free energy of folding for the mRNA (AG f oi d, )- (Fig. 1G) Strong (SEQ ID NO: 210) and weak (SEQ ID NO: 211) consensus sequences for the rpiL* - 21 to -11 RBS region targeted by mutagenesis (residue locations are stated relative to the translation start site) are provided. Frequency logos were generated for the 11 strongest and 11 weakest RBSs by comparing the frequency of each nucleotide at each position in that group with the frequency of that nucleotide in that position in the full library. Position -20 and -19 were not randomized and are thus are not shown in the frequency logos.

Figs. 2A-2E. Design and characterization of genetic sensors. (Figs. 2A-2D)

Response curves for NanoLuc under the regulated control of the rhamnose- (Rha) (Fig. 2A), chondroitin sulfate- (ChS) (Fig. 2B), arabinogalactan- (AG) (Fig. 2C), or IPTG- (Fig. 2D) inducible promoters. LacOl operator sites were inserted in various regions (01, 02, 03) of the PcfxA promoter (see also Figs. 7A-7B). Inducer concentrations were applied as follows: three-fold serial dilutions starting at lOmM Rha (Fig. 2A); three-fold serial dilutions starting at 0.4% for ChS (Fig. 2B) and AG (Fig. 2C); and four-fold serial dilutions starting at 500μΜ for IPTG (Fig. 2D). The leftmost data point in each plot represents the background luminescence in the absence of inducer. Response curves were fit to a Hill function (solid lines). (Fig. 2E) Orthogonality matrix of sugar-inducible genetic systems incubated with lOmM rhamnose (Rha), 0.2% chondroitin sulfate (ChS), 0.2% arabinogalactan (AG), or lOOmM IPTG compared to no inducer. Error bars represent the standard deviation of three biological replicates made on different days. Figs. 3A-3G. Synthetic genetic memory. (Fig. 3A) Integrases mediate recombination of DNA between integrase binding sites (attB/attP), resulting in the inversion of the intervening spacers. (Fig. 3B) Schematic of the location of the promoter-RBS -integrase system and the memory array cassettes in the B. thetaiotaomicron chromosome. (Fig. 3C) Integrase-mediated DNA inversion at each integrase target sequence in the memory array cassette was detected by polymerase chain reaction (PCR). Primer pairs (arrows) anneals to the interface of the integrase recognition sites and to the spacer region between recognition sites. PCR amplification occurs only after an inversion event (solid lines below the primer arrows indicate expected amplicons). (Fig. 3D) Representative PCR products are shown after recombination. - indicates no integrase, + indicates the integrase is present. PAM4-rpiL* was used to control expression of each integrase. (Fig. 3E) Schematic of the rhamnose-inducible recombinase circuit. Transcriptional activator RhaR, produced from the endogenous locus, is activated in the presence of rhamnose causing expression of Intl2 from P r h a . Intl2 mediates recombination between the Intl2 attB and attP recognition sequences. (Fig. 3F) Response curve of Intl2 memory circuit. Intl2 was placed under the control of a subset of P3763- rpiL*C51. Inducer concentrations were nine-fold serial dilutions starting at 10 mM rhamnose. The leftmost data point represents the recombination in the absence of inducer. Cells were grown 8 hours at 37 °C before harvesting cells and isolating DNA. qPCR was used to measure the fold-change in flipping relative to the 10 mM rhamnose sample using the Intl2 gene for reference. Data were fit with a Hill function to guide the eye. (Fig. 3G) Intl2- mediated recombination versus time. Cells were induced with 10 mM rhamnose at t=0. qPCR was used to measure the fold-change in flipping relative to the t=8 sample using the Intl2 gene for reference. For Figs. 3F-G, error bars represent the standard deviation of three biological replicates made on different days.

Figs. 4A-4F. CRISPRi-mediated repression of recombinant and endogenous genes.

(Fig. 4A) Schematic of dCas9-based repression of NanoLuc. LacI Q is expressed from P BT II and represses transcription from the PLac023 promoter. Addition of IPTG inactivates LacI Q to allow expression of dCas9 from PLac023. dCas9 complexes with guide RNA (sgRNA) constitutively expressed from the Pi promoter to prevent the transcription of NanoLuc from the PcfiA promoter. Guide RNAs were designed to target the coding sequence of NanoLuc (NL1-4) or the P C fiA promoter (PR1-2). (Fig. 4B) Response curves of dCas9-mediated targeting the coding sequence of NanoLuc (NL1-4), the promoter (PR1-2) or a nonsense sequence (NS). Fourfold serial dilutions of IPTG starting at 500 μΜ or no inducer were added to cultures. Response curves were fit to a Hill Function (solid lines). (Fig. 4C) Fold repression elicited by various gRNAs in the presence (500 μΜ) of inducer. Bars are colored to correspond to part B. (Fig. 4D) Genomic location of endogenous genes targeted using CRISPRi. (Fig. 4E) Minimum inhibitory concentrations (MICs) of polymyxin B for cells with CRISPRi targeted against BT1854 (dCas9 BT i85 4 ) compared with wild-type (WT) cells or non-specific control cells (dCas9 s)- Reported values are the mode of three independent biological replicates made on three separate days. (Fig. 4F) CRISPRi was targeted against BT1754 (dCas9 BT i75 4 ). Growth curves of wild-type (WT) (black), dCas9 BT i75 4 (pink) or dCas9NS (gray) cells in minimal media supplemented with 0.5% glucose (MM-Glc) or 0.5% fructose (MM-Fru) in the presence (full line) or absence (dotted line) of lOOmM IPTG . Error bars represent the standard deviation of three biological replicates made on different days.

Figs. 5A-5D. In vivo function of genetic parts within B. thetaiotaomicron colonizing the mouse gut. (Fig. 5A) Experimental timeline. Specific pathogen free (SPF) Swiss Webster mice were treated for 10 days with ciprofloxacin and metronidazole and gavaged with B. thetaiotaomicron 2 days after cessation of treatment. (Figs. 5B-5C) Luciferase activity in fecal pellets of mice inoculated with strains possessing the arabinogalactan (AG) inducible P 02 68 (Fig. 5B) or IPTG-inducible CRISPRi dCas9 N L3 (Fig. 5C) systems. Mice were provided drinking water supplemented with 5% arabinogalactan (Fig. 5B: solid line), or 25 mM IPTG (Fig. 5C: solid line) after stool collection on Day 2 (grey box), or were maintained on normal drinking water throughout the entire experiment (dashed lines). Inducer water was removed on Day 4 after stool collection. Grey boxes indicate the period of time that mice were exposed to inducer- supplemented drinking water. Luminescence values were normalized to cell density as determined by qPCR using NanoLuc-specific primers. (Fig. 5D) SPF mice were colonized with B. thetaiotaomicron containing the rhamnose-inducible integrase construct P3763-rpiL*C51-Intl2. All mice were exposed to 0.3% rhamnose (w/w) in the plant-based chow. In addition, half of the mice had their drinking water supplemented with 500 mM rhamnose after stool collection on Day 1 ("Chow + Rha", solid line) while the other half of the mice were maintained on normal drinking water throughout the entire experiment ("Chow", dashed line). Mice receiving rhamnose-supplemented water on Days 1 and 2 (grey box) were returned to normal water on Day 3 after stool collection. Absolute quantities of flipped and unflipped memory array in fecal DNA were determined by qPCR using standard curves (Experimental Procedures). Recombination frequency is expressed as the ratio of cells containing a flipped memory array (Flipped) divided by the sum total of cells containing a flipped or unflipped array (Total). For day 3 "Chow" samples, n=3. For all other days, n=6 for both treatment groups. For Figs. 5B-D, individual points represent independent biological replicates and the line represents the mean of the group. *P<0.05; **P<0.01.

Figs. 6A-6B. PAM promoter sequences and induction with fucose. (Fig. 6A) Promoters PAM1, PAM2, PAM3, and PAM4 were constructed by introducing a 26 bp sequence (gray) at 4 locations in the constitutive BT1311 promoter (PBT1311). Predicted - 33, -7, and +1 sites of the PBT1311 promoter are shown in bold. Fig. 6A depicts SEQ ID NOs: 212 to 216 from top to bottom, respectively. (Fig. 6B) Activity of promoters PAM1, PAM2, PAM3, and PAM4 were measured in the presence (filled bars) or absence (open bars) of fucose (10 mM). Error bars represent the standard deviation of three biological replicates made on three different days (n=3).

Figs. 7A-7B. Synthetic IPTG-inducible promoters. (Fig. 7A) Synthetic IPTG- inducible promoters were constructed by placing LacOl operator sites (red) upstream of the - 33 element (01), between the -33 and -7 elements (02) and/or directly downstream of the transcription start site (03) of the strong P C f X A promoter. Predicted -33, -7 and +1 sites are shown in bold. These promoters are regulated by the E. coli LacIQ repressor expressed from PBT1311. Fig. 7A depicts SEQ ID NOs: 217 to 220 from top to bottom, respectively. (Fig. 7B) Response curves for the synthetic IPTG-inducible systems. Cells were incubated with no inducer or four-fold serial dilutions of IPTG starting at 500 μΜ. Data sets for PLacO and PL c023 were fit to a Hill function (solid line). Error bars represent the standard deviation of three biological replicates made on three different days (n=3).

Figs. 8A-8B. Integrase characterization. (Fig. 8A) Representative PCR products are shown for wild-type (unflipped) memory array at each integrase recognition sequence. "-" indicates no integrase, "+" indicates the integrase is present. PAM4-rpiL* was used to control expression of each integrase. (Fig. 8B) Cell growth of the P3763-rpiL*C51-Intl2 strain is shown as optical density (OD) at 600 nm as a function of rhamnose concentration. Inducer concentrations were three-fold serial dilutions starting at 10 mM rhamnose. The leftmost data point represents the recombination in the absence of inducer. Cells were grown 8 hours at 37°C before measuring the OD600 value for each culture. Error bars represent the standard deviation of three biological replicates made on three different days (n=3).

Figs. 9A-9C. Colonization of the mouse gut with engineered B. thetaiotaomicron strains. (Figs. 9A-9B) Cell densities of the arabinogalactan-inducible P0268 (Fig. 9A) or the dCas9N L 3 (Fig. 9B) strains in the fecal pellets of inoculated mice. 5% arabinogalactan (Fig. 9A: solid line) or 25 mM IPTG (Fig. 9B: solid line) was added to the drinking water of mice on Day 2 after stool collection (solid lines) and mice were returned to normal water on Day 4 after stool collection. The control groups (dashed lines) remained on normal water for the duration of the experiment. Grey boxes indicate the period of time over which mice were exposed to inducer in their drinking water. Bacterial loads were quantified by analyzing DNA extracted from fecal pellet using qPCR. The number of cells was determined using

NanoLuc- specific primers and a standard curve generated with purified NanoLuc amplicons. Results were normalized to the weight of fecal material analyzed. (Fig. 9C) Bacterial load of the rhamnose-inducible integrase strain in the fecal pellets of inoculated mice. All mice were exposed to 0.3% rhamnose (w/w) in the plant-based chow. Rhamnose supplemented drinking water was provided to half of the mice ("Chow + Rha", solid line) on Day 1 after stool collection and normal water was returned on Day 3 after stool collection (grey box). The other half of the mice ("Chow", dashed line) remained on normal water for the duration of the experiment. Cell density was calculated as the sum of flipped and unflipped (wild-type) memory array as determined by qPCR on DNA isolated from fecal samples. Results were normalized to the weight of fecal material analyzed. For day 3 "Chow" samples, n=3. For all other days, n=6 for both treatment groups. For A-C, individual points represent independent biological replicates and the line represents the mean of the group.

Fig. 10 shows a plasmid map of pNBUl, which includes a NBU1 integrase for insertion into a single site in a Bacteroides chromosome.

Fig. 11 is a graph showing that pNBUl may be used to target, for example, B.

thetaiotaomicron, B.fragilis, B. ovatus, B. vulgatus, B. caccae, B. eggerthii, B. vulgatus and Parabacteroides distasonis.

DETAILED DESCRIPTION OF THE INVENTION

To date, multiple microorganisms have served as chassis for engineered microbial therapies of human disease. However, compared to organisms such as E. coli and L. lactis, which undergo depletion or clearance within days of administration, Bacteroides populations exhibit low variation in abundance and long-term colonization. Nonetheless, few genetic parts and inducible systems are available for B. thetaiotaomicron, for example, and its relatives due, in part, to unique promoter and RBS architectures in Bacteroides, which have precluded the direct incorporation of genetic systems developed in other organisms. For example, unlike most other prokaryotes, the unique major sigma factor in Bacteroides binds to a -33/- 7 consensus sequence (TTTG/TAnnTTTG), the strength of translation initiation is poorly correlated with the level of ribosome binding site (RBS) complementarity to the 16S rRNA of the host organism, and compared to the E. coli RBS, Bacteroides RBS strength is more sensitive to secondary structures, depleted in GC content, and predicted to rely more heavily on interactions with ribosomal protein SI. Further, promoter and RBS

characterization have employed several reporter outputs, preventing direct comparison of parts. A lack of genetic part libraries hinders the introduction of multi-gene pathways, such as those that could produce a metabolic product designed to treat disease.

The present disclosure provides, in some aspects, a set of genetic tools for precise and robust engineering of Bacteroides {e.g., B. thetaiotomicron) or Parabacteroides for microbiome applications (as well as other applications). Provided herein is a library of biological parts, comprised of constitutive promoters, inducible promoters, and ribosomal binding sites (RBSs) that each span output dynamic ranges over several orders of magnitude (Fig. 1A). Constitutive promoters and RBSs were used to characterize the input expression levels required to generate recombinase-based DNA-encoded memory in B.

thetaiotaomicron, for example. Externally switchable DNA-based memory devices were then constructed by integrating inducible promoters with recombinases. Additionally, inducible promoters were used to control CRISPRi-based regulation of synthetic and endogenous genes. Finally, multiple of regulatory tools provided herein were integrated together and their proper in vivo function validated within B. thetaiotomicron that colonized the gut of mice. With this toolbox of genetic parts, Bacteroides (e.g., B. thetaiotaomicron) or Parabacteroides can be used as a platform for predictable gene expression and circuit design for microbiome engineering.

Bacteroides and Parabacteroides

Bacteroides is a genus of Gram-negative, non-spore-forming, anaerobic, and rod- shaped bacteria. They have an outer membrane, a peptidoglycan layer, and a cytoplasmic membrane. The main by-products of their anaerobic respiration are acetic acid, isovaleric acid, propionic acid and succinic acid. They are involved in many important metabolic activities in the human colon including fermentation of carbohydrates, utilization of nitrogenous substances, and biotransformation of bile acids and other steroids. Most intestinal bacteria are saccharolytic, which means that they obtain carbon and energy by hydrolysis of carbohydrate molecules.

The genomes of the circular chromosomes of many Bacteroides species and strains have been studied; research is being done on sequencing Bacteroides species in order to understand their pathogenic properties. All Bacteroides have G-C composition of 40-48%. Much of the genome is controlled by sigma factors which respond to environmental factors. There have been a total of three genome projects done on two different species of

Bacteroides. The three genomes sequenced were that of Bacteroides thetaiotaomicron VPI- 5482, Bacteroides fragilis YCH46, and Bacteroides fragilis NCTC 9343. Information and a schematic representation of the Bacteroides thetaiotaomicron VPI-5482 chromosome can be found at National Center for Biotechnology Information (NCBI).

Engineered nucleic acids of the present disclosure may be introduced into a variety of different organisms, including Bacteroides. Examples of species of Bacteroides

contemplated herein include, without limitation, B. acidifaciens, B. caccae, B. distasonis, B. gracilis, B. fragilis, B. dorei, B. oris, B. ovatus, B. putredinis, B. pyogenes, B. stercoris, B. suis, B. tectus, B. thetaiotaomicron, B. vulgatus, B. eggerthii, B. merdae, B. stercoris, and B. uniformis.

Engineered nucleic acids of the present disclosure may also be introduced into Parabacteroides (Sakamoto M and Benno Y. Int J Syst Evol Microbiol. 2006 Jul;56(Pt 7): 1599-605, incorporated by reference), which is closely related to Bacteroides. Examples of species of Parbacteroides contemplated herein include, without limitation, P. chartae, P. chinchilla, P. distasonis, P. faecis, P. goldsteinii, P. gordonii, P. johnsonii, and P. merdae.

Engineered Nucleic Acids

A "nucleic acid" is at least two nucleotides covalently linked together, and in some instances, may contain phosphodiester bonds {e.g. , a phosphodiester "backbone"). An "engineered nucleic acid" is a nucleic acid that does not occur in nature. It should be understood, however, that while an engineered nucleic acid as a whole is not naturally- occurring, it may include nucleotide sequences that occur in nature. In some embodiments, an engineered nucleic acid comprises nucleotide sequences from different organisms {e.g., from different species). For example, in some embodiments, an engineered nucleic acid includes a murine nucleotide sequence, a bacterial nucleotide sequence, a human nucleotide sequence, and/or a viral nucleotide sequence. Engineered nucleic acids include recombinant nucleic acids and synthetic nucleic acids. A "recombinant nucleic acid" is a molecule that is constructed by joining nucleic acids {e.g., isolated nucleic acids, synthetic nucleic acids or a combination thereof) and, in some embodiments, can replicate in a living cell. A "synthetic nucleic acid" is a molecule that is amplified or chemically, or by other means, synthesized. A synthetic nucleic acid includes those that are chemically modified, or otherwise modified, but can base pair with naturally-occurring nucleic acid molecules. Recombinant and synthetic nucleic acids also include those molecules that result from the replication of either of the foregoing.

In some embodiments, a nucleic acid of the present disclosure is considered to be a nucleic acid analog, which may contain, at least in part, other backbones comprising, for example, phosphoramide, phosphorothioate, phosphorodithioate, O-methylphophoroamidite linkages and/or peptide nucleic acids. A nucleic acid may be single-stranded (ss) or double- stranded (ds), as specified, or may contain portions of both single-stranded and double- stranded sequence. In some embodiments, a nucleic acid may contain portions of triple- stranded sequence. A nucleic acid may be DNA, both genomic and/or cDNA, RNA or a hybrid, where the nucleic acid contains any combination of deoxyribonucleotides and ribonucleotides (e.g., artificial or natural), and any combination of bases, including uracil, adenine, thymine, cytosine, guanine, inosine, xanthine, hypoxanthine, isocytosine and isoguanine.

Nucleic acids of the present disclosure may include one or more genetic elements. A "genetic element" refers to a particular nucleotide sequence that has a role in nucleic acid expression (e.g., promoter, enhancer, terminator) or encodes a discrete product of an engineered nucleic acid (e.g., a nucleotide sequence encoding a guide RNA, a protein and/or an RNA interference molecule).

Nucleic acids of the present disclosure may be produced using standard molecular biology methods (see, e.g., Green and Sambrook, Molecular Cloning, A Laboratory Manual, 2012, Cold Spring Harbor Press).

In some embodiments, nucleic acids are produced using GIBSON ASSEMBLY® Cloning (see, e.g., Gibson, D.G. et al. Nature Methods, 343-345, 2009; and Gibson, D.G. et al. Nature Methods, 901-903, 2010, each of which is incorporated by reference herein). GIBSON ASSEMBLY® typically uses three enzymatic activities in a single-tube reaction: 5' exonuclease, the Ύ extension activity of a DNA polymerase and DNA ligase activity. The 5' exonuclease activity chews back the 5' end sequences and exposes the complementary sequence for annealing. The polymerase activity then fills in the gaps on the annealed regions. A DNA ligase then seals the nick and covalently links the DNA fragments together. The overlapping sequence of adjoining fragments is much longer than those used in Golden Gate Assembly, and therefore results in a higher percentage of correct assemblies.

In some embodiments, a compressed biosynthetic pathway is delivered to a cell on a vector. A "vector" refers to a nucleic acid (e.g., DNA) used as a vehicle to artificially carry genetic material (e.g., an engineered nucleic acid) into a cell where, for example, it can be replicated and/or expressed. In some embodiments, a vector is an episomal vector (see, e.g., Van Craenenbroeck K. et al. Eur. J. Biochem. 261 , 5665, 2000, incorporated by reference herein). A non-limiting example of a vector is a plasmid (e.g., Fig. 3). Plasmids are double- stranded generally circular DNA sequences that are capable of automatically replicating in a host cell. Plasmid vectors typically contain an origin of replication that allows for semi- independent replication of the plasmid in the host and also the transgene insert. Plasmids may have more features, including, for example, a "multiple cloning site," which includes nucleotide overhangs for insertion of a nucleic acid insert, and multiple restriction enzyme consensus sites to either side of the insert. Another non-limiting example of a vector is a viral vector.

Genetic Elements

Expression of engineered nucleic acids is driven by a promoter operably linked to a nucleic acid containing, for example, a nucleic acid encoding a molecule of interest. A "promoter" refers to a control region of a nucleic acid sequence at which initiation and rate of transcription of the remainder of a nucleic acid sequence are controlled. A promoter drives expression or drives transcription of the nucleic acid sequence that it regulates. A promoter may also contain sub-regions at which regulatory proteins and molecules may bind, such as RNA polymerase and other transcription factors. Promoters may be constitutive, inducible, activatable, repressible, tissue-specific or any combination thereof.

Herein, a promoter is considered to be "operably linked" when it is in a correct functional location and orientation in relation to a nucleic acid sequence it regulates to control ("drive") transcriptional initiation and/or expression of that sequence.

A promoter may be one naturally associated with a gene or sequence, as may be obtained by isolating the 5' non-coding sequences located upstream of the coding segment of a given gene or sequence. Such a promoter can be referred to as "endogenous."

In some embodiments, a coding nucleic acid sequence may be positioned under the control of a recombinant or heterologous promoter, which refers to a promoter that is not normally associated with the encoded sequence in its natural environment. Such promoters may include promoters of other genes; promoters isolated from any other cell; and synthetic promoters or enhancers that are not "naturally occurring" such as, for example, those that contain different elements of different transcriptional regulatory regions and/or mutations that alter expression through methods of genetic engineering that are known in the art. In addition to producing nucleic acid sequences of promoters and enhancers synthetically, sequences may be produced using recombinant cloning and/or nucleic acid amplification technology, including polymerase chain reaction (PCR) (see U.S. Pat. No. 4,683,202 and U.S. Pat. No. 5,928,906).

In some embodiments, a promoter is an "inducible promoter," which refer to a promoter that is characterized by regulating (e.g., initiating or activating) transcriptional activity when in the presence of, influenced by or contacted by an inducer signal. An inducer signal may be endogenous or a normally exogenous condition (e.g., light), compound (e.g., chemical or non-chemical compound) or protein that contacts an inducible promoter in such a way as to be active in regulating transcriptional activity from the inducible promoter. Thus, a "signal that regulates transcription" of a nucleic acid refers to an inducer signal that acts on an inducible promoter. A signal that regulates transcription may activate or inactivate transcription, depending on the regulatory system used. Activation of transcription may involve directly acting on a promoter to drive transcription or indirectly acting on a promoter by inactivation a repressor that is preventing the promoter from driving transcription.

Conversely, deactivation of transcription may involve directly acting on a promoter to prevent transcription or indirectly acting on a promoter by activating a repressor that then acts on the promoter.

The administration or removal of an inducer signal results in a switch between activation and inactivation of the transcription of the operably linked nucleic acid sequence. Thus, the active state of a promoter operably linked to a nucleic acid sequence refers to the state when the promoter is actively regulating transcription of the nucleic acid sequence (i.e., the linked nucleic acid sequence is expressed). Conversely, the inactive state of a promoter operably linked to a nucleic acid sequence refers to the state when the promoter is not actively regulating transcription of the nucleic acid sequence (i.e., the linked nucleic acid sequence is not expressed).

An inducible promoter of the present disclosure may be induced by (or repressed by) one or more physiological condition(s), such as changes in light, pH, temperature, radiation, osmotic pressure, saline gradients, cell surface binding, and the concentration of one or more extrinsic or intrinsic inducing agent(s). An extrinsic inducer signal or inducing agent may comprise, without limitation, amino acids and amino acid analogs, saccharides and polysaccharides, nucleic acids, protein transcriptional activators and repressors, cytokines, toxins, petroleum-based compounds, metal containing compounds, salts, ions, enzyme substrate analogs, hormones or combinations thereof. Inducible promoters of the present disclosure include any inducible promoter described herein or known to one of ordinary skill in the art. Examples of inducible promoters include, without limitation, chemically/biochemically-regulated and physically- regulated promoters such as alcohol-regulated promoters, tetracycline-regulated promoters (e.g., anhydrotetracycline (aTc)-responsive promoters and other tetracycline -responsive promoter systems, which include a tetracycline repressor protein (tetR), a tetracycline operator sequence (tetO) and a tetracycline transactivator fusion protein (tTA)), steroid- regulated promoters (e.g. , promoters based on the rat glucocorticoid receptor, human estrogen receptor, moth ecdysone receptors, and promoters from the steroid/retinoid/thyroid receptor superfamily), metal-regulated promoters (e.g., promoters derived from metallothionein (proteins that bind and sequester metal ions) genes from yeast, mouse and human), pathogenesis-regulated promoters (e.g., induced by salicylic acid, ethylene or

benzothiadiazole (BTH)), temperature/heat- inducible promoters (e.g., heat shock promoters), and light-regulated promoters (e.g., light responsive promoters from plant cells).

In some embodiments, an inducer signal of the present disclosure is an N-acyl homoserine lactone (AHL), which is a class of signaling molecules involved in bacterial quorum sensing. Quorum sensing is a method of communication between bacteria that enables the coordination of group based behavior based on population density. AHL can diffuse across cell membranes and is stable in growth media over a range of pH values. AHL can bind to transcriptional activators such as LuxR and stimulate transcription from cognate promoters.

In some embodiments, an inducer signal of the present disclosure is

anhydrotetracycline (aTc), which is a derivative of tetracycline that exhibits no antibiotic activity and is designed for use with tetracycline-controlled gene expression systems, for example, in bacteria.

In some embodiments, an inducer signal of the present disclosure is isopropyl β-D-l- thiogalactopyranoside (IPTG), which is a molecular mimic of allolactose, a lactose metabolite that triggers transcription of the lac operon, and it is therefore used to induce protein expression where the gene is under the control of the lac operator. IPTG binds to the lac repressor and releases the tetrameric repressor from the lac operator in an allosteric manner, thereby allowing the transcription of genes in the lac operon, such as the gene coding for beta-galactosidase, a hydrolase enzyme that catalyzes the hydrolysis of β-galactosides into monosaccharides. The sulfur (S) atom creates a chemical bond which is non-hydrolyzable by the cell, preventing the cell from metabolizing or degrading the inducer. IPTG is an effective inducer of protein expression, for example, in the concentration range of 100 μΜ to 1.0 mM. Concentration used depends on the strength of induction required, as well as the genotype of cells or plasmid used. If laclq, a mutant that over-produces the lac repressor, is present, then a higher concentration of IPTG may be necessary. In blue- white screen, IPTG is used together with X-gal. Blue- white screen allows colonies that have been transformed with the recombinant plasmid rather than a non-recombinant one to be identified in cloning experiments.

Other inducible promoter systems are known in the art and may be used in accordance with the present disclosure.

In some embodiments, inducible promoters of the present disclosure function in prokaryotic cells (e.g., bacterial cells). Examples of inducible promoters for use prokaryotic cells include, without limitation, bacteriophage promoters (e.g. Pis Icon, T3, T7, SP6, PL) and bacterial promoters (e.g., Pbad, PmgrB, Ptrc2, Plac/ara, Ptac, Pm), or hybrids thereof (e.g. PLlacO, PLtetO). Examples of bacterial promoters for use in accordance with the present disclosure include, without limitation, positively regulated E. coli promoters such as positively regulated σ70 promoters (e.g., inducible pBad/araC promoter, Lux cassette right promoter, modified lamdba Prm promote, plac Or2-62 (positive), pBad/AraC with extra REN sites, pBad, P(Las) TetO, P(Las) CIO, P(Rhl), Pu, FecA, pRE, cadC, hns, pLas, pLux), oS promoters (e.g., Pdps), σ32 promoters (e.g., heat shock) and σ54 promoters (e.g., glnAp2); negatively regulated E. coli promoters such as negatively regulated σ70 promoters (e.g., Promoter (PRM+), modified lamdba Prm promoter, TetR - TetR-4C P(Las) TetO, P(Las) CIO, P(Lac) IQ, RecA_DlexO_DLac01, dapAp, FecA, Pspac-hy, pel, plux-cl, plux-lac, CinR, CinL, glucose controlled, modified Pr, modified Prm+, FecA, Pcya, rec A (SOS), Rec A (SOS), EmrR_regulated, Betl_regulated, pLac_lux, pTet_Lac, pLac/Mnt, pTet/Mnt, LsrA/cI, pLux/cI, Lacl, LacIQ, pLacIQl, pLas/cI, pLas/Lux, pLux/Las, pRecA with LexA binding site, reverse BBa_R0011, pLacI/ara-1, pLacIq, rrnB PI, cadC, hns, PfhuA, pBad/araC, nhaA, OmpF, RcnR), oS promoters (e.g., Lutz-Bujard LacO with alternative sigma factor σ38), σ32 promoters (e.g., Lutz-Bujard LacO with alternative sigma factor σ32), and σ54 promoters (e.g., glnAp2); negatively regulated B. subtilis promoters such as repressible B. subtilis σΑ promoters (e.g., Gram-positive IPTG-inducible, Xyl, hyper-spank) and σΒ promoters. Other inducible microbial promoters may be used in accordance with the present disclosure.

A "ribosomal binding site (RBS)" is a sequence on mRNA that is bound by the ribosome when initiating protein translation. The ribosome searches for this site and binds to it through base-pairing of nucleotides. Once the ribosome has bound, it recruits initiation factors and begins the translation process. Bacteroides possess a unique RBS where homology to the 16S rRNA does not play a role in the strength of translation initiation.

The present disclosure contemplates a variety of RBSs including, without limitation, those listed in Table 2.

Recombinases

A "recombinase," as used herein, is a site-specific enzyme that recognizes short DNA sequence(s), which sequence(s) are typically between about 30 base pairs (bp) and 40 bp, and that mediates the recombination between these recombinase recognition sequences, which results in the excision, integration, inversion, or exchange of DNA fragments between the recombinase recognition sequences. For example, in some embodiments, Bacteroides cells of the present disclosure may be engineered to comprise at least two engineered nucleic acids, comprising a region containing a promoter and a nucleotide sequence encoding a ribosomal binding site (RBS) operably linked to a nucleotide sequence encoding a recombinase, and the other comprising a promoter and a nucleotide sequence encoding a RBS operably linked to a nucleotide sequence encoding a molecule of interest, wherein the nucleotide sequence encoding the molecule of interest is flanked by a pair of cognate recombinase recognition sequences. In such embodiments, expression of the molecule of interest is regulated by recombinase activity, or inactivity, of the other circuit.

Recombinases can be classified into two distinct families: serine recombinases (also referred to herein as serine integrases) and tyrosine recombinases (also referred to herein as tyrosine integrases), based on distinct biochemical properties. Serine recombinases and tyrosine recombinases are further divided into bidirectional recombinases and unidirectional recombinases. Examples of bidirectional serine recombinases for use herein include, without limitation, β-six, CinH, ParA and γδ; and examples of unidirectional serine recombinases include, without limitation, Int7, Int8, Int9, Intl2, Bxbl, (|)C31, TP901, TGI, φΒΤΙ, R4, (pRVl, (pFCl, MRU, A118, U153 and gp29. Examples of bidirectional tyrosine

recombinases for use herein include, without limitation, Cre, FLP, and R; and unidirectional tyrosine recombinases include, without limitation, Lambda, HK101, HK022 and pSAM2.

The serine and tyrosine recombinase names stem from the conserved nucleophilic amino acid residue that the recombinase uses to attack the DNA and which becomes covalently linked to the DNA during strand exchange. The outcome of recombination depends, in part, on the location and orientation of two short repeated DNA sequences that are to be recombined, typically less than 30 bp long. Recombinases bind to these repeated sequences, which are specific to each recombinase, and are herein referred to as "recombinase recognition sequences." Thus, as used herein, a recombinase is "specific for" a recombinase recognition sequence when the recombinase can mediate inversion or excision between the repeated nucleotide sequences. As used herein, a recombinase may also be said to recognize its "cognate recombinase recognition sequences," which flank an intervening genetic element (e.g., promoter, terminator, or nucleotide sequence encoding the molecule of interest). A genetic element is said to be "flanked" by recombinase recognition sites when the element is located between and immediately adjacent to two repeated nucleotide sequences.

Recombinases can also be classified as irreversible or reversible. As used herein, an "irreversible recombinase" refers to a recombinase that can catalyze recombination between two complementary recombination sites, but cannot catalyze recombination between the hybrid sites that are formed by this recombination without the assistance of an additional factor. Thus, an "irreversible recognition site" refers to a recombinase recognition site that can serve as the first of two nucleotide recognition sequences for an irreversible recombinase and that is modified to a hybrid recognition site following recombination at that site. A "complementary irreversible recognition site" refers to a recombinase recognition site that can serve as the second of two nucleotide recognition sequences for an irreversible recombinase and that is modified to a hybrid recombination site following homologous recombination at that site.

Irreversible recombinases, and nucleic acids that encode the irreversible

recombinases, are described in the art and can be obtained using routine methods. Examples of irreversible recombinases include, without limitation, phiC31 (cpC31) recombinase, coliphage P4 recombinase (Ow & Ausubel, J. Bacteriol. 155, 704-713 (1983)), coliphage lambda integrase (Lorbach et al, J. Mol. Biol., 296, 1175-81 (2000)), Listeria Al 18 phage recombinase (Loessner et al., Mol. Micro. 35, 324-340 (2000)), and actinophage R4 Sre recombinase (Matsuura et al, J Bacteriol. 178, 3374-3376 (1996)), HKlOl, HK022, pSAM2, Bxbl, TP901, TGI, φΒΤΙ, cpRVl, cpFCl, MRU, U153 and gp29.

Conversely, a "reversible recombinase" refers to a recombinase that can catalyze recombination between two complementary recombinase recognition sites and, without the assistance of an additional factor, can catalyze recombination between the sites that are formed by the initial recombination event, thereby reversing it. The product-sites generated by recombination are themselves substrates for subsequent recombination. Examples of reversible recombinase systems include, without limitation, the Cre-lox and the Flp-frt systems, R, β-six, CinH, Par A and γδ.

In some embodiments, the recombinase is serine recombinase. Thus, in some embodiments, the recombinase is considered to be irreversible. In some embodiments, the recombinase is a tyrosine recombinase. Thus, in some embodiments, the recombinase is considered to be reversible.

The recombinases provided herein are not meant to be exclusive examples of recombinases that can be used in embodiments of the present disclosure. The complexity of the engineered nucleic acids of the present disclosure can be expanded by mining databases for new orthogonal recombinases or designing synthetic recombinases with defined DNA specificities (Groth, A. C. & Calos, M. P. J Mol Biol 335, 667-678, (2004); Gordley, R. M., et al. Proc Natl Acad Sci USA 106, 5053-5058 (2009)). Other examples of recombinases that are useful in the engineered nucleic acids described herein are known to those of skill in the art, and any new recombinase that is discovered or generated is expected to be able to be used in the different embodiments of the present disclosure.

Therapeutic, Prophylactic and Diagnostic Molecules

The tools provided herein may be used to express, inhibit expression of, or reduce expression of a molecule of interest {e.g., a gene or protein of interest). A molecule, herein, may be, for example, any molecule that can be used to provide benefit to a subject (including without limitation prophylactic or therapeutic benefit) or that can be used for diagnosis and/or detection (for example, imaging) in vitro or in vivo.

In some embodiments, a "nucleotide sequence encoding a molecule of interest" is a nucleotide sequence encoding a protein of interest. Proteins of interest include, for example, antibodies, single chain antibodies, antibody fragments, enzymes, co-factors, receptors, ligands, transcription factors and other regulatory factors, antigens, cytokines and

chemokines.

Aspects of the present disclosure provide methods of treating a condition in a subject {e.g., a human subject) comprising administering to a subject a Bacteroides bacterium, as described herein. In some embodiments, the Bacteroides bacterium comprises (a) an engineered nucleic acid comprising a region containing a promoter and a nucleotide sequence encoding a ribosomal binding site (RBS) operably linked to a nucleotide sequence encoding molecule of interest, such as a therapeutic or prophylactic molecule of interest. CRISPR Interference

Aspects of the present disclosure provide cells (e.g., Bacteroides bacteria) that comprise (a) an engineered nucleic acid comprising a region containing a promoter and a nucleotide sequence encoding a ribosomal binding site (RBS) operably linked to a nucleotide sequence encoding a catalytically-inactive Cas9 nuclease, and (b) an engineered nucleic acid comprising a promoter and a nucleotide sequence encoding a RBS operably linked to a nucleotide sequence encoding a guide RNA, wherein the guide RNA targets a nucleotide sequence encoding a molecule of interest.

CRISPR interference (CRISPRi) is a genetic perturbation technique that permits sequence-specific repression or activation of gene expression. The technique uses catalytically-inactive Cas9 (also referred to as dead Cas9 or dCas9) lacking endonuclease activity to regulate genes in an RNA-guided manner. Targeting specificity is determined by complementary base-pairing of a single guide RNA (sgRNA) to genomic loci, for example. CRISPRi relies on the generation of catalytically inactive Cas9. This is accomplished, for example, by introducing point mutations in the two catalytic residues (D10A and H840A) of the gene encoding Cas9. In doing so, dCas9 is unable to cleave dsDNA but retains the ability to target DNA. Taken together sgRNA and dCas9 provide a minimum system for gene- specific regulation in any organism.

In some embodiments, CRISPRi, as provided herein, is used to inhibit or reduce (e.g., by greater than 10%, such as 20% to 98%, or 50% to 90%) transcription of a molecule of interest (e.g., an endogenous gene of interest) in, for example, a Bacteroides bacterium.

Applications

The present disclosure provides, inter alia, a versatile set of genetic technologies for the manipulation of, for example, the abundant gut symbiont Bacteroides (e.g., B.

thetaiotaomicron), expanding on the number and expression range of genetic parts previously available for Bacteroidetes (range: 10 ) and achieving ranges of expression similar to those of libraries characterized for other gut-associated bacteria, including E. coli (range: 10 4 -10 5 ) and lactic acid bacteria (range: 10 ).

For microbiome engineering applications, the ability to precisely modulate gene expression in commensal organisms may enable functional studies of the microbiome, non- invasive monitoring of in vivo environments, and long-term targeted therapeutics. For example, the constitutive and inducible systems, integrases, and CRISPRi regulators, as provided herein, may be integrated for higher-order computation in B. thetaiotaomicron. These engineered commensals may be used to map the dose-dependent and temporal effects of specific surface polysaccharides or heterologous pathways on colonization and

maintenance of the gut microbiota and on host health. Higher-order combinations of inducible promoters linked with integrases may achieve Boolean logic with embedded cellular memory, enabling surveillance of the gut environment. Furthermore, environmental sensing coupled with precision expression control of heterologous pathways in B.

thetaiotaomicron may be exploited, in some embodiments, for on-demand, localized delivery of therapeutic molecules. The present disclosure also shows that the CRISPRi system can be used to dynamically manipulate bacterial processes in Bacteroides (e.g., B. thetaiotaomicron) by targeting endogenous genes. dCas9-mediated repression may be induced, for example, in a commensal library of Bacteroides (e.g., B. thetaiotaomicron) harboring distinct guide RNAs to identify genes required for Bacteroides (e.g., B. thetaiotaomicron) maintenance or interspecies interactions, for example. With these genetic resources, Bacteroides (e.g., B. thetaiotaomicron) is a useful platform for cellular sensing, computation and actuation at the host-microbe interface in the gut.

EXAMPLES

Example 1. Landing pads for genetic part and device characterization

All genetic parts in this study were characterized using the integration vector pNBU2 to ensure genetic stability of the constructs (Fig. IB). The pNBU2 plasmid encodes the intN2 tyrosine integrase, which mediates sequence-specific recombination between the attN site of pNBU2 and one of two attBT sites located in the 3' ends of the two tRNA Ser genes, BT_t70 and BT_t71, on the B. thetaiotaomicron chromosome (Fig. IB). Insertion of the pNBU2 plasmid inactivates the tRNA Ser gene, and simultaneous insertion into both BT_t70 and BT_t71 is unlikely due to the essentiality of tRNA Ser . pNBU2-based vectors have been used for single-copy complementation in B. thetaiotaomicron in in vitro studies (Koropatkin NM et al. Nat. Rev. Microbiol. 10:323-35, 2012) and in vivo mouse models (Martens EC et al. Cell Host Microbe 4:447-57, 2008).

B. thetaiotaomicron genetic parts were characterized with NanoLuc luciferase (Hall MP et al. ACS Chem. Biol. 7:1848-1857, 2012), which is a small (19 kDa) modified shrimp luciferase. Efforts to use members of the green fluorescent protein family and a FMN-based fluorescent reporter were not successful. NanoLuc oxidizes the exogenously-added substrate furimazine to produce glow-type bioluminescence (E max = 460nm) with a signal half-life of 2 hr. By comparison, bacterial luciferase LuxAB (79 kDa) exhibited rapid signal decay when used to characterize gene expression in Bacteroides (Mastropaolo MD et al. Microbiology 155:2683-93, 2009).

Example 2. Expression control through promoter and RBS design

To expand the range of constitutive gene expression that can be implemented in Bacteroides, promoter-RBS combinations were constructed and characterized (Fig. 1C). Four promoter variants were constructed based on the constitutive promoter for the B.

thetaiotaomicron housekeeping sigma factor BT1311 (PBT II) (Vingadassalom D, et al. Mol. Microbiol. 56:888-902, 2005). Specifically, a 26-bp sequence was substituted or inserted into PBTI3II in regions composing and surrounding the -33 and -7 promoter sequences (Figs. 6A-6B). Promoter activity is affected by mutations in these regions (Bayley DP, et al. FEMS Microbiol. Lett. 193:149-54, 2000) or the equivalent regions in the promoters of other bacteria. The resulting promoters, designated PAMI, PAM2, PAM3, and PAM4, retained the BT1311 RBS and were used to control expression of the NanoLuc reporter in the pNBU2 vector backbone (Wang J, et al. J. Bacteriol. 182:3559-3571, 2000). The PAM promoters spanned a 20-fold range of expression and had decreased expression levels relative to the PBT II parent promoter. For comparison to prior work, the activities of promoter-RBS pairs, PcfxA, PcfiA, Pi and P ce p A (Wegmann U, et al. Appl. Environ. Microbiol. 79: 1980-9, 2013;

Parker AC, et al. Antimicrob. Agents Chemother. 37:1028-1036, 1993; Rogers MB, et al. J. Bacteriol. 176:4376-4384, 1994; and Goto T, et al. J. Antibiot. (Tokyo). 66:239-242, 2013) were also measured (Fig. ID).

The PAM promoters were then combined with RBSs of varying strength to increase the range of expression levels. The RBS is poorly understood in Bacteroides species, and the presence of a consensus Shine-Delgarno (SD) sequence based on the Bacteroides 16S rRNA does not greatly enhance translation initiation. RBSs GH022, GH023, and GH078 (Wegmann U, et al. Appl. Environ. Microbiol. 79:1980-9, 2013) were first used. As reported, this set of RBSs covered a limited range of expression spanning less than one order of magnitude (Fig. ID). Given that ribosomal proteins are predicted to be the most highly expressed proteins in most bacterial species, a ribosomal protein RBS (rpiL* in Fig. ID) was selected to increase the range of available RBSs. In addition, a weak B. thetaiotaomicron RBS (RC500) was constructed (Fig. ID). The RBS library consisting of RC500, GH022, GH023, GH078, and rpiL* spanned a >10 -fold range when paired with each PAM-derived promoter. When combined, these PA M promoters and RBSs could achieve expression levels over a 10 4 -fold range.

To identify a set of RBSs for fine-tuning gene expression in B. thetaiotaomicron, three randomized RBS libraries targeting the most conserved positions of the Bacteroides ribosomal protein RBSs were generated. Libraries were based on the rpiL* RBS and were characterized under the control of P BT II- The low GC content (14%) of the rpiL* RBS reduced the likelihood of introducing secondary structures during randomization. For each library, 3 nucleotides in and around the rpiL* RBS Shine Delgarno sequence were targeted. These positions are within or near the RBS region predicted to interact with the ribosomal SI protein (nt -21 to -11 relative to the start codon of NanoLuc, Fig. 1C) (Bloom SM, et al. Cell Host Microbe 9:390-403, 1991). Coverage of 67-80% of the 64 potential members as achieved in each library, resulting in 142 RBS sequences (Fig. IE, Table 1). These RBSs were screened and sequenced and a set of 8 was identified that span 10 -fold expression range in approximately even increments (Table 2).

RBS strength in Bacteroides species is reported to be sensitive to secondary structure and GC content, likely due to the inability to form mRNA-16S rRNA interactions. Only a weak positive correlation was observed between the minimum free energy of RBS folding and expression of the NanoLuc reporter (r = 0.19) in the rpiL* library (Fig. IF). To visualize the impact of GC content on RBS strength within this library, frequency logos were generated to compare the frequency of each nucleotide at each diversified position in the target sequence relative to the frequency of that nucleotide in the full library. As seen in Fig. 1G, the strongest RBSs were GC-depleted relative to the overall library, and the weakest RBSs sequences had a higher likelihood of containing a G or C at most positions tested. These data support findings that A/U rich regions upstream of the SD sequence enhance RBS strength. The RBS libraries provided herein highlight the distinct GC content depletion of Bacteroides RBSs compared to other bacterial species, which results in part failure when constructs are transferred into Bacteroides from other species.

Example 3. Genetic sensors and inducible systems

To create inducible systems for use in B. thetaiotaomicron, parts from a large repertoire of systems that govern carbohydrate utilization were used, which included cytoplasmic transcription factors, extracytoplasmic function sigma/anti-sigma pairs, and hybrid two-component systems (HTCS), among others (64). In B. thetaiotaomicron, rhamnose metabolism is mediated by the AraC/XylS -family transcriptional activator, RhaR, which activates transcription at the P BT3763 promoter (Patel EH, et al. Res. Microbiol.

159:678-84, 2008). To assay the functionality of P BT3763 as an inducible system, 250 bp of the promoter-RBS region was cloned upstream of the start codon of BT3763 into the pNBU2 expression vector to drive expression of NanoLuc. Gene expression was conditional on the concentration of rhamnose and demonstrated a response curve with an output dynamic range of 104-fold (Fig. 2A). Fitting the response curve to a Hill function revealed a threshold K of 0.3 mM and a Hill coefficient n = 1.4.

Two-component systems are signal-transduction mechanisms widespread in bacteria for sensing external stimuli. Bacteroides sp. possess a unique variant of these systems, called hybrid two-component systems (HTCSs), that incorporate both the sensor histidine kinase and response regulator of classical two-component systems into a single polypeptide chain. Putative HTCSs, BT3334 and BT0267, were identified in transcriptomic studies to control expression of the chondroitin sulfate (ChS)-inducible P BT 3324 promoter and arabinogalactan (AG)-inducible P BTO 268 promoter, respectively (64, 83). The promoter regions upstream of the BT3324 and BT0268 genes were used as the basis for two polysaccharide sensors.

Chondroitin sulfate induction of P BT 3324 and arabinogalactan induction P BTO 268 led to a 60-fold and 29-fold regulation of output gene expression, respectively (Figs. 2B and 2C).

Next, an IPTG-inducible system was developed. Pairs of LacOl operator sites were inserted in the strong P C f X A promoter in three locations: upstream of the -33 element (01), between the -33 and -7 elements (02) or just downstream of the transcription start site (03) (Figs. 7A-7B). The LacI Q repressor was expressed from the strong BT1311 promoter to achieve tight control of NanoLuc expression. Compared to the unmodified P C f X A promoter, the addition of synthetic operator sites diminished the maximum expression of NanoLuc (Figs. 7A-7B). This strategy produced two IPTG-inducible promoters that with thresholds at K = 86μΜ and K = 6μΜ (PLac023)- The induction of these systems elicits an 8- and 22-fold change in gene expression, respectively (Fig. 2D).

As the orthogonality of genetic parts is crucial for their simultaneous use, the degree of cross-talk between each inducible system was tested by incubating each engineered strain with the full set of carbohydrate inducers. The inducers themselves bear little structural similarity: rhamnose, a methyl-pentose sugar; ChS, a sulfated glycocosaminoglycan composed of chains of acetylgalactosamine and glucuronic acid residues; AG, a

polysaccharide composed of arabinose and galactose units; and IPTG, a molecular mimic of allolactose. Functionally, each inducible system was highly orthogonal to each other, with no cross-reactivity observed with any of the combinations (Fig. 2E). Example 4. Synthetic genetic memory

To enable genetic memory in B. thetaiotaomicron, serine integrases were

implemented, which permanently invert DNA between two recognition sequences (Fig. 3A). Recently, 11 orthogonal integrases and their recognition sequences were characterized in E. coli (Yang L, et al. Nat. Methods 11, 2014). In this study, a DNA "memory array" composed of a linear concatenation of integrase recognition sequences was used to record the expression of one or multiple integrases in response to a stimulus. Each integrase and its cognate recognition sequence in the memory array functioned as a switch that could be permanently flipped in response to integrase expression.

To equip B. thetaiotaomicron with permanent genetic memory, serine integrases that function in B. thetaiotaomicron were first identified by cloning the integrases into a strong constitutive expression vectors (PAM4-rpiL*, 1.2 X 10 " RLU/CFU). Using allelic exchange, the DNA memory array containing the integrase recognition sequences were incorporated into the B. thetaiotaomicron chromosome to provide a stable, single-copy record of DNA inversion (Figs. 3B and 3C). Integrase expression vectors were conjugated into the B.

thetaiotaomicron memory array strain. Genomic DNA was isolated from transconjugants and analyzed by PCR to detect flipping. Four integrases, Int7, Int8, Int9 and Intl2, each catalyzed recombination at the respective recognition sequence in the memory array (Fig. 3D), and DNA inversion was not detected in the absence of an integrase (Fig. 8A).

To create an inducible memory switch, Intl2 was cloned under the control of the rhamnose-inducible promoter with the rpiL*RBS variant C51 (Fig. 3E) {see also Fig. 1C, Table 1). The Intl2 recombinase switch responded to increasing concentrations of rhamnose (Fig. 3F) within 2 hours (Fig. 3G), with no background detected in the absence of inducer. Notably, expression of Intl2 did not impact growth of B. thetaiotaomicron, even when maximally expressed (Fig. 8B).

Example 5. CRISPRi-mediated gene knockdown

CRISPRi can provide a facile toolbox for constructing synthetic gene circuits and modulating endogenous genes in B. thetaiotaomicron. To demonstrate the use of CRISPRi- mediated gene knockdown for synthetic constructs, a set of guide RNAs (sgRNAs) that control expression of NanoLuc was first created (Fig. 4A). The production of dCas9 was regulated by the IPTG-inducible PLac023 system while sgRNAs were constitutively expressed from the Pi promoter. Four gRNAs targeting the coding sequence of NanoLuc (NL1-4) and two targeting the P C fiA promoter driving NanoLuc expression (PR1-2) were designed (Fig. 4A). A nonsense sgRNA (NS) with no sequence identity to either P C fiA or NanoLuc was used as a negative control. All of the specifically targeted guide RNAs repressed the expression of NanoLuc (Fig. 4B) by 20-45 fold with IPTG induction of dCas9 expression (Fig. 4C), thus implementing genetic NOT gates in B. thetaiotaomicron. The IPTG-to-NanoLuc response function of sgRNAs targeting the coding sequence or promoter exhibited similar Hill coefficients and lower dissociation constants to the IPTG-to-NanoLuc transfer function of the PLac023 promoter on its own (n = 1.1 to 1.4; K= 0.6 to 1.4μΜ IPTG).

To demonstrate the programmable knockdown of endogenous genes in B.

thetaiotaomicron, sgRNAs were designed to target mechanisms implicated in the resilience of Bacteroides in the human microbiota. Resistance to inflammation-associated cationic antimicrobial peptides, such as polymyxin B, is essential for the stability of commensal organisms in the dynamic gut environment. In B. thetaiotaomicron, LpxF, the gene product of BT1854, is required for the dephosphorylation of lipid A that leads to high levels of resistance to antimicrobial peptides. Using the minimum inhibitory concentration (MIC) of polymyxin B as a phenotypic readout, an sgRNA was designed to specifically suppress BT1854 expression. Similar to wild-type (WT) B. thetaiotaomicron, strains containing dCas9NS demonstrated high levels of polymyxin B resistance in the presence or absence of dCas9 induction with IPTG. However, in cells containing the sgRNA targeted against BT1854 (dCas9 BT i 854 ), the induction of dCas9 with led to sensitization of the cells to polymyxin B treatment, with a 8 to 16-fold decrease in MIC compared to WT and the nonspecific dCas9NS control (Fig. 4E).

Next, whether dCas9-mediated repression of carbohydrate-utilization pathways could alter the metabolic capabilities of B. thetaiotaomicron was explored, which pathways are important for the bacterium's ability to successfully and persistently colonize the mammalian gut. Fructose-containing carbohydrates are catabolized by the gene products of the BT1757- 1763/BT1765 polysaccharide utilization locus, which is subject to regulation by the HTCS sensor, BT1754 (Sonnenburg ED, et al. Cell 141:1241-52, 2010). BT1754 is essential for growth on fructose-containing carbohydrates and genetic inactivation of BT1754 leads to retarded growth in minimal media (MM) containing fructose as the sole carbon source. To modulate the ability of B. thetaiotaomicron to utilize fructose, a specific guide RNA was designed to repress BT1754 and integrated this system into the B. thetaiotaomicron genome along with an IPTG-inducible dCas9 cassette (dCas9 BT i 754 ). Induction of dCas9 BT i 754 did not affect the growth rate of cells on MM-glucose compared to WT cells and dCas9 s- The generation time G = (logio2 » t)/logio(B/Bo) ~ lhr (where t is the time interval, and B 0 and B are the initial and final concentrations of bacteria, respectively), indicating that neither dCas9 induction nor repression of BT1754 impacts growth on glucose media (Fig. 4F). However, induction of dCas9 BT i 754 drastically decreased the growth rate of the cells in MM-fructose (G = 4.7 hr) while the growth of WT and dCas9 s cells in MM-fructose remained similar (G = 1 hr) to growth in MM-glucose (Fig. 4F). Thus, inducible dCas9-mediated repression of endogenous genes can alter both the resistance and metabolic profiles of B. thetaiotaomicron. Example 6. Function of genetic parts in B. thetaiotaomicron colonizing the mouse gut

Next investigated was whether the function of the B. thetaiotaomicron genetic parts and modules can be maintained in the context of a complex microbiota. As wild-type strains of Bacteroides spp. are unable to stably colonize conventional specific -pathogen free (SPF) mice, an antibiotic regimen that promotes B. thetaiotaomicron colonization without sterilizing the gut microbiota was employed (Fig. 5A) (Lee SM, et al. Nature 501:426-9,

2013; Bloom SM, et al. Cell Host Microbe 9:390-403, 2011). A ten-day treatment of animals with ciprofloxacin and metronidazole prior to bacterial inoculation was sufficient to maintain stable and high levels of colonization for the duration of the experiments (up to 12 days tested) (Figs. 9A-9C).

Using this model, the functionality of the inducible systems were tested, CRISPRi, and integrases in vivo. First, SPF mice were colonized with the strain containing the arabinogalactan-inducible P0268 promoter driving expression of NanoLuc (Fig. 9A). Within a day of addition of arabinogalactan to the drinking water of the mice, luciferase activity in fecal pellets increased approximately 75-fold (Fig. 5B). Following removal of inducer from the drinking water, luciferase activity in the fecal pellets of mice fed inducer rapidly returned to baseline, demonstrating tight temporal control of gene expression dependent on

arabinogalactan.

To investigate whether more complex genetic circuits perform in the context of the mouse microbiome, the dCas9 L3 repressor cascade was evaluated, which is composed of the CRISPRi system as well as the PLac023 IPTG-inducible promoter, within stably colonized B. thetaiotaomicron. Within 24 hours of adding IPTG to drinking water, CRISPRi elicited approximately a 20-fold reduction in gene expression compared to the uninduced control (Fig. 5C). The fold repression observed in vivo is similar to that measured in vitro. Luciferase activity returned to baseline 6 days following the removal of IPTG from drinking water. Moreover, expression of dCas9 and NanoLuc did not significantly impact in vivo fitness compared to uninduced controls (Figs. 9A and 9B). Thus, inducible promoters as well as exogenously regulated CRISPRi can be implemented for on-demand activation or repression of synthetic genetic circuits in members of a mammalian microbiome.

To test the function of recombinases in vivo, mice were colonized with a B.

thetaiotaomicron strain containing the rhamnose-inducible Intl2 integrase memory switch (Fig. 3E). Rhamnose biosynthetic pathways are absent in higher vertebrates, but rhamnose is a common component of the plant and bacterial cell wall. All mice were fed with plant-based chow that was determined to be composed of 0.3% rhamnose (w/w). In addition, after one day of colonization, the drinking water of half of the mice was supplemented with 0.5M rhamnose for two days to further induce the memory switch. Stool was collected over the course of the experiment, and the absolute number of unflipped (wild-type) and flipped Intl2 recognition sequences was determined by qPCR using standard curves generated with purified, homogenous template DNA. Recombination frequency is reported as the ratio of flipped to total memory array sequences (Fig. 5D). A background recombination rate of

-11% per day was detected in mice fed on rhamnose-containing chow but not supplemented with rhamnose in their drinking water (Fig. 5D, "Chow"). In mice supplemented with exogenous rhamnose (Fig. 5D, "Chow + Rha"), the recombinase switch achieved >90% flipping in <1 day, a statistically significant increase over mice not supplemented with rhamnose in the water (p<0.01; Fig. 5D). Together, these results indicate that inducible recombinase systems can be implemented within B. thetaiotaomicron living in the mouse gut.

Example 7. Generation ofpNBUl

An integration vector, designated pNBUl, was created to introduce recombinant DNA into a wide range of Bacteroides species (Fig. 10). IntNl integrase catalyzes site-specific genomic integration of the plasmid into recipient Bacteroides strains. Following transfer of pNBUl, the IntNl integrase is expressed, binds to its cognate attP site on the plasmid and catalyzes integration of the plasmid backbone at attB sites located in the Bacteroides genome. pNBUl shows a greater host range and efficiency relative to the pNBU2 plasmid, discussed above. pNBUl is capable of facilitating gene expression in multiple Bacteroides spp, including, for example, B. thetaiotaomicron, B.fragilis, B. ovatus, B. vulgatus, B. caccae, B. eggerthii and Parabacteroides distasonis. Further, pNBUl comprises a variant of the IntNl attP site (SEQ ID NO: 207) that exhibits high specificity and low off-target integration events. Table 1.

Table 2. Part Name Type DNA sequence SEQ ID NO.

TTACAAAGAAAATTCGACAAACTGTTATTTTTCTA

Synthetic IPTG- TCTATTTAAATTGTGAGCGGATAACAATTTGAATT

PLac012 inducible GTGAGCGGATAACAATTACCTTTGTCGGCAAATA 147

promo ter+RBS AAGATATTCTCGTCAAACAAATATAAATAATATA

AAC

TTACAAAGAAAATTCGACAAACTGTTATTTTTCTA

Synthetic IPTG- TCTATTTAAATTGTGAGCGGATAACAATTTGGGTG

PLacOO inducible GGAAACTTTAGTTATGTACCTTTGTCGGCAATTGT 148

promo ter+RBS GAGCGGATAACAATTAAATAAAGATATTCTCGTC

AAACAAATATAAATAATATAAAC

TTACAAAGAAAATTCGACAAACTGTTATTTTTCTA

Synthetic IPTG- TCTATTTATTTGAATTGTGAGCGGATAACAATTAC

PLac023 inducible CTTTGTCGGCAATTGTGAGCGGATAACAATTAAA 149

promo ter+RBS TAAAGATATTCTCGTCAAACAAATATAAATAATA

TAAAC

GTGGTGAATGTGAAACCAGTAACGTTATACGATG

TCGCAGAGTATGCCGGTGTCTCTTATCAGACCGTT

TCCCGCGTGGTGAACCAGGCCAGCCACGTTTCTG

CGAAAACGCGGGAAAAAGTGGAAGCGGCGATGG

CGGAGCTGAATTACATTCCCAACCGCGTGGCACA

ACAACTGGCGGGCAAACAGTCGTTGCTGATTGGC

GTTGCCACCTCCAGTCTGGCCCTGCACGCGCCGTC

GCAAATTGTCGCGGCGATTAAATCTCGCGCCGAT

CAACTGGGTGCCAGCGTGGTGGTGTCGATGGTAG

AACGAAGCGGCGTCGAAGCCTGTAAAGCGGCGGT

GCACAATCTTCTCGCGCAACGCGTCAGTGGGCTG

ATCATTAACTATCCGCTGGATGACCAGGATGCCA

TTGCTGTGGAAGCTGCCTGCACTAATGTTCCGGCG

TTATTTCTTGATGTCTCTGACCAGACACCCATCAA

CAGTATTATTTTCTCCCATGAGGACGGTACGCGAC

Transcriptional TGGGCGTGGAGCATCTGGTCGCATTGGGTCACCA

Laclq 150

repressor GCAAATCGCGCTGTTAGCGGGCCCATTAAGTTCT

GTCTCGGCGCGTCTGCGTCTGGCTGGCTGGCATA

AATATCTCACTCGCAATCAAATTCAGCCGATAGC

GGAACGGGAAGGCGACTGGAGTGCCATGTCCGGT

TTTCAACAAACCATGCAAATGCTGAATGAGGGCA

TCGTTCCCACTGCGATGCTGGTTGCCAACGATCAG

ATGGCGCTGGGCGCAATGCGCGCCATTACCGAGT

CCGGGCTGCGCGTTGGTGCGGATATCTCGGTAGT

GGGATACGACGATACCGAGGACAGCTCATGTTAT

ATCCCGCCGTTAACCACCATCAAACAGGATTTTC

GCCTGCTGGGGCAAACCAGCGTGGACCGCTTGCT

GCAACTCTCTCAGGGCCAGGCGGTGAAGGGCAAT

CAGCTGTTGCCCGTCTCACTGGTGAAAAGAAAAA

CCACCCTGGCGCCCAATACGCAAACCGCCTCTCC

CCGCGCGTTGGCCGATTCATTAATGCAGCTGGCA

CGACAGGTTTCCCGACTGGAAAGCGGGCAGTGA

TTACAAAGAAAATTCGACAAACTGTTATTTTTCTA

Constitutive TCTATTTATTTGGGTGGGAAACTTTAGTTATGTAC

PcfxA 151

promo ter+RBS CTTTGTCGGCAAATAAAGATATTCTCGTCAAACA

AATATAAATAATATAAAC

TGATCTGGAAGAAGCAATGAAAGCTGCTGTTAAG

TCTCCGAATCAGGTATTGTTCCTGACAGGTGTATT

CCCATCCGGTAAACGCGGATACTTTGCAGTTGAT

CTGACTCAGGAATAAATTATAAATTAAGGTAAGA

Constitutive

PBT1311 AGATTGTAGGATAAGCTAATGAAATAGAAAAAG 152

promo ter+RBS

GATGCCGTCACACAACTTGTCGGCATTCTTTTTTG

TTTTATTAGTTGAAAATATAGTGAAAAAGTTGCCT

AAATATGTATGTTAACAAATTATTTGTCGTAACTT

TGCACTCCAAATCTGTTTTTAACATATGGCACTA Part Name Type DNA sequence SEQ ID NO.

GATAAAGTTTGGAAGATAAAGCTAAAAGTTCTTA

Constitutive

Pl-RBS TCTTTGCAGTCCGAAATAAAGACATATAAAAGAA 153

promo ter+RBS

AAGACACC

GGAGTGAGCTTCTCGGATTTTATTTGTATTTTTGC

CATGCCTGATGAGGTTTTGTTTGATTATTTTTTTGC

Constitutive AACACTAAGTTAAGTGAATCCTCTGACATGGCAA

PcfiA 154

promo ter+RBS AATCCTGAGCAACTTTTTGTTGCTCAGGTACTTAA

AAAAAATATTTTATAATAGTGTTGCGGAATTAAG

GTAAAAGAATAAA

Constitutive CAAATTTGCGCGCCACAATTATTATTCATACCTTT

PcepA 155

promo ter+RBS GTGGACCGTATTACAAAGAACCCAATCATAT

Constitutive GATAAAGTTTGGAAGATAAAGCTAAAAGTTCTTA

PI 156

promoter TCTTTGCAGT

ATGGATAAGAAATACTCAATAGGCTTAGCTATCG

GCACAAATAGCGTCGGATGGGCGGTGATCACTGA

TGAATATAAGGTTCCGTCTAAAAAGTTCAAGGTT

CTGGGAAATACAGACCGCCACAGTATCAAAAAAA

ATCTTATAGGGGCTCTTTTATTTGACAGTGGAGAG

ACAGCGGAAGCGACTCGTCTCAAACGGACAGCTC

GTAGAAGGTATACACGTCGGAAGAATCGTATTTG

TTATCTACAGGAGATTTTTTCAAATGAGATGGCG

AAAGTAGATGATAGTTTCTTTCATCGACTTGAAG

AGTCTTTTTTGGTGGAAGAAGACAAGAAGCATGA

ACGTCATCCTATTTTTGGAAATATAGTAGATGAA

GTTGCTTATCATGAGAAATATCCAACTATCTATCA

TCTGCGAAAAAAATTGGTAGATTCTACTGATAAA

GCGGATTTGCGCTTAATCTATTTGGCCTTAGCGCA

TATGATTAAGTTTCGTGGTCATTTTTTGATTGAGG

GAGATTTAAATCCTGATAATAGTGATGTGGACAA

ACTATTTATCCAGTTGGTACAAACCTACAATCAAT

TATTTGAAGAAAACCCTATTAACGCAAGTGGAGT

AGATGCTAAAGCGATTCTTTCTGCACGATTGAGT

AAATCAAGACGATTAGAAAATCTCATTGCTCAGC

TCCCCGGTGAGAAGAAAAATGGCTTATTTGGGAA

TCTCATTGCTTTGTCATTGGGTTTGACCCCTAATTT

Catalytically- TAAATCAAATTTTGATTTGGCAGAAGATGCTAAA dCas9 inactive nuclease TTACAGCTTTCAAAAGATACTTACGATGATGATTT 157

for CRISPRi AGATAATTTATTGGCGCAAATTGGAGATCAATAT

GCTGATTTGTTTTTGGCAGCTAAGAATTTATCAGA

TGCTATTTTACTTTCAGATATCCTAAGAGTAAATA

CTGAAATAACTAAGGCTCCCCTATCAGCTTCAAT

GATTAAACGCTACGATGAACATCATCAAGACTTG

ACTCTTTTAAAAGCTTTAGTTCGACAACAACTTCC

AGAAAAGTATAAAGAAATCTTTTTTGATCAATCA

AAAAACGGATATGCAGGTTATATTGATGGGGGAG

CTAGCCAAGAAGAATTTTATAAATTTATCAAACC

AATTTTAGAAAAAATGGATGGTACTGAGGAATTA

TTGGTGAAACTAAATCGTGAAGATTTGCTGCGCA

AGCAACGGACCTTTGACAACGGCTCTATTCCCCA

TCAAATTCACTTGGGTGAGCTGCATGCTATTTTGA

GAAGACAAGAAGACTTTTATCCATTTTTAAAAGA

CAATCGTGAGAAGATTGAAAAAATCTTGACTTTT

CGAATTCCTTATTATGTTGGTCCATTGGCGCGTGG

CAATAGTCGTTTTGCATGGATGACTCGGAAGTCT

GAAGAAACAATTACCCCATGGAATTTTGAAGAAG

TTGTCGATAAAGGTGCTTCAGCTCAATCATTTATT

GAACGCATGACAAACTTTGATAAAAATCTTCCAA

ATGAAAAAGTACTACCAAAACATAGTTTGCTTTA

TGAGTATTTTACGGTTTATAACGAATTGACAAAG

GTCAAATATGTTACTGAAGGAATGCGAAAACCAG Part Name Type DNA sequence SEQ ID NO.

CATTTCTTTCAGGTGAACAGAAGAAAGCCATTGT

TGATTTACTCTTCAAAACAAATCGAAAAGTAACC

GTTAAGCAATTAAAAGAAGATTATTTCAAAAAAA

TAGAATGTTTTGATAGTGTTGAAATTTCAGGAGTT

GAAGATAGATTTAATGCTTCATTAGGTACCTACC

ATGATTTGCTAAAAATTATTAAAGATAAAGATTTT

TTGGATAATGAAGAAAATGAAGATATCTTAGAGG

ATATTGTTTTAACATTGACCTTATTTGAAGATAGG

GAGATGATTGAGGAAAGACTTAAAACATATGCTC

ACCTCTTTGATGATAAGGTGATGAAACAGCTTAA

ACGTCGCCGTTATACTGGTTGGGGACGTTTGTCTC

GAAAATTGATTAATGGTATTAGGGATAAGCAATC

TGGCAAAACAATATTAGATTTTTTGAAATCAGAT

GGTTTTGCCAATCGCAATTTTATGCAGCTGATCCA

TGATGATAGTTTGACATTTAAAGAAGACATTCAA

AAAGCACAAGTGTCTGGACAAGGCGATAGTTTAC

ATGAACATATTGCAAATTTAGCTGGTAGCCCTGCT

ATTAAAAAAGGTATTTTACAGACTGTAAAAGTTG

TTGATGAATTGGTCAAAGTAATGGGGCGGCATAA

GCCAGAAAATATCGTTATTGAAATGGCACGTGAA

AATCAGACAACTCAAAAGGGCCAGAAAAATTCGC

GAGAGCGTATGAAACGAATCGAAGAAGGTATCA

AAGAATTAGGAAGTCAGATTCTTAAAGAGCATCC

TGTTGAAAATACTCAATTGCAAAATGAAAAGCTC

TATCTCTATTATCTCCAAAATGGAAGAGACATGT

ATGTGGACCAAGAATTAGATATTAATCGTTTAAG

TGATTATGATGTCGATGCCATTGTTCCACAAAGTT

TCCTTAAAGACGATTCAATAGACAATAAGGTCTT

AACGCGTTCTGATAAAAATCGTGGTAAATCGGAT

AACGTTCCAAGTGAAGAAGTAGTCAAAAAGATGA

AAAACTATTGGAGACAACTTCTAAACGCCAAGTT

AATCACTCAACGTAAGTTTGATAATTTAACGAAA

GCTGAACGTGGAGGTTTGAGTGAACTTGATAAAG

CTGGTTTTATCAAACGCCAATTGGTTGAAACTCGC

CAAATCACTAAGCATGTGGCACAAATTTTGGATA

GTCGCATGAATACTAAATACGATGAAAATGATAA

ACTTATTCGAGAGGTTAAAGTGATTACCTTAAAA

TCTAAATTAGTTTCTGACTTCCGAAAAGATTTCCA

ATTCTATAAAGTACGTGAGATTAACAATTACCAT

CATGCCCATGATGCGTATCTAAATGCCGTCGTTGG

AACTGCTTTGATTAAGAAATATCCAAAACTTGAA

TCGGAGTTTGTCTATGGTGATTATAAAGTTTATGA

TGTTCGTAAAATGATTGCTAAGTCTGAGCAAGAA

ATAGGCAAAGCAACCGCAAAATATTTCTTTTACT

CTAATATCATGAACTTCTTCAAAACAGAAATTAC

ACTTGCAAATGGAGAGATTCGCAAACGCCCTCTA

ATCGAAACTAATGGGGAAACTGGAGAAATTGTCT

GGGATAAAGGGCGAGATTTTGCCACAGTGCGCAA

AGTATTGTCCATGCCCCAAGTCAATATTGTCAAG

AAAACAGAAGTACAGACAGGCGGATTCTCCAAG

GAGTCAATTTTACCAAAAAGAAATTCGGACAAGC

TTATTGCTCGTAAAAAAGACTGGGATCCAAAAAA

ATATGGTGGTTTTGATAGTCCAACGGTAGCTTATT

CAGTCCTAGTGGTTGCTAAGGTGGAAAAAGGGAA

ATCGAAGAAGTTAAAATCCGTTAAAGAGTTACTA

GGGATCACAATTATGGAAAGAAGTTCCTTTGAAA

AAAATCCGATTGACTTTTTAGAAGCTAAAGGATA

TAAGGAAGTTAAAAAAGACTTAATCATTAAACTA

CCTAAATATAGTCTTTTTGAGTTAGAAAACGGTCG

TAAACGGATGCTGGCTAGTGCCGGAGAATTACAA Part Name Type DNA sequence SEQ ID NO.

AAAGGAAATGAGCTGGCTCTGCCAAGCAAATATG

TGAATTTTTTATATTTAGCTAGTCATTATGAAAAG

TTGAAGGGTAGTCCAGAAGATAACGAACAAAAA

CAATTGTTTGTGGAGCAGCATAAGCATTATTTAG

ATGAGATTATTGAGCAAATCAGTGAATTTTCTAA

GCGTGTTATTTTAGCAGATGCCAATTTAGATAAA

GTTCTTAGTGCATATAACAAACATAGAGACAAAC

CAATACGTGAACAAGCAGAAAATATTATTCATTT

ATTTACGTTGACGAATCTTGGAGCTCCCGCTGCTT

TTAAATATTTTGATACAACAATTGATCGTAAACG

ATATACGTCTACAAAAGAAGTTTTAGATGCCACT

CTTATCCATCAATCCATCACTGGTCTTTATGAAAC

ACGCATTGATTTGAGTCAGCTAGGAGGTGACTGA

NNNNNNNNNNNNNNNNNNNNGTTTTAGAGCTAG

Guide RNA for AAATAGCAAGTTAAAATAAGGCTAGTCCGTTATC sgRNA 158

CRISPRi AACTTGAAAAAGTGGCACCGAGTCGGTGCTTTTT

T

ATGGTTTTTACTCTGGAAGATTTTGTTGGCGATTG

GCGTCAGACCGCGGGTTATAATTTGGATCAAGTC

CTGGAACAGGGTGGCGTAAGCTCTCTGTTCCAGA

ACCTGGGTGTGAGCGTGACGCCGATTCAGCGCAT

CGTTCTGTCCGGCGAGAACGGTCTGAAAATTGAT

ATTCATGTGATCATCCCGTACGAAGGCCTGAGCG

GTGACCAAATGGGTCAAATCGAGAAAATCTTTAA

Luciferase AGTCGTCTACCCAGTTGACGATCACCACTTCAAG

NanoLuc 159

reporter GTTATCTTGCATTACGGTACGCTGGTGATTGATGG

TGTGACCCCGAATATGATTGACTATTTCGGCCGTC

CGTATGAAGGCATTGCCGTTTTTGACGGTAAAAA

GATCACCGTCACCGGTACCCTGTGGAATGGCAAT

AAGATTATTGACGAGCGTCTGATTAACCCGGACG

GCAGCCTGCTGTTCCGCGTGACCATCAACGGTGT

CACGGGTTGGCGTCTGTGCGAGCGCATCCTGGCA

TAA

TGATCTGGAAGAAGCAATGAAAGCTGCTGTTAAG

TCTCCGAATCAGGTATTGTTCCTGACAGGTGTATT

CCCATCCGGTAAACGCGGATACTTTGCAGTTGAT

Synthetic

CTGACTCAGGAATAAATTATAAATTAAGGTAAGA

constitutive

PAM1 AGATTGTAGGATAAGCTAATGAAATAGAAAAAG 160

promoter +

GATGCCGTCACACAACTTGTCGGCATTCTTTTTTG PBT1311 RBS

ctttgcaacagcatagctcagcacagAAGTTGCCTAAATATGTA

TGTTAACAAATTATTTGTCGTAACTTTGCACTCCA

AATCTGTTTTTAACATATGGCACTA

TGATCTGGAAGAAGCAATGAAAGCTGCTGTTAAG

TCTCCGAATCAGGTATTGTTCCTGACAGGTGTATT

CCCATCCGGTAAACGCGGATACTTTGCAGTTGAT

Synthetic CTGACTCAGGAATAAATTATAAATTAAGGTAAGA constitutive AGATTGTAGGATAAGCTAATGAAATAGAAAAAG

PAM2 161

promoter + GATGCCGTCACACAACTTGTCGGCATTCTTTTTTG PBT1311 RBS TTTTATTAGTTGAAAATATAGTGAAAAAGTTGCCT

AAATATGTATGTTAACAAATTctttgcaacagcatagctcagc

acagGCACTCCAAATCTGTTTTTAACATATGGCACT

A

TGATCTGGAAGAAGCAATGAAAGCTGCTGTTAAG

TCTCCGAATCAGGTATTGTTCCTGACAGGTGTATT

Synthetic CCCATCCGGTAAACGCGGATACTTTGCAGTTGAT constitutive CTGACTCAGGAATAAATTATAAATTAAGGTAAGA

PAM3 162

promoter + AGATTGTAGGATAAGCTAATGAAATAGAAAAAG PBT1311 RBS GATGCCGTCACACAACTTGTCGGCATTCTTTTTTG

TTTTATTAGTTGAAAATATAGTGAAAActttgcaacagca

tagctcagcacagATTATTTGTCGTAACTTTGCACTCCAA Part Name Type DNA sequence SEQ ID NO.

ATCTGTTTTTAACATATGGCACTA

TGATCTGGAAGAAGCAATGAAAGCTGCTGTTAAG

TCTCCGAATCAGGTATTGTTCCTGACAGGTGTATT

CCCATCCGGTAAACGCGGATACTTTGCAGTTGAT

Synthetic CTGACTCAGGAATAAATTATAAATTAAGGTAAGA constitutive AGATTGTAGGATAAGCTAATGAAATAGAAAAAG

PAM4 163

promoter + GATGCCGTCACACAACTTGTCGGCATTCTTTTTTG PBT1311 RBS TTTTATTAGTTGAAAATATAGTGAAAAAGTTGCCT

AAATATGTATGTTAACAAATTATTTGTCGTAACTT

TGCACTCCctttgcaacagcatagctcagcacagAAATCTGTTTT

TAACAT

ATGAAAGTGGCCATTTATGTTCGTGTTAGCACCG

ATGAACAGGCCAAAGAAGGTTTTAGCATTCCGGC

ACAGCGTGAACGTCTGCGTGCATTTTGTGCAAGC

CAGGGTTGGGAAATTGTGCAAGAATATATTGAAG

AAGGTTGGAGCGCAAAAGATCTGGATCGTCCGCA

GATGCAGCGTCTGCTGAAAGATATCAAAAAAGGC

AACATTGATATTGTGCTGGTGTATCGTCTGGATCG

CCTGACCCGTAGCGTTCTGGATCTGTATCTGCTGC

TGCAGACCTTTGAAAAATACAATGTGGCATTTCG

TAGCGCCACCGAAGTTTATGATACCAGCACCGCA

ATGGGTCGTCTGTTTATTACCCTGGTTGCAGCACT

GGCACAGTGGGAACGTGAAAATCTGGCAGAACGT

GTTAAATTTGGTATCGAGCAGATGATCGATGAAG

GTAAAAAACCGGGTGGTCATAGCCCGTATGGTTA

CAAATTTGATAAAGACTTCAATTGCACCATTATTG

AGGAAGAAGCAGACGTTGTTCGTATGATCTATCG

CATGTATTGTGATGGTTATGGCTATCGTAGCATTG

CAGATCGTCTGAATGAACTGATGGTTAAACCGCG

TATTGCCAAAGAATGGAATCATAATAGCGTGCGT

GATATCCTGACCAACGATATCTATATTGGCACCTA

Int7 Serine integrase TCGTTGGGGTGATAAAGTTGTTCCGAATAATCATC 164

CGCCTATTATTAGCGAAACCCTGTTCAAAAAAGC

CCAGAAAGAAAAAGAAAAACGTGGCGTTGATCG

TAAACGCGTTGGTAAATTTCTGTTTACCGGTCTGC

TGCAGTGTGGTAATTGTGGTGGCCATAAAATGCA

GGGCCATTTTGATAAACGTGAGCAGAAAACCTAT

TACCGTTGTACCAAATGTCACCGCATTACCAACG

AAAAAAACATTCTGGAACCGCTGCTGGATGAAAT

TCAGCTGCTGATTACCAGCAAAGAATACTTTATG

AGCAAATTCAGCGACCGCTATGATCAGCAAGAGG

TTGTTGATGTTAGCGCACTGACAAAAGAACTGGA

AAAAATCAAACGCCAGAAAGAGAAATGGTACGA

TCTGTATATGGATGATCGTAACCCGATTCCGAAA

GAAGAACTGTTTGCCAAAATTAACGAACTGAACA

AAAAAGAAGAAGAAATCTATAGCAAGCTGAGCG

AAGTGGAAGAAGATAAAGAACCGGTTGAAGAGA

AATATAACCGCCTGAGCAAAATGATCGATTTTAA

ACAGCAGTTTGAGCAGGCCAACGACTTTACCAAA

AAAGAGCTGCTGTTCAGCATCTTCGAAAAGATTG

TGATTTATCGCGAGAAAGGCAAGCTGAAAAAAAT

CACCCTGGATTACACCCTGAAATAA Part Name Type DNA sequence SEQ ID NO.

ATGAAAGTTGCCGTTTATTGTCGTGTTAGCACCCT

GGAACAGAAAGAACATGGTCATAGCATTGAAGA

ACAAGAGCGTAAACTGAAAAGCTTCTGCGATATT

AATGATTGGACCGTGTATGATACCTATATCGATG

CAGGTTATAGCGGTGCAAAACGTGATCGTCCGGA

ACTGCAGCGTCTGATGAATGATATTAACAAATTT

GATCTGGTGCTGGTGTATAAACTGGATCGTCTGA

CCCGTAATGTTCGTGATCTGCTGGACCTGCTGGAA

ATCTTTGAAAAAAATGATGTGAGCTTTCGTAGCG

CCACCGAAGTTTATGATACCACCACCGCAATGGG

TCGTCTGTTTGTTACCCTGGTTGGTGCAATGGCAG

AATGGGAACGTGAAACCATTCGTGAACGTACCCA

GATGGGTAAACTGGCAGCACTGCGTAAAGGTATT

ATGCTGACCACCCCTCCGTTTTATTATGACCGTGT

GGATAATAAGTTTGTGCCGAACAAATACAAAGAC

GTTATTCTGTGGGCATATGACGAAGCAATGAAAG

GTCAGAGCGCAAAAGCAATTGCACGCAAACTGAA

TAATAGCGATATTCCGCCTCCGAATAATACCCAG

TGGCAGGGTCGTACCATTACCCATGCCCTGCGTA

ATCCGTTTACCCGTGGTCATTTTGATTGGGGTGGT

Int8 Serine integrase GTGCATATTGAAAATAACCATGAACCGATCATCA 165

CCGATGAGATGTATGAGAAAGTTAAAGATCGCCT

GAATGAACGCGTGAACACCAAAAAAGTTCGTCAT

ACCAGCATTTTTCGTGGCAAACTGGTTTGTCCGGT

TTGTAATGCACGCCTGACCCTGAATAGCCATAAA

AAGAAAAGCAATAGCGGCTATATCTTTGTGAAAC

AGTACTACTGCAACAACTGTAAAGTTACCCCGAA

TCTGAAACCGGTGTACATCAAAGAAAAAGAAGTG

ATTAAAGTTTTTTACAATTATCTGAAACGCTTCGA

TCTGGAAAAATATGAGGTTACCCAGAAACAGAAC

GAACCGGAAATCACCATCGATATCAATAAAGTTA

TGGAACAGCGCAAACGCTACCATAAACTGTATGC

AAGCGGTCTGATGCAAGAAGATGAACTGTTTGAC

CTGATTAAAGAAACCGATCAGACCATTGCCGAAT

ATGAAAAACAGAATGAAAACCGCGAAGTGAAGC

AGTATGATATCGAAGATATCAAACAGTATAAAGA

TCTGCTGTTAGAAATGTGGGATATCAGCTCCGAT

GAAGATAAAGAGGACTTTATCAAAATGGCGATTA

AAAACATCTATTTTGAATATATCATTGGCACCGGT

AACACCAGCCGTAAACGTAATAGCCTGAAAATTA

CGAGCATTGAATTCTATTAA

ATGAAAGTGGCCATTTATACCCGTGTTAGCACCCT

GGAACAGAAAGAAAAAGGTCATAGCATCGAAGA

ACAAGAACGTAAACTGCGTGCATATAGCGATATC

AACGATTGGAAAATCCACAAAGTTTATACCGATG

CAGGTTATAGCGGTGCCAAAAAAGATCGTCCGGC

ACTGCAAGAAATGCTGAATGAAATTGATAACTTC

GATCTGGTGCTGGTGTATAAACTGGATCGTCTGA

CCCGTAGCGTTAAAGATCTGCTGGAAATTCTGGA

ACTGTTTGAAAACAAAAACGTGCTGTTTCGTAGC

Int9 Serine integrase GCCACCGAAGTTTATGATACCACCAGTGCAATGG 166

GTCGTCTGTTTGTTACCCTGGTTGGTGCAATGGCA

GAATGGGAACGTACCACCATTCAAGAACGCACCG

CCATGGGTCGCCGTGCAAGCGCACGTAAAGGTCT

GGCAAAAACCGTTCCGCCTTTCTATTATGATCGCG

TGAATGATAAATTTGTGCCGAACGAGTACAAAAA

GGTTCTGCGTTTTGCAGTTGAAGAAGCAAAAAAA

GGCACCAGCCTGCGTGAAATTACCATTAAACTGA

ACAACAGCAAATACAAAGCACCGCTGGGTAAAA

ATTGGCATCGTAGCGTGATTGGTAATGCACTGAC Part Name Type DNA sequence SEQ ID NO.

CAGTCCGGTTGCACGTGGTCATCTGGTTTTTGGTG

ATATTTTTGTGGAAAACACCCACGAAGCCATTATT

AGCGAAGAGGAATATGAAGAAATCAAGCTGCGC

ATTAGCGAAAAAACCAATAGCACCATTGTGAAAC

ACAACGCCATTTTTCGTAGCAAACTGCTGTGTCCG

AATTGCAATCAGAAACTGACCCTGAATACCGTTA

AACATACCCCGAAAAACAAAGAGGTGTGGTACA

GCAAACTGTATTTTTGCAGCAATTGCAAAAACAC

CAAAAATAAGAACGCCTGCAACATCGATGAAGGT

GAAGTTCTGAAACAGTTCTACAACTATCTGAAGC

AGTTTGATCTGACCAGCTACAAAATTGAAAACCA

GCCGAAAGAAATTGAGGATGTGGGCATTGATATT

GAAAAACTGCGTAAAGAACGTGCCCGTTGTCAGA

CCCTGTTTATTGAAGGTATGATGGATAAAGATGA

AGCCTTTCCGATTATTAGCCGCATCGATAAAGAA

ATCCACGAGTATGAAAAACGCAAAGACAACGAT

AAAGGCAAAACCTTTAACTATGAAAAGATTAAAA

ACTTCAAATATAGCCTGCTGAACGGCTGGGAACT

GATGGAAGATGAACTGAAAACCGAGTTTATCAAG

ATGGCGATCAAAAACATCCACTTTGAGTATGTGA

AAGGCATCAAAGGTAAACGTCAGAACAGCCTGA

AAATTACCGGCATCGAATTCTATTAA

ATGAAAGTGGCCATTTATACCCGTGTTAGCAGCG

CAGAACAGGCAAATGAAGGTTATAGCATTCACGA

GCAGAAGAAGAAACTGATCAGCTATTGCGAAATC

CACGATTGGAACGAGTATAAAGTTTTTACCGATG

CAGGTATTAGCGGTGGTAGCATGAAACGTCCGGC

ACTGCAAAAACTGATGAAACATCTGAGTTCATTT

GATCTGGTGCTGGTGTATAAACTGGATCGTCTGA

CCCGTAATGTTCGTGATCTGCTGGATATGCTGGAA

GAATTTGAACAGTATAACGTGAGCTTTAAAAGCG

CCACCGAAGTTTTTGATACCACCAGTGCAATTGG

CAAACTGTTTATTACCATGGTTGGTGCAATGGCA

GAATGGGAACGTGAAACCATTCGTGAACGTAGCC

TGTTTGGTAGCCGTGCAGCAGTTCGTGAAGGTAA

CTATATTCGTGAAGCACCGTTTTGCTATGATAACA

TTGAAGGTAAACTGCACCCGAACGAATATGCCAA

AGTTATTGATCTGATTGTGAGCATGTTCAAAAAA

GGCATTAGCGCCAATGAAATTGCACGTCGTCTGA

ATAGCAGCAAAGTTCATGTTCCGAACAAAAAAAG

CTGGAATCGTAATAGCCTGATTCGTCTGATGCGTA

Intl2 Serine integrase 167

GTCCGGTTCTGCGTGGTCATACCAAATATGGTGAT

ATGCTGATTGAAAACACCCATGAACCGGTGCTGA

GCGAACATGATTATAATGCAATTAACAACGCCAT

CAGCAGCAAAACCCATAAAAGCAAAGTTAAACA

CCATGCCATTTTTCGTGGTGCACTGGTTTGTCCGC

AGTGTAATCGTCGTCTGCATCTGTATGCAGGCACC

GTTAAAGATCGTAAAGGCTATAAATACGATGTGC

GTCGCTATAAATGTGAAACCTGCAGCAAAAACAA

AGATGTGAAGAATGTGAGCTTCAACGAAAGCGAA

GTGGAAAACAAATTCGTCAATCTGCTGAAAAGCT

ACGAGCTGAACAAATTTCATATCCGTAAAGTGGA

ACCGGTGAAAAAAATCGAGTATGACATCGATAAG

ATTAACAAACAGAAAATTAACTATACCCGCAGTT

GGAGCCTGGGCTATATTGAAGATGATGAATATTT

CGAGCTGATGGAAGAAATCAACGCCACCAAAAA

AATGATCGAAGAACAGACCACCGAGAATAAACA

GAGCGTTAGCAAAGAGCAGATTCAGAGCATTAAC

AACTTTATCCTGAAAGGCTGGGAAGAACTGACCA

TCAAAGATAAAGAGGAACTGATTCTGAGCACCGT Part Name Type DNA sequence SEQ ID NO.

GGATAAAATCGAATTTAACTTCATCCCGAAAGAT AAAAAACATAAAACCAATACCCTGGATATTAACA ATATTCACTTTAAATTCTAA

GH022 RBS CATATAAAAGAAAAGACACC 168

GH023 RBS GAAATAAAGACATATAAAAGAAAAGACACC 169

GH078 RBS AAAAGGATCTATTATAAGGAGGCACTCACC 170

AATAGGCCTTTCGGTCCACACTCTCTATAGGCAA

RC500 RBS 171

A

CGCATTTTAAAATAAAATAAATTATTTATTTAATT

rpiL* RBS 172

AAACGAAT

Table 3.

It should be understood that the pNBU2 vector described herein may be substituted with pNBUl(SEQ ID NO: 209), provided herein. Thus, any of any of the components provided in Tables 1-3 and 5, for example, may be used in an pNBUl or pNBU2 vector backbone. In some embodiments, a pNBUl backbone is used instead of the pNBU2 backbone for any one or more of the constructs described in Table 4. Table 4.

Identifier Plasmid Relevant Features

NanoLuc expressed constitutively from PAMl, pNBU2 pAT751 pNBU2-PAMl -NanoLuc backbone, AmpR

NanoLuc expressed constitutively from PAM2, pNBU2 pAT752 pNBU2-PAM2-NanoLuc backbone, AmpR

NanoLuc expressed constitutively from PAM3, pNBU2 pAT753 pNBU2-PAM3-NanoLuc backbone, AmpR

NanoLuc expressed constitutively from PAM4, pNBU2 pAT754 pNBU2-PAM4-NanoLuc backbone, AmpR

NanoLuc expressed constitutively from PBT1311 and pAT587 pNBU2-PBT 1311 -GH022-NanoLuc GH022 RBS, pNBU2 backbone, AmpR

NanoLuc expressed constitutively from PBT1311 and pAT588 pNBU2-PBT1311-GH023 -NanoLuc GH023 RBS, pNBU2 backbone, AmpR

NanoLuc expressed constitutively from PBT1311 and pAT590 pNBU2-PBT 1311 -GH049-NanoLuc GH049 RBS, pNBU2 backbone, AmpR

NanoLuc expressed constitutively from PBT1311 and pAT593 pNBU2-PBT 1311 -rpiL*-NanoLuc rpiL* RBS, pNBU2 backbone, AmpR

NanoLuc expressed constitutively from PBT1311 and pAT695 pNBU2-PBT 1311 -RC500-NanoLuc RC500 RBS, pNBU2 backbone, AmpR

NanoLuc expressed constitutively from PAMl and pAT772 pNBU2-PAMl-GH078-NanoLuc GH078 RBS, pNBU2 backbone, AmpR

NanoLuc expressed constitutively from PAMl and pAT773 pNBU2-P AM 1 -GH022-NanoLuc GH022 RBS, pNBU2 backbone, AmpR

NanoLuc expressed constitutively from PAMl and pAT774 pNBU2-PAMl-GH023-NanoLuc GH023 RBS, pNBU2 backbone, AmpR

NanoLuc expressed constitutively from PAMl and pAT775 pNBU2-PAMl-rpiL*-NanoLuc rpiL* RBS, pNBU2 backbone, AmpR

NanoLuc expressed constitutively from PAMl and pAT776 pNBU2-PAMl-RC500-NanoLuc RC500 RBS, pNBU2 backbone, AmpR

NanoLuc expressed constitutively from PAM2 and pAT779 pNBU2-PAM2-GH078-NanoLuc GH078 RBS, pNBU2 backbone, AmpR

NanoLuc expressed constitutively from PAM2 and pAT780 pNBU2-PAM2-GH022-NanoLuc GH022 RBS, pNBU2 backbone, AmpR

NanoLuc expressed constitutively from PAM2 and pAT781 pNBU2-PAM2-GH023-NanoLuc GH023 RBS, pNBU2 backbone, AmpR

NanoLuc expressed constitutively from PAM2 and pAT782 pNBU2-PAM2-rpiL*-NanoLuc rpiL* RBS, pNBU2 backbone, AmpR

NanoLuc expressed constitutively from PAM2 and pAT783 pNBU2-PAM2-RC500-NanoLuc RC500 RBS, pNBU2 backbone, AmpR

NanoLuc expressed constitutively from PAM3 and pAT786 pNBU2-PAM3-GH078-NanoLuc GH078 RBS, pNBU2 backbone, AmpR

NanoLuc expressed constitutively from PAM3 and pAT787 pNBU2-PAM3-GH022-NanoLuc GH022 RBS, pNBU2 backbone, AmpR

NanoLuc expressed constitutively from PAM3 and pAT788 pNBU2-PAM3-GH023-NanoLuc GH023 RBS, pNBU2 backbone, AmpR

NanoLuc expressed constitutively from PAM3 and pAT789 pNBU2-PAM3 -rpiL* -NanoLuc rpiL* RBS, pNBU2 backbone, AmpR

NanoLuc expressed constitutively from PAM3 and pAT790 pNBU2-PAM3-RC500-NanoLuc RC500 RBS, pNBU2 backbone, AmpR

NanoLuc expressed constitutively from PAM4 and pAT793 pNBU2-PAM4-GH078-NanoLuc GH078 RBS, pNBU2 backbone, AmpR

NanoLuc expressed constitutively from PAM4 and pAT794 pNBU2-PAM4-GH022-NanoLuc GH022 RBS, pNBU2 backbone, AmpR Identifier Plasmid Relevant Features

NanoLuc expressed constitutively from PAM4 and pAT795 pNBU2-PAM4-GH023-NanoLuc GH023 RBS, pNBU2 backbone, AmpR

NanoLuc expressed constitutively from PAM4 and pAT796 pNBU2-PAM4-rpiL*-NanoLuc rpiL* RBS, pNBU2 backbone, AmpR

NanoLuc expressed constitutively from PAM4 and pAT797 pNBU2-PAM4-RC500-NanoLuc RC500 RBS, pNBU2 backbone, AmpR

Int7 expressed constitutively from PAM4 and the rpiL* pAT890 pNBU2-PAM4-rpiL*-int7 RBS, pNBU2 backbone, AmpR

Int8 expressed constitutively from PAM4 and the rpiL* pAT891 pNBU2-PAM4-rpiL*-int8 RBS, pNBU2 backbone, AmpR

Int9 expressed constitutively from PAM4 and the rpiL* pAT892 pNBU2-PAM4-rpiL*-int9 RBS, pNBU2 backbone, AmpR

Intl2 expressed constitutively from PAM4 and the pAT895 pNBU2-PAM4-rpiL*-intl2 rpiL* RBS, pNBU2 backbone, AmpR

Memory arrary integration vector for insertion between BT2113 and BT2114 in the B. thetaiotaomicron pAT847 pExchange-tdk-BT2107 -MA chromosome, AmpR

Rhamnose-inducible Intl2 expression vector with pAT937 pNBU2-Prha-rpiL*C51-Intl2 rpiL*C51 RBS, pNBU2 backbone, AmpR

Table 5.

Table 6.

IntNl attP Site (SEQ ID NO: 207)

CTACGTTCAACCAAAAGAAATAATGACTTACTGCTATATTTTTTGCACGTGTGGGGAAAA TGTGGG GAAAATTCAAGCAAAAGAAAAAGCTAAGTATTGAACTATCAAATACTTAGCTTTCTTTCT TGTACC CAGACCCCGCATTTGAAATAATTAAAGTGGGGAAAATGTGGGTAAAAAGAAAAATGCGGA AAAA CGCCACAATTACACTGTATTTCAATATGTTATAATCCTATTAAATTTTAATCCAAGTTTA ATCGAAT TGCAAAATATTTAGCAGATGTGGGGAAAATGCTGGGGAAAATATTTATATTTGCAGCAGA GTAAA AT

IntNl coding sequence (SEQ ID NO: 208)

ATGAAAGTAACCTTTATCATTAAAAAAGCAGCCAAACGATATGATACAGAATCCATGGCT ACAAT CTATGTCCGTTTTAGAAACGGAAGGCAGTTAGACTCCGTTGCTCCTACTCAGTTAGCCAT CAATCC CAATCTATGGGATGATAAAGACGAATGTGTAAAAACGAAAGCTGTCTGCAATGAAGAAAT GCGTA CCCATATAAATGAAGAGATACGCCAGTTGAAAACCTATATCGAGAAGGTATATCAACAAG AAAAG GAAGCAATAGACAAAGAATGGCTAAAAACAACACTTGATAAATTTTACCATCCTGAAAAA TATTT TTTGCCGGAGGAAGTGGTTATCAAGCCTACCATTGGAGAACTATTCGATGAATTTCTAAA CAAGCA CCCTTTGTCGGAAGTACGAAAGAAAAATTTCCGGGTTGTCAAAAGAGCCTTACTGCGTTA TGAACT ATATGTAAGGGCTACAAAGAGAGGACAAAAGGGCTTTATCCTTGATGTGGATTTGGTAAC ACCTG ACACGCTTCGGGATATGTGGGATTTCTTTCAGAACGAATACCAGTATTATGAACTTTACC CGAGCA TTTATGAAGCCATTCCCGAAAAGAGGACACCACAGCCCAGAAGCAAAAACACGCTGATAG ACTGT TTTTCAAGAATACGCACATTCTTCCTGTGGTGCTTCGATAACAAACGCACCACAAACAGA CCTTTC GACAAGTTTCCGATAGAGGAGTGTACATATGGTACACCTTATTATATAACACTCGAAGAA AGGGA CAGGATTTTTAATGCAGACCTTTCTGCCACCCCACAACTGGCAATACAGAGGGATATATT CATATT TCAGACACTGATAGGATGCAGGGTGAGCGACCTGTACCGAATGACCAAACTAAATGTGGT CAATG AAGCCATAGAATATATTCCCAAGAAAACCAAAGAGGGGAATCCGGTTACGGTACGTGTTC CACTT AACGACAAAGCGAAAGAAATCCTTGAACGCTACAAAGAATATGAGGGAAAACTGTTGCCG TTCAT ATCCGAGCAAAAGTACAATGATGCCATAAAAAAGATATTCAAATTAGCTGGAGTTGACCG CATCG TAACAATCTTAGACCCGTTGACGCACAACGAAATCAAACGACCTATTTATGAAGTGGCAA GCAGC CATCTGGCAAGACGTACGTTTATCGGCAATATCTATAAAAAAGTGAAAGACCCGAACCTT GTTTCC GCACTGTCGGGACACAAGGAGGGAAGCAAAGCTTTCAGACGATACAGGGATATTGACGAA GAAA TGAAGAAAGACCTTGTAAAACTACTGGACTGA pNBUl-L23R-NL (SEQ ID NO: 209)

ATGGTTTTTACTCTGGAAGATTTTGTTGGCGATTGGCGTCAGACCGCGGGTTATAATTTG GATCAA GTCCTGGAACAGGGTGGCGTAAGCTCTCTGTTCCAGAACCTGGGTGTGAGCGTGACGCCG ATTCAG CGCATCGTTCTGTCCGGCGAGAACGGTCTGAAAATTGATATTCATGTGATCATCCCGTAC GAAGGC CTGAGCGGTGACCAAATGGGTCAAATCGAGAAAATCTTTAAAGTCGTCTACCCAGTTGAC GATCA CCACTTCAAGGTTATCTTGCATTACGGTACGCTGGTGATTGATGGTGTGACCCCGAATAT GATTGA CTATTTCGGCCGTCCGTATGAAGGCATTGCCGTTTTTGACGGTAAAAAGATCACCGTCAC CGGTAC CCTGTGGAATGGCAATAAGATTATTGACGAGCGTCTGATTAACCCGGACGGCAGCCTGCT GTTCCG CGTGACCATCAACGGTGTCACGGGTTGGCGTCTGTGCGAGCGCATCCTGGCATAATGAAC TGCACT TGCTTTGATAATTAATGATAAACAATCTAAAAGCACTCTAATCGTTATCGGAGTGCTTTT AGATTA CTAATCAAATTGCTTCTACTAATTGCCTATCTTCCAGTGATGGAACAGCATTTGTGCATT GGCTGCA ACAATCAGCCTTGATCTGGAAGAAGCAATGAAAGCTGCTGTTAAGTCTCCGAATCAGGTA TTGTTC CTGACAGGTGTATTCCCATCCGGTAAACGCGGATACTTTGCAGTTGATCTGACTCAGGAA TAAATT ATAAATTAAGGTAAGAAGATTGTAGGATAAGCTAATGAAATAGAAAAAGGATGCCGTCAC ACAAC TTGTCGGCATTCTTTTTTGTTTTATTAGTTGAAAATATAGTGAAAAAGTTGCCTAAATAT GTATGTT AACAAATTATTTGTCGTAACTTTGCACTCCAAATCTGTTTTTAACATATGGCACTAGTGG TGAATGT GAAACCAGTAACGTTATACGATGTCGCAGAGTATGCCGGTGTCTCTTATCAGACCGTTTC CCGCGT GGTGAACCAGGCCAGCCACGTTTCTGCGAAAACGCGGGAAAAAGTGGAAGCGGCGATGGC GGAG CTGAATTACATTCCCAACCGCGTGGCACAACAACTGGCGGGCAAACAGTCGTTGCTGATT GGCGTT GCCACCTCCAGTCTGGCCCTGCACGCGCCGTCGCAAATTGTCGCGGCGATTAAATCTCGC GCCGAT CAACTGGGTGCCAGCGTGGTGGTGTCGATGGTAGAACGAAGCGGCGTCGAAGCCTGTAAA GCGGC GGTGCACAATCTTCTCGCGCAACGCGTCAGTGGGCTGATCATTAACTATCCGCTGGATGA CCAGGA TGCCATTGCTGTGGAAGCTGCCTGCACTAATGTTCCGGCGTTATTTCTTGATGTCTCTGA CCAGACA CCCATCAACAGTATTATTTTCTCCCATGAGGACGGTACGCGACTGGGCGTGGAGCATCTG GTCGCA TTGGGTCACCAGCAAATCGCGCTGTTAGCGGGCCCATTAAGTTCTGTCTCGGCGCGTCTG CGTCTG GCTGGCTGGCATAAATATCTCACTCGCAATCAAATTCAGCCGATAGCGGAACGGGAAGGC GACTG GAGTGCCATGTCCGGTTTTCAACAAACCATGCAAATGCTGAATGAGGGCATCGTTCCCAC TGCGAT GCTGGTTGCCAACGATCAGATGGCGCTGGGCGCAATGCGCGCCATTACCGAGTCCGGGCT GCGCG TTGGTGCGGATATCTCGGTAGTGGGATACGACGATACCGAGGACAGCTCATGTTATATCC CGCCGT TAACCACCATCAAACAGGATTTTCGCCTGCTGGGGCAAACCAGCGTGGACCGCTTGCTGC AACTCT CTCAGGGCCAGGCGGTGAAGGGCAATCAGCTGTTGCCCGTCTCACTGGTGAAAAGAAAAA CCACC CTGGCGCCCAATACGCAAACCGCCTCTCCCCGCGCGTTGGCCGATTCATTAATGCAGCTG GCACGA CAGGTTTCCCGACTGGAAAGCGGGCAGTGAGCTTTCCTCGGTACCAAATTCCAGAAAAGA GGCCT CCCGAAAGGGGGGCCTTTTTTCGTTTTGGTCCTACTTGTGCCTGTTCTATTTCCGAACCG ACCGCTT GTATGAATCCATCAAAATTCGTTTTCTCTATGTTGGATTCCTTGTTGCTCATATTGTGAT GATAATTT CTACAAATATAGTCATTGGTAACTATCTATGAAACTGTTTGATACTTTTATCAGTCCAGT AGTTTTA CAAGGTCTTTCTTCATTTCTTCGTCAATATCCCTGTATCGTCTGAAAGCTTTGCTTCCCT CCTTGTGT CCCGACAGTGCGGAAACAAGGTTCGGGTCTTTCACTTTTTTATAGATATTGCCGATAAAC GTACGT CTTGCCAGATGGCTGCTTGCCACTTCATAAATAGGTCGTTTGATTTCGTTGTGCGTCAAC GGGTCTA AGATTGTTACGATGCGGTCAACTCCAGCTAATTTGAATATCTTTTTTATGGCATCATTGT ACTTTTG CTCGGATATGAACGGCAACAGTTTTCCCTCATATTCTTTGTAGCGTTCAAGGATTTCTTT CGCTTTG TCGTTAAGTGGAACACGTACCGTAACCGGATTCCCCTCTTTGGTTTTCTTGGGAATATAT TCTATGG CTTCATTGACCACATTTAGTTTGGTCATTCGGTACAGGTCGCTCACCCTGCATCCTATCA GTGTCTG AAATATGAATATATCCCTCTGTATTGCCAGTTGTGGGGTGGCAGAAAGGTCTGCATTAAA AATCCT GTCCCTTTCTTCGAGTGTTATATAATAAGGTGTACCATATGTACACTCCTCTATCGGAAA CTTGTCG AAAGGTCTGTTTGTGGTGCGTTTGTTATCGAAGCACCACAGGAAGAATGTGCGTATTCTT GAAAAA CAGTCTATCAGCGTGTTTTTGCTTCTGGGCTGTGGTGTCCTCTTTTCGGGAATGGCTTCA TAAATGC TCGGGTAAAGTTCATAATACTGGTATTCGTTCTGAAAGAAATCCCACATATCCCGAAGCG TGTCAG GTGTTACCAAATCCACATCAAGGATAAAGCCCTTTTGTCCTCTCTTTGTAGCCCTTACAT ATAGTTC ATAACGCAGTAAGGCTCTTTTGACAACCCGGAAATTTTTCTTTCGTACTTCCGACAAAGG GTGCTT GTTTAGAAATTCATCGAATAGTTCTCCAATGGTAGGCTTGATAACCACTTCCTCCGGCAA AAAATA TTTTTCAGGATGGTAAAATTTATCAAGTGTTGTTTTTAGCCATTCTTTGTCTATTGCTTC CTTTTCTT GTTGATATACCTTCTCGATATAGGTTTTCAACTGGCGTATCTCTTCATTTATATGGGTAC GCATTTC TTCATTGCAGACAGCTTTCGTTTTTACACATTCGTCTTTATCATCCCATAGATTGGGATT GATGGCT AACTGAGTAGGAGCAACGGAGTCTAACTGCCTTCCGTTTCTAAAACGGACATAGATTGTA GCCATG GATTCTGTATCATATCGTTTGGCTGCTTTTTTAATGATAAAGGTTACTTTCATAGACTTT CAGGTTG AATTTTACTCTGCTGCAAATATAAATATTTTCCCCAGCATTTTCCCCACATCTGCTAAAT ATTTTGC AATTCGATTAAACTTGGATTAAAATTTAATAGGATTATAACATATTGAAATACAGTGTAA TTGTGG CGTTTTTCCGCATTTTTCTTTTTACCCACATTTTCCCCACTTTAATTATTTCAAATGCCG GGTCTGGG TACAAGAAAGAAAGCTAAGTATTTGATAGTTCAATACTTAGCTTTTTCTTTTGCTTGAAT TTTCCCC ACATTTTCCCCACACGTGCAAAAAATATAGCAGTAAGTCATTATTTCTTTTGGTTGAACG TAGAGA GTAGCGATATTAAAAGAATCCGATGAGAAAAGACTAATATTTATCTATCCATTCAGTTTG ATTTTT CAGGACTTTACATCGTCCTGAAAGTATTTGTTGGTACCGGTACCGAGGACGCGTAAACAT TTACAG TTGCATGTGGCCTATTGTTTTTAGCCGTTAAATATTTTATAACTATTAAATAGCGATACA AATTGTT CGAAACTAATATTGTTTATATCATATATTCTCGCATGTTTTAAAGCTTTATTAAATTGAT TTTTTGTA AACAGTTTTTCGTACTCTTTGTTAACCCATTTCATTACAAAAGTTTCATATTTTTTTCTC TCTTTAAA TGCCATTTTTGCTGGCTTTCTTTTTAATACAATTAATGTGCTATCCACTTTAGGTTTTGG ATGGAAAT AATACCTAGGAATTTTTGCTAATATAGAAATATCTACCTCTGCCATTAACAGCAATGCTA GTGATC TGTTTGTATCTAATAACATTTTAGCAAAACCATATTCCACTATTAAATAACTTATTGTGG CTGAACT TTCAAAAACAATTTTTCGAATTATATTTGTGCTTATGTTGTAAGGTATGCTGCCAAATAT TTTATAT GGATTGTGGCTAGGAAATGTAAATTTCAGTATATCATCATTTACTATTTGATAGTTAGGA TAATTTA AGAGCTTATTACGAGTTACCTCACATAATTTAGAATCAATTTCTATCGCCGTTACAAAAT TACATCT CTTTACCAATCCAGCAGTAAAATGACCTTTCCCTGCACCTATTTCAAAGATGTTATCTTT TTCATCT AAACTTATGCAATTCATTATTTTTTCTATGTGATATTTTGAAGTAATAAAATTTTGACTA TCTTTTAT ATTTACTTTGTTCATTATAACCTCTCCTTAATTTATTGCATCTCTTTTCGAATATTTATG TTTTTTGAG AAAAGAACGTACTCATGGTTCATCCCGATATGCGTATCGGTCTGTATATCAGCAACTTTC TATGTG TTTCAACTACAATAGTCATCTATTCTCATCTTTCTGAGTCCACCCCCTGCAAAGCCCCTC TTTACGA CATAAAAATTCGGTCGGAAAAGGTATGCAAAAGATGTTTCTCTCTTTAAGAGAAACTCTT CGGGAT GCAAAAATATGAAAATAACTCCAATTCACCAAATTATATAGCGACTTTTTTACAAAATGC TAAAAT TTGTTGATTTCCGTCAAGCAATTGTTGAGCAAAAATGTCTTTTACGATAAAATGATACCT CAATATC AACTGTTTAGCAAAACGATATTTCTCTTAAAGAGAGAAACACCTTTTTGTTCACCAATCC CCGACT TTTAATCCCGCGGCCATGATTGAAAAAGGAAGAGTATGAGTATTCAACATTTCCGTGTCG CCCTTA TTCCCTTTTTTGCGGCATTTTGCCTTCCTGTTTTTGCTCACCCAGAAACGCTGGTGAAAG TAAAAGA TGCTGAAGATCAGTTGGGTGCACGAGTGGGTTACATCGAACTGGATCTCAACAGCGGTAA GATCC TTGAGAGTTTTCGCCCCGAAGAACGTTTTCCAATGATGAGCACTTTTAAAGTTCTGCTAT GTGGCG CGGTATTATCCCGTATTGACGCCGGGCAAGAGCAACTCGGTCGCCGCATACACTATTCTC AGAATG ACTTGGTTGAGTACTCACCAGTCACAGAAAAGCATCTTACGGATGGCATGACAGTAAGAG AATTA TGCAGTGCTGCCATAACCATGAGTGATAACACTGCGGCCAACTTACTTCTGACAACGATC GGAGG ACCGAAGGAGCTAACCGCTTTTTTGCACAACATGGGGGATCATGTAACTCGCCTTGATCG TTGGGA ACCGGAGCTGAATGAAGCCATACCAAACGACGAGCGTGACACCACGATGCCTGTAGCAAT GGCAA CAACGTTGCGCAAACTATTAACTGGCGAACTACTTACTCTAGCTTCCCGGCAACAATTAA TAGACT GGATGGAGGCGGATAAAGTTGCAGGACCACTTCTGCGCTCGGCCCTTCCGGCTGGCTGGT TTATTG CTGATAAATCTGGAGCCGGTGAGCGTGGGTCTCGCGGTATCATTGCAGCACTGGGGCCAG ATGGT AAGCCCTCCCGTATCGTAGTTATCTACACGACGGGGAGTCAGGCAACTATGGATGAACGA AATAG ACAGATCGCTGAGATAGGTGCCTCACTGATTAAGCATTGGTAACTGTCAGACCAAGTTTA CTCATA ACGCGTCAATTCGAGGGGGATCAATTCCGTGATAGGTGGGCTGCCCTTCCTGGTTGGCTT GGTTTC ATCAGCCATCCGCTTGCCCTCATCTGTTACGCCGGCGGTAGCCGGCCAGCCTCGCAGAGC AGGATT CCCGTTGAGCACCGCCAGGTGCGAATAAGGGACAGTGAAGAAGGAACACCCGCTCGCGGG TGGG CCTACTTCACCTATCCTGCCCGGCTGACGCCGTTGGATACACCAAGGAAAGTCTACACGA ACCCTT TGGCAAAATCCTGTATATCGTGCGAAAAAGGATGGATATACCGAAAAAATCGCTATAATG ACCCC GAAGCAGGGTTATGCAGCGGAAAACGGAATTGATCCGGCCACGATGCGTCCGGCGTAGAG GATCT GAAGATCAGCAGTTCAACCTGTTGATAGTACGTACTAAGCTCTCATGTTTCACGTACTAA GCTCTC ATGTTTAACGTACTAAGCTCTCATGTTTAACGAACTAAACCCTCATGGCTAACGTACTAA GCTCTC ATGGCTAACGTACTAAGCTCTCATGTTTCACGTACTAAGCTCTCATGTTTGAACAATAAA ATTAAT ATAAATCAGCAACTTAAATAGCCTCTAAGGTTTTAAGTTTTATAAGAAAAAAAAGAATAT ATAAG GCTTTTAAAGCTTTTAAGGTTTAACGGTTGTGGACAACAAGCCAGGGATGTAACGCACTG AGAAG CCCTTAGAGCCTCTCAAAGCAATTTTGAGTGACACAGGAACACTTAACGGCTGACATGGG AATTCC CCTCCACCGCGGTGGTTACAAAGAAAATTCGACAAACTGTTATTTTTCTATCTATTTATT TGAATTG TGAGCGGATAACAATTACCTTTGTCGGCAATTGTGAGCGGATAACAATTAAATAAAGATA TTCTCG TCAAACAAATATAAATAATATAAAC While several inventive embodiments have been described and illustrated herein, those of ordinary skill in the art will readily envision a variety of other means and/or structures for performing the function and/or obtaining the results and/or one or more of the advantages described herein, and each of such variations and/or modifications is deemed to be within the scope of the inventive embodiments described herein. More generally, those skilled in the art will readily appreciate that all parameters, dimensions, materials, and configurations described herein are meant to be exemplary and that the actual parameters, dimensions, materials, and/or configurations will depend upon the specific application or applications for which the inventive teachings is/are used. Those skilled in the art will recognize, or be able to ascertain using no more than routine experimentation, many equivalents to the specific inventive embodiments described herein. It is, therefore, to be understood that the foregoing embodiments are presented by way of example only and that, within the scope of the appended claims and equivalents thereto, inventive embodiments may be practiced otherwise than as specifically described and claimed. Inventive embodiments of the present disclosure are directed to each individual feature, system, article, material, kit, and/or method described herein. In addition, any combination of two or more such features, systems, articles, materials, kits, and/or methods, if such features, systems, articles, materials, kits, and/or methods are not mutually inconsistent, is included within the inventive scope of the present disclosure.

All definitions, as defined and used herein, should be understood to control over dictionary definitions, definitions in documents incorporated by reference, and/or ordinary meanings of the defined terms.

All references, patents and patent applications disclosed herein are incorporated by reference with respect to the subject matter for which each is cited, which in some cases may encompass the entirety of the document.

The indefinite articles "a" and "an," as used herein in the specification and in the claims, unless clearly indicated to the contrary, should be understood to mean "at least one."

The phrase "and/or," as used herein in the specification and in the claims, should be understood to mean "either or both" of the elements so conjoined, i.e., elements that are conjunctively present in some cases and disjunctively present in other cases. Multiple elements listed with "and/or" should be construed in the same fashion, i.e., "one or more" of the elements so conjoined. Other elements may optionally be present other than the elements specifically identified by the "and/or" clause, whether related or unrelated to those elements specifically identified. Thus, as a non-limiting example, a reference to "A and/or B", when used in conjunction with open-ended language such as "comprising" can refer, in one embodiment, to A only (optionally including elements other than B); in another embodiment, to B only (optionally including elements other than A); in yet another embodiment, to both A and B (optionally including other elements); etc.

As used herein in the specification and in the claims, the phrase "at least one," in reference to a list of one or more elements, should be understood to mean at least one element selected from any one or more of the elements in the list of elements, but not necessarily including at least one of each and every element specifically listed within the list of elements and not excluding any combinations of elements in the list of elements. This definition also allows that elements may optionally be present other than the elements specifically identified within the list of elements to which the phrase "at least one" refers, whether related or unrelated to those elements specifically identified. Thus, as a non-limiting example, "at least one of A and B" (or, equivalently, "at least one of A or B," or, equivalently "at least one of A and/or B") can refer, in one embodiment, to at least one, optionally including more than one, A, with no B present (and optionally including elements other than B); in another

embodiment, to at least one, optionally including more than one, B, with no A present (and optionally including elements other than A); in yet another embodiment, to at least one, optionally including more than one, A, and at least one, optionally including more than one, B (and optionally including other elements); etc.

It should also be understood that, unless clearly indicated to the contrary, in any methods claimed herein that include more than one step or act, the order of the steps or acts of the method is not necessarily limited to the order in which the steps or acts of the method are recited.

In the claims, as well as in the specification above, all transitional phrases such as "comprising," "including," "carrying," "having," "containing," "involving," "holding,"

"composed of," and the like are to be understood to be open-ended, i.e., to mean including but not limited to. Only the transitional phrases "consisting of and "consisting essentially of shall be closed or semi-closed transitional phrases, respectively, as set forth in the United States Patent Office Manual of Patent Examining Procedures, Section 2111.03.