Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
RECOMBINANTLY ENGINEERED CELLS EXPRESSING CHLORITE DISMUTASE AND METHODS FOR USING SAME IN CELL CULTURE
Document Type and Number:
WIPO Patent Application WO/2015/085199
Kind Code:
A1
Abstract:
The present disclosure relates to recombinantly engineered cells that contain a chlorite dismutase polypeptide and methods for culturing such cells in a culture medium containing chlorite in an amount sufficient to reduce the growth rate or kill contaminating microorganisms without killing the recombinantly engineered cells. Also provided are methods for the production of a fermentation product using the recombinantly engineered cells.

Inventors:
COATES JOHN D (US)
Application Number:
PCT/US2014/068842
Publication Date:
June 11, 2015
Filing Date:
December 05, 2014
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
UNIV CALIFORNIA (US)
International Classes:
C12N1/20; C12N9/02; C12N9/88; C12P7/06; C12P7/16
Domestic Patent References:
WO2012166964A12012-12-06
Foreign References:
US5891339A1999-04-06
Other References:
MARCIN LOS ET AL: "Bacteriophage contamination: is there a simple method to reduce its deleterious effects in laboratory cultures and biotechnological factories?", J. APPL. GENET, 1 January 2004 (2004-01-01), pages 111 - 120, XP055175614, Retrieved from the Internet [retrieved on 20150311]
FRANÇOISE BICHAI ET AL: "Assessing the Disinfecting Power of Chlorite in Drinking Water", WATER QUAL. RES. J. CANADA COPYRIGHT, 1 January 2006 (2006-01-01), pages 375 - 382, XP055175680, Retrieved from the Internet [retrieved on 20150311]
BENNETT R. STREIT ET AL: "Chemical and Steady-State Kinetic Analyses of a Heterologously Expressed Heme Dependent Chlorite Dismutase +", BIOCHEMISTRY, vol. 47, no. 19, 1 May 2008 (2008-05-01), pages 5271 - 5280, XP055177283, ISSN: 0006-2960, DOI: 10.1021/bi800163x
HEINEMANN I U ET AL: "The biochemistry of heme biosynthesis", ARCHIVES OF BIOCHEMISTRY AND BIOPHYSICS, ACADEMIC PRESS, US, vol. 474, no. 2, 15 June 2008 (2008-06-15), pages 238 - 251, XP022701458, ISSN: 0003-9861, [retrieved on 20080221], DOI: 10.1016/J.ABB.2008.02.015
Attorney, Agent or Firm:
WARD, Michael R. et al. (425 Market StreetSan Francisco, CA, US)
Download PDF:
Claims:
CLAIMS

We claim:

1. A method of culturing a cell, comprising: a) culturing a recombinant!}' engineered cell comprising a chlorite dismutase polypeptide in a culture medium under conditions whereby the chlorite dismutase polypeptide is expressed in said cell, wherein said culture medium may also include one or more contami noting microorgan ism s ; and

b) treating said culture medium with chlorite in an amount sufficient to reduce the

growth rate or kill said one or more contaminating microorganisms without killing said cell, therebv cullurins said ceil.

2. The method of claim 1 , wherein the culture medium is treated with chlorite prior to

culturing the cell.

3. The method of claim 1 , wherein the chlorite is produced in the culture medium by

electrochemical generation.

4. The method of claim 1, further comprising prior to step (a), adding the cell to the culture medium.

5. The method of claim 4, wherein the chlorite is added to the culture medium concurrently with the addition of the cell to the culture medium.

6. The method of claim 1 , wherein the culture medium is treated with the chlorite

intermittently.

7. The method of claim 1 , further comprising growing the cell in a bioreactor.

8. The method of claim 7, wherein the culture medium containing the cell is treated with chlorite prior to growing the cell in the bioreactor.

9. The method of claim 7, wherein the culture medium containing the cell is treated with chlorite while growing the cell in the bioreactor.

10. The method of any one of claims 1-9, wherein the cell is a bacterial cell, a yeast cell, or a fungal cell.

11. The method of any one of claims 1-9, wherein the cell is a bacterial cell.

12. The method of claim 11, wherein the bacterial cell is a Clostridium cell.

13. The method of any one of claims 1-9, wherein the cell is a yeast cell.

14. The method of any one of claims 1-9, wherein the cell is an animal cell.

15. The method of claim 14, wherein the animal cell is a mammalian or insect cell.

16. The method of any one of claims 1-15, wherein the cell does not endogenously express a chlorite dismutase.

17. The method of any one of claims 1-16, wherein the chlorite dismutase polypeptide is derived from a perchlorate reducing bacterium.

18. The method of any one of claims 1-16, wherein the chlorite dismutase polypeptide is at least 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 99%, or 100% identical to a chlorite dismutase polypeptide having a RefSeq accession number selected from the group consisting of YP_005026408.1, YP_285781.1, AAM92878.1, WP_014235269.1, AAT07043.1, WP 009867516.1, CAC14884.1, WP_013516316.1, ACA21503.1, YP 004267835.1, EFH80711.1, YP 004178041.1, YP 004367213.1, YP_004058724.1, and YP 004172359.1.

19. The method of any one of claims 1-18, wherein the cell further comprises the proteins necessary to produce a fermentation product.

20. The method of claim 19, wherein the cell is further recombinantly engineered to express one or more of the proteins necessary to produce a fermentation product.

21. The method of claim 19 or claim 20, further comprising culturing the cell under

conditions sufficient for the cell to produce a fermentation product.

22. The method of any one of claims 19-21, wherein the fermentation product is a hydrocarbon or hydrocarbon derivative.

23. The method of claim 22, wherein the hydrocarbon or hydrocarbon derivative comprises ethanol or butanol.

24. The method of any one of claims 1-23, wherein the cell further comprises one or more proteins necessary for heme biosynthesis.

25. The method of claim 24, wherein the cell is further recombinantly engineered to express the one or more proteins necessary for heme biosynthesis.

26. The method of claim 24 or claim 25, wherein the one or more proteins necessary for heme biosynthesis are selected from the group consisting of uroporphyrinogen III decarboxylase, coproporphyrinogen III oxidase, protoporphyrinogen IX oxidase, and ferrochelatase.

27. The method of any one of claims 1-26, wherein the one or more contaminating

microorganisms are selected from the group consisting of prokaryotic microorganisms, bacteria, archaea, eukaryotic microorganisms, fungal cells, yeast, algae, and

bacteriophages.

28. The method of any one of claims 1-27, wherein the one or more contaminating

microorganisms are bacteria.

29. The method of claim 28, wherein the bacteria is selected from the group consisting of

Lactobacillus, Clostridium, Pediococcus, Enterococcus, Acetohacter, and Gluconohacter.

30. The method of any one of claims 1-26, wherein the one or more contaminating

microorganisms are bacteriophages.

31. A recombinantly engineered cell comprising a chlorite dismutase polypeptide.

32. The cell of claim 31, wherein the cell is a bacterial cell, a yeast cell, or a fungal cell.

33. The cell of claim 31, wherein the cell is an animal cell.

34. The cell of claim 33, wherein the animal cell is a mammalian or insect cell.

35. The cell of any one of claims 31 -34. wherein the cell does not endogenously express a chlorite dismutase.

36. The cell of any one of claims 31 -35, wherein the chlorite dismutase polypeptide is

derived from a perchlorate reducing bacterium.

37. The cell of any one of claims 31 -35, wherein the chlorite dismutase polypeptide is at least 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 99%, or 100% identical to a chlorite dismutase polypeptide having a RefSeq accession number selected from the group consisting of YP_005026408.1, YP_285781.1,

AAM92878.1, WP_014235269.1, AAT07043.1, WP 009867516.1, CAC14884.1, WP_013516316.1, ACA21503.1, YP 004267835.1, EFH80711.1, YP 004178041.1,

YP 004367213.1, YP_004058724.1, and YP 004172359.1.

38. The cell of any one of claims 31-37, wherein the chlorite dismutase polypeptide is

expressed under the control of a constitutive promoter.

39. The ceil of any one of claims 1-37, wherein the chlorite dismutase polypeptide is

expressed under the control of an inducible promoter.

40. The ceil of any one of claims 31-39, wherein the ceil further comprises the proteins

necessary to produce a fermentation product.

41. The cell of claim 40, wherein the cell is further recombinantly engineered to express one or more of the proteins necessary to produce a fermentation product.

42. The cell of claim 40 or claim 41, wherein the fermentation product is a hydrocarbon or hydrocarbon derivative.

43. The cell of claim 42, wherein the hydrocarbon or hydrocarbon derivative comprises

ethanol or butanol.

44. The cell of any one of claims 31-43, wherein the cell further comprises one or more proteins necessary for heme biosynthesis.

45. The cell of claim 44, wherein the cell is further recombinantly engineered to express the one or more of the proteins necessary for heme biosynthesis.

46. The cell of claim 44 or claim 45, wherein the one or more proteins necessary for heme biosynthesis are selected from the group consisting of uroporphyrinogen III decarboxylase, coproporphyrinogen III oxidase, protoporphyrinogen IX oxidase, and ferrochelatase.

47. A culture medium comprising the cell of any one of claims 31 -46.

48. The culture medium of claim 47, further comprising chlorite ions.

49. The culture medium of claim 48, wherein the chlorite ions are at a concentration

sufficient to kill one or more microorganisms with which said cell is susceptible to contamination.

Description:
RECOMBINANTLY ENGINEERED CELLS EXPRESSING CHLORITE DISMUTASE AND METHODS FOR USING SAME IN CELL CULTURE

CROSS REFERENCES TO RELATED APPLICATIONS

[0001] This application claims the priority benefit of U.S. Provisional Application Serial No. 61/912,487, filed December 5, 2013, which is incorporated herein by reference in its entirety.

FIELD OF THE INVENTION

[0002] The present disclosure relates to recombinantly engineered cells containing a chlorite dismutase polypeptide, and methods of using such cells for culturing in a medium treated with chlorite to reduce the growth rate or kill contaminating microorganisms. The methods and compositions described herein may further be used to produce a fermentation product, such as a hydrocarbon, a hydrocarbon derivative, ethanol, or butanol.

BACKGROUND

[0003] Methods to manufacture fuels and other value-added chemicals through industrial biotechnology have the potential to improve these important industries by introducing greener and more sustainable processes. The use of biotechnologies to manufacture fuels and value- added chemicals can reduce the generation of waste and carbon dioxide emissions while improving energy efficiency, as compared to using fossil fuels (Tang, W.L. and Zhao, H.

(2009). Biotechnol J 4(12): 1725-39). Industrial biotechnology also represents a growing market; the World Economic Forum estimates that the revenue potential for bio-based energy, fuels, chemicals, and materials by the year 2020 is nearly $300 billion. New biotechnological advances are constantly expanding the capabilities of engineered biological systems to produce useful products.

[0004] One problem common to all biotechnological processes is hygiene. Biological contamination in industrial fermentation processes constitutes one of the most devastating threats to the productivity of the biotechnology facilities. There are several types of infections that can occur, including eukaryotic, prokaryotic, and bacteriophage. These infections are usually the result of poor process hygiene, whether they are due to operator failure or simply the impossible logistics associated with large scale processes. For example, due to global bacteriophage abundance, bioprocesses based on bacterial activities are in constant threat of bacteriophage infections (Los et al. (2004). J Appl Genet 45(1): 111 20).

[0005] With the planned development of industrial, fermentation-based biofuel production, the scale of bioprocesses is moving beyond anything previously conceived. Thus, there exists a need for improving hygienic methods for culturing cells and producing fermentation products.

BRIEF SUMMARY

[0006] In order to meet this need, the present disclosure provides methods and

compositions for engineering a cell to express a chlorite dismutase polypeptide, and culturing the cell in the presence of chlorite in an amount sufficient to kill contaminating

microorganisms while not killing the cultured cell. In one aspect, these methods and compositions are useful in improving industrial fermentation hygiene where the cultured cell produces a fermentation product, e.g., a hydrocarbon, hydrocarbon derivative, ethanol, or butanol. Moreover, the present disclosure is based at least in part on the surprising discovery that chlorite dismutase, a metabolic enzyme endogenously utilized by perchlorate reducing bacteria, can protect a host cell from the toxic effects of a chlorite-containing disinfectant.

[0007] Accordingly, certain aspects of the present disclosure relate to a method of culturin a cell, by: a) culturing a recombinantly engineered cell containing a chlorite dismutase polypeptide in a culture medium under conditions whereby the chlorite dismutase polypeptide is expressed in the cell, where the culture medium may also include one or more contarnmati microorganisms; and b) treating the culture medium w th chlorite in an amount sufficient to reduce the growth rate or kill the one or more contaminating microorganisms without killing the cell, thereby culturing the cell. [0008] In certain embodiments, the culture medium is treated with chlorite prior to culturing the cell. In certain embodiments, the chlorite is produced in the culture medium by electrochemical generation. In certain embodiments, the method further includes prior to step (a), adding the cell to the culture medium. In certain embodiments, the chlorite is added to the culture medium concurrently with the addition of the cell to the culture medium. In certain embodiments, the culture medium is treated with the chlorite intermittently. In certain embodiments, the method further includes growing the cell in a bioreactor. In certain embodiments, the culture medium containing the cell is treated with chlorite prior to growing the cell in the bioreactor. In certain embodiments, the culture medium containing the cell is treated with chlorite while growing the cell in the bioreactor. In certain embodiments that may be combined with any of the preceding embodiments, the cell is a bacterial cell, a yeast cell, or a fungal cell. In certain embodiments that may be combined with any of the preceding embodiments, the cell is a bacterial cell. In certain embodiments that may be combined with any of the preceding embodiments, the bacterial cell is a Clostridium cell. In certain embodiments that may be combined with any of the preceding embodiments, the cell is a yeast cell. In certain embodiments, the cell is an animal cell. In certain embodiments, the animal cell is a mammalian or insect cell. In certain embodiments that may be combined with any of the preceding embodiments, the cell does not endogenously express a chlorite dismutase. In certain embodiments that may be combined with any of the preceding embodiments, the chlorite dismutase polypeptide is derived from a perchlorate reducing bacterium. In certain embodiments that may be combined with any of the preceding embodiments, the chlorite dismutase polypeptide is at least 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 99%, or 100% identical to a chlorite dismutase polypeptide having a RefSeq accession number selected from YP_005026408.1, YP_285781.1,

AAM92878.1, WP_014235269.1, AAT07043.1, WP 009867516.1, CAC14884.1,

WP_013516316.1, ACA21503.1, YP 004267835.1, EFH80711.1, YP 004178041.1,

YP_004367213.1, YP_004058724.1, and YP_004172359.1. In certain embodiments that may be combined with any of the preceding embodiments, the cell further contains the proteins necessary to produce a fermentation product. In certain embodiments that may be combined with any of the preceding embodiments, the cell is further recombinantly engineered to express one or more of the proteins necessary to produce a fermentation product. In certain embodiments that may be combined with any of the preceding embodiments, the method further includes culturing the cell under conditions sufficient for the cell to produce a fermentation product. In certain embodiments that may be combined with any of the preceding embodiments, the fermentation product is a hydrocarbon or hydrocarbon derivative. In certain embodiments, the hydrocarbon or hydrocarbon derivative contains ethanol or butanol. In certain embodiments that may be combined with any of the preceding embodiments, the cell further contains one or more proteins necessary for heme biosynthesis. In certain

embodiments, the cell is further recombinantly engineered to express the one or more of the proteins necessary for heme biosynthesis. In certain embodiments that may be combined with any of the preceding embodiments, the one or more proteins necessary for heme biosynthesis are selected from uroporphyrinogen III decarboxylase, coproporphyrinogen III oxidase, protoporphyrinogen IX oxidase, and ferrochelatase. In certain embodiments that may be combined with any of the preceding embodiments, the one or more contaminating

microorganisms are selected from prokaryotic microorganisms, bacteria, archaea, eukaryotic microorganisms, fungal cells, yeast, algae, and bacteriophages. In certain embodiments that may be combined with any of the preceding embodiments, the one or more contaminating microorganisms are bacteria. In certain embodiments, the bacteria are selected from

Lactobacillus, Clostridium, Pediococcus, Enterococcus, Acelobacter, and Gluconobacter. In certain embodiments that may be combined with any of the preceding embodiments, the one or more contaminating microorganisms are bacteriophages.

[0009] Other aspects of the present disclosure relate to a recombinantly engineered cell containing a chlorite dismuta.se polypeptide.

[0010] In certain embodiments, the cell is a bacterial cell, a yeast cell, or a fungal cell. In certain embodiments, the cell is an animal cell. In certain embodiments, the animal cell is a mammalian or insect cell. In certain embodiments that may be combined with any of the preceding embodiments, the cell does not endogenously express a chlorite dismutase. In certain embodiments that may be combined with any of the preceding embodiments, the chlorite dismutase polypeptide is derived from a perchlorate reducing bacterium. In certain embodiments that may be combined with any of the preceding embodiments, the chlorite dismutase polypeptide is at least 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 99%, or 100% identical to a chlorite dismutase polypeptide having a RefSeq accession number selected from YP_005026408.1, YP_285781.1,

AAM92878.1, WP_014235269.1, AAT07043.1, WP 009867516.1, CAC14884.1,

WP_013516316.1, ACA21503.1, YP_004267835.1, EFH80711.1, YP_004178041.1,

YP_004367213.1, YP_004058724.1, and YP_004172359.1. In certain embodiments that may be combined with any of the preceding embodiments, the chlorite dismutase polypeptide is expressed under the control of a constitutive promoter. In certain embodiments that may be combined with any of the preceding embodiments, the chlorite dismutase polypeptide is expressed under the control of an inducible promoter. In certain embodiments that may be combined with any of the preceding embodiments, the cell further contains the proteins necessary to produce a fermentation product. In certain embodiments that may be combined with any of the preceding embodiments, the cell is further recombinantly engineered to express one or more of the proteins necessary to produce a fermentation product. In certain embodiments that may be combined with any of the preceding embodiments, the fermentation product is a hydrocarbon or hydrocarbon derivative. In certain embodiments that may be combined with any of the preceding embodiments, the hydrocarbon or hydrocarbon derivative contains ethanol or butanol. In certain embodiments that may be combined with any of the preceding embodiments, the cell further contains one or more proteins necessary for heme biosynthesis. In certain embodiments, the cell is further recombinantly engineered to express the one or more of the proteins necessary for heme biosynthesis. In certain embodiments that may be combined with any of the preceding embodiments, the one or more proteins necessary for heme biosynthesis are selected from uroporphyrinogen III decarboxylase,

coproporphyrinogen III oxidase, protoporphyrinogen IX oxidase, and ferrochelatase.

[0011] Other aspects of the present disclosure relate to a culture medium containing the cell of any of the preceding embodiments. In certain embodiments, the culture further includes chlorite ions. In certain embodiments, the chlorite ions are at a concentration sufficient to kill one or more microorganisms with which the cell is susceptible to contamination. DESCRIPTION OF THE FIGURES

[0012] FIG. 1 depicts a model of the respiratory pathway of perchlorate reduction in perchlorate reducing bacteria.

[0013] FIG. 2 depicts the structure of the conserved "core" of the perchlorate reduction genomic island (PRI). Four exemplary PRIs are shown (species of origin are as labeled); chlorite dismutase genes are labeled "cld."

[0014] FIG. 3 depicts the aerobic growth of Azospira suillum strain PS grown in the presence or absence of 40 μΜ chlorite. The graph demonstrates the effect of chlorite with respect to growth on a wild-type (wt) strain, or a matched strain lacking the chlorite dismutase gene {Acid). Four combinations are shown: wt strain grown in the absence of chlorite (dark circles), wt strain grown in the presence of 40 μΜ chlorite (open circles), Acid strain grown in the absence of chlorite (dark squares), and Acid strain grown in the presence of 40 μΜ chlorite (open squares).

[0015] FIG. 4 depicts the aerobic growth of Shewanella oneidensis strain MR-1 in the presence of 0, 250, 500, or 1000 μΜ chlorite. Strain RAM13 (circles) contains an empty expression vector, while strain ICC38 (squares) is recombinantly engineered to constitutively express a chlorite dismutase.

[0016] FIG. 5 demonstrates that culturing a bacterial strain expression chlorite dismutase in chlorite protects the strain from phage-mediated killing. The growth of Pseudomonas chloritidismutans (as shown by OD 6 oo nm ) over time is depicted for cultures grown in LB alone (squares), LB plus 60 μΜ chlorite (circles), LB plus 60 μΜ chlorite plus phage (triangles), LB plus heat-killed phage (asterisks), and LB plus phage (pluses).

[0017] FIG. 6 depicts the growth of wild-type Caulobacter crescentus bearing an empty plasmid (pJS14) in medium supplemented with ImM (pluses) or 4 mM chlorite (triangles), as compared to medium with no chlorite (circles).

[0018] FIG. 7 depicts the growth of wild-type Caulobacter crescentus bearing a plasmid expressing chlorite dismutase under the control of a xylose-inducible promoter (pJS14- xylX::cld) in medium supplemented with ImM (pluses) or 4 mM chlorite (triangles), as compared to medium with no chlorite (circles).

[0019] FIG. 8 shows the results of competition experiments where a chlorite dismutase- expressing Caulobacter strain (KR3557) and a non-chlorite dismutase -expressing Caulobacter strain (KR260) were co-cultured in the presence or absence of 0.5 mM chlorite. Growth of each strain was observed over time by measuring cfu/mL.

[0020] FIG. 9 plots the concentration of chlorite present in cultures over time. Cultures included a chlorite dismutase-expressing Caulobacter strain (KR3557), a non-chlorite dismutase -expressing Caulobacter strain (KR260), or a mixed culture of both strains.

[0021] FIG. 10 illustrates a set of centromere-based yeast plasmids designed to express chlorite dismutase at different levels.

[0022] FIG. 11 illustrates an exemplary pathway for heme biosynthesis in bacteria.

DETAILED DESCRIPTION

[0023] The following description sets forth exemplary methods, parameters and the like. It should be recognized, however, that such description is not intended as a limitation on the scope of the present disclosure but is instead provided as a description of exemplary embodiments.

[0024] The present disclosure relates generally to methods of culturing a cell, whereby the cell is recombinantly engineered to express a chlorite dismutase polypeptide and where the culture medium containing the cell is treated with chlorite dismutase to reduce the growth rate or kill contaminating microorganisms without killing the cultured cell. Further embodiments relate generally to the use of this method for the generation of a fermentation product. The present disclosure also relates to compositions including a cell recombinantly engineered to express a chlorite dismutase polypeptide. It also relates to compositions including a culture medium that contains a cell recombinantly engineered to express a chlorite dismutase polypeptide. [0025] In particular, the present disclosure is based, at least in part, on the surprising discovery that a host cell recombinantly engineered to express chlorite dismutase is protected from the toxic effects of chlorite by the activity of the chlorite dismutase. Furthermore, by recombinantly engineering a cell that would not normally express chlorite dismutase to express such an enzyme, the cell is surprisingly able to grow in culture medium containing a concentration of chlorite that would, without chlorite dismutase expression, kill the cell, prevent or slow its growth. Since chlorite is a common component in many disinfecting solutions, adding chlorite to culture medium containing a cell recombinantly engineered to express chlorite dismutase allows the recombinant cell to grow while reducing the growth, or killing, contaminating microorganisms.

[0026] Accordingly, the present disclosure provides methods and compositions for culturing a cell that is recombinantly engineered to express a chlorite dismutase in the presence of chlorite in an amount sufficient to reduce the growth rate or kill additional contaminating microorganism(s) without killing the recombinantly engineered cell. Further methods and compositions are provided for culturing the recombinantly engineered cell where the cell is also recombinantly engineered to produce a fermentation product. These methods and compositions allow the biological production of important fuels and other commodity chemicals with an improved level of process sanitation by reducing the growth rate or killing contaminating microorganisms without interfering with the recombinant cells generating a product.

Chlorite Dismutase

[0027] Certain aspects of the present disclosure relate to a cell that is recombinantly engineered to express a chlorite dismutase polypeptide. As used herein, "chlorite dismutase" (used interchangeably with "chlorite dismutase polypeptide") refers to an enzyme that catalyzes the reaction converting chlorite (C10 2 ~ ) into chloride (CI ) and oxygen (0 2 ). The term "chlorite 0(2)-lyase" may also be used interchangeably. The EC classification for chlorite dismutase is EC 1.13.11.49. In the present disclosure, any enzyme demonstrated or predicted to have the activity corresponding to EC classification 1.13.11.49 is considered to be a chlorite dismutase polypeptide. The GO term ID for chlorite dismutase is GO: 0050587. In the present disclosure, any protein demonstrated or predicted to have the cellular component, biological process, or molecular function corresponding to the GO term ID GO: 0050587 is considered to be a chlorite dismutase polypeptide. The Pfam protein family for chlorite dismutase is

PF06778. In the present disclosure, any protein annotated to belong to the family

corresponding to the Pfam classification PF06778 is considered to be a chlorite dismutase polypeptide.

[0028] The active site of a chlorite dismutase contains a heme group. The active enzyme is a multimeric complex of chlorite dismutase monomers, and enzymes have been purified with different amounts of multimers, from two to six monomers per complex. Enzymatically, chlorite dismutase is highly unique. Chlorite dismutase is one of only four known enzymatic systems able to catalyze covalent O-O bond formation (Bardiya N and Bae JH (2011).

Microbiol Res 166(4):237-54). The amino acid sequence of chlorite dismutase is also highly distinctive. For example, no other enzyme is more than 24% similar to the D. agitata chlorite dismutase (Bender et al. (2002). Appl Environ Microbiol 68(10):4820-6).

[0029] The active site of a chlorite dismutase contains a heme group. The active enzyme is a multimeric complex of chlorite dismutase monomers, and enzymes have been purified with different amounts of multimers, from two to six monomers per complex. Enzymatically, chlorite dismutase is highly unique. The amino acid sequence of chlorite dismutase is also highly distinctive; for example, no other enzyme is more than 24% similar to the D. agitata chlorite dismutase (Bender et al. (2002). Appl Environ Microbiol 68(10):4820-6).

[0030] Without wishing to be bound by theory, it is believed that chlorite is a strong oxidizing agent that is highly reactive and has antimicrobial and antiviral effects due to its oxidation of cell membrane components or viral proteins, leading to cell lysis or denaturation of viral proteins. Chloride ions, by contrast, are non-reactive and harmless. Chloride ions are used for a variety of cellular processes: e.g. , regulation of pH and organic solute transport through chloride channel proteins. In some embodiments, chlorite dismutase polypeptides of the present disclosure can localize to the outer cell membrane of a host cell of the present disclosure. Without wishing to be bound to theory, the expression of chlorite dismutase on the outer membrane of a cell is thought to enable the cell to convert harmful chlorite anions present in a culture medium into harmless chloride and oxygen before the chlorite is able to oxidize cellular components and reduce the growth rate or kill the cell. In some embodiments, the present disclosure employs chlorite dismutase activity to demonstrate a detoxification function that may be utilized through recombinant engineering to improve culturing processes, such as fermentation sanitation, among other applications.

[0031] In some embodiments, suitable chlorite dismutase polypeptides of the present disclosure may be obtained from perchlorate reducing bacteria. "Perchlorate reducing bacteria," or "PRB," as used herein refers to any prokaryotic microorganism that possesses an endogenous capability of metabolizing a (per)chlorate anion into chloride. All known PRBs contain a gene encoding a chlorite dismutase polypeptide. Chlorite dismutase polypeptides have been found in at least 13 bacterial and 3 archaeal phyla (Goblirsch, B. et al. J Mol Biol 408(3):379-98 (2011)), and over 50 PRB species have been isolated (Coates, J. D. &

Achenbach, L. A. Nat Rev Micro 2, 569-580 (2004)). In addition, several non-PRB species have also been found to contain a chlorite dismutase polypeptide. All known PRBs are facultative anaerobes or microaerophilic. They represent a phylogenetically diverse set of microorganisms within the phylum of Proteobacteria. PRBs have been found in both natural and human-contaminated environments, including soil, hot springs, wastewater treatment plants, animal waste lagoons, and even Antarctica (Coates, J. D. & Achenbach, L. A. Nat Rev Micro 2, 569-580 (2004); Bardiya N and Bae JH (2011). Microbiol Res 166(4):237-54). PRBs use perchlorate or chlorate reduction as part of their endogenous respiration, completely oxidizing an organic carbon source (most commonly acetate but sugars, lactate, pyruvate and others have been described) to generate carbon dioxide and water. In this process,

(per)chlorate is used as an electron acceptor. All of the genes necessary for (per)chlorate reduction, including the enzymatic components, accessory components, and regulatory components, are encoded by genes clustered together on a highly conserved perchlorate reduction genomic island (PRI) in perchlorate reducing microorganisms or a chlorate reduction composite transposon interior (CRI) in chlorate reducing microorganisms that are thought to have spread among these organisms through horizontal gene transfer (Melnyk RA et al. (2011). Appl Environ Microbiol 77(20):7401-4; Clark IC et al. (2013) mBio 4:e00379-13). [0032] Examples of suitable chlorite dismutase polypeptides include, without limitation, those listed in Table 1 , and homologs thereof.

Table 1

NCBI GI Accession Number NCBI RefSeq Accession Number

372486843 YP_005026408.1

71908194 YP_285781.1

504001275 WP_014235269.1

22212277 AAM92878.1

11121510 CAC14884.1

46948122 AAT07043.1

168203360 ACA21503.1

325106767 YP_004267835.1

320102450 YP_004178041.1

328949878 YP_004367213.1

313680985 YP_004058724.1

320335648 YP_004172359.1

161723183 NP_244692.2

10176450 BAB07544.1

161511066 NP_391647.2

384177417 YP_005558802.1

350268047 YP_004879354.1

221311731 ZP_03593578.1

321313325 YP_004205612.1

398305248 ZP_10508834.1

398308738 ZP_10512212.1

296331406 ZP_06873878.1

57866176 YP_187831.1

394231816 EJD77439.1

394250268 EJD95462.1

365225074 EHM66327.1

27467276 NP_763913.1

394237370 EJD82862.1

242241926 ZP .04796371.1

251810009 ZP_04824482.1

365231827 EHM72844.1

374820240 EHR84334.1

319650951 ZP_08005086.1 NCBI GI Accession Number NCBI RefSeq Accession Number

52082283 YP_081074.1

387591609 EIJ83926.1

288554459 YP_003426394.1

294501915 YP_003565615.1

384044252 YP_005492269.1

295707263 YP_003600338.1

394284803 EJE28902.1

154687885 YP_001423046.1

384267298 YP_005423005.1

389572462 ZP_10162547.1

242372811 ZP_04818385.1

373855300 ZP_09598046.1

387928340 ZP_10131018.1

384161381 YP_005543454.1

149182787 ZP_01861250.1

384166285 YP_005547664.1

308175490 YP_003922195.1

314932813 ZP_07840182.1

223043041 ZP_03613089.1

194016490 ZP_03055104.1

157694165 YP_001488627.1

341596274 EGS38890.1

311070287 YP_003975210.1

205375386 ZP_03228175.1

341843773 EGS84994.1

365156917 ZP_09353202.1

49482817 YP_040041.1

341842114 EGS83547.1

57651462 YP_185518.1

253732974 ZP_04867139.1

258422774 ZP_05685677.1

15923577 NP_371111.1

21282271 NP_645359.1

317130810 YP_004097092.1

374397432 EHQ68642.1

152977571 YP_001377088.1

386830232 YP_006236886.1 NCBI GI Accession Number NCBI RefSeq Accession Number

334276682 EGL94935.1

385780852 YP_005757023.1

82750292 YP_416033.1

56965670 YP_177404.1

358051694 ZP_09145835.1

239636797 ZP_04677799.1

330685907 EGG97536.1

384546866 YP_005736119.1

228994103 ZP_04154003.1

401726885 EJT00094.1

229000173 ZP_04159742.1

379795059 YP_005325057.1

311032064 ZP_07710154.1

229087876 ZP_04219988.1

70727404 YP_254320.1

386715945 YP_006182269.1

323489299 ZP 08094531.1

299536034 ZP_07049352.1

389816722 ZP_10207674.1

126650921 ZP_01723132.1

212640580 YP_002317100.1

224475733 YP_002633339.1

251798799 YP_003013530.1

312112678 YP_003990994.1

336237140 YP_004589756.1

386728346 YP_006194729.1

314937089 ZP_07844436.1

228475108 ZP_04059835.1

374823144 EHR87147.1

393202157 YP_006463999.1

163943070 YP_001647954.1

401311354 EJS16661.1

206970225 ZP_03231178.1

229020608 ZP_04177344.1

392958928 ZP_10324425.1

218906569 YP_002454403.1

401135057 EJQ42663.1 NCBI GI Accession Number NCBI RefSeq Accession Number

253575332 ZP_04852670.1

229124893 ZP_04254069.1

401648378 EJS65974.1

229034026 ZP_04188974.1

401174736 EJQ81943.1

229153549 ZP_04281727.1

229099815 ZP_04230739.1

30023418 NP_835049.1

228924134 ZP_04087410.1

228903866 ZP 04067981.1

228968520 ZP_04129508.1

42784562 NP_981809.1

228988615 ZP 04148701.1

30265410 NP_847787.1

75758554 ZP_00738674.1

118480419 YP_897570.1

401795731 AFQ09590.1

340355774 ZP_08678448.1

401193405 EJR00411.1

374602821 ZP_09675809.1

239828647 YP_002951271.1

289551609 YP_003472513.1

401241592 EJR47979.1

401093093 EJQ01212.1

354584844 ZP_09003736.1

401252388 EJR58649.1

229065036 ZP_04200333.1

401138051 EJQ45626.1

315649374 ZP_07902463.1

401643508 EJS61205.1

152112371 A4ITP5.2

138896991 YP_001127444.1

196040963 ZP_03108260.1

401077984 EJP86305.1

229014553 ZP_04171669.1

222152077 YP_002561237.1

261409225 YP_003245466.1 NCBI GI Accession Number NCBI RefSeq Accession Number

329926866 ZP_08281269.1

319891573 YP_004148448.1

386320088 YP_006016251.1

347751826 YP_004859391.1

374710660 ZP_09715094.1

52695600 1T0T

319768491 YP_004133992.1

56421951 YP_149269.1

297531616 YP_003672891.1

261420821 YP_003254503.1

375010605 YP_004984238.1

301056856 YP_003795067.1

336115528 YP_004570295.1

73663434 YP_302215.1

297585491 YP_003701271.1

365233554 EHM74501.1

347549512 YP_004855840.1

16801283 NP .471551.1

255022443 ZP_05294429.1

23100478 NP_693945.1

217963726 YP_002349404.1

46908349 YP_014738.1

16804152 NP_465637.1

290894126 ZP_06557098.1

386732857 YP_006206353.1

116873548 YP_850329.1

399054122 ZP_10742752.1

400171111 ZP_10786628.1

365227200 EHM68402.1

381184708 ZP_09893243.1

315304220 ZP_07874585.1

308070609 YP_003872214.1

390455464 ZP_10240992.1

375310122 ZP_09775400.1

392971201 ZP_10336597.1

289435463 YP_003465335.1

310643795 YP_003948553.1 NCBI GI Accession Number NCBI RefSeq Accession Number

398812996 ZP_10571701.1

313632394 EFR99424.1

350547382 ZP_08916707.1

374320146 YP_005073275.1

226314140 YP_002774036.1

47570390 ZP 00241031.1

297191327 ZP_06908725.1

254818553 ZP_05223554.1

379755414 YP_005344086.1

334138528 ZP_08511946.1

296171936 ZP 06852981.1

371546895 EHN75321.1

21224373 NP_630152.1

357004127 ZP_09069126.1

294632032 ZP_06710592.1

302557663 ZP_07310005.1

381210903 ZP_09917974.1

392416039 YP_006452644.1

400536789 ZP_10800323.1

329940188 ZP_08289470.1

29828763 NP_823397.1

340627677 YP_004746129.1

41408895 NP_961731.1

291436567 ZP_06575957.1

386843184 YP_006248242.1

385995584 YP_005913882.1

291454875 ZP_06594265.1

289570850 ZP_06451077.1

15609813 NP_217192.1

289444218 ZP_06433962.1

313607606 EFR83886.1

299822310 ZP_07054196.1

290956576 YP_003487758.1

383823529 ZP_09978719.1

240169729 ZP_04748388.1

345021484 ZP_08785097.1

54025706 YP_119948.1 NCBI GI Accession Number NCBI RefSeq Accession Number

297203093 ZP_06920490.1

304408214 ZP_07389863.1

383638911 ZP_09951317.1

357021607 ZP_09083838.1

342306657 BAK54746.1

15922480 NP_378149.1

375142573 YP_005003222.1

395771103 ZP_10451618.1

389865542 YP_006367783.1

357402256 YP_004914181.1

294815684 ZP_06774327.1

254393128 ZP_05008286.1

374320324 YP_005073453.1

118468878 YP_887113.1

357410441 YP_004922177.1

15790882 NP_280706.1

383819186 ZP_09974462.1

172056260 YP_001812720.1

374608155 ZP_09680954.1

13541622 NP_111310.1

365861224 ZP_09400999.1

302554904 ZP_07307246.1

328885880 CCA59119.1

269126008 YP_003299378.1

345009959 YP_004812313.1

345855716 ZP_08808383.1

118618640 YP_906972.1

284990411 YP_003408965.1

295835898 ZP_06822831.1

374985838 YP_004961333.1

333919450 YP_004493031.1

386382344 ZP_10067963.1

372458364 CCF13558.1

378814713 ZP_09837647.1

302541889 ZP_07294231.1

345002765 YP_004805619.1

302537561 ZP_07289903.1 NCBI GI Accession Number NCBI RefSeq Accession Number

378717676 YP_005282565.1

239991583 ZP_04712247.1

326775901 ZP_08235166.1

182435262 YP_001822981.1

357392404 YP_004907245.1

15898949 NP_343554.1

291448583 ZP_06587973.1

359767828 ZP_09271610.1

398781574 ZP_10545606.1

385777371 YP_005649939.1

357009391 ZP_09074390.1

262202198 YP_003273406.1

379709823 YP_005265028.1

339007351 ZP_08639926.1

318061066 ZP_07979787.1

271967974 YP_003342170.1

259507404 ZP_05750304.1

229586215 YP_002844717.1

379736665 YP_005330171.1

25028348 NP_738402.1

309811739 ZP_07705517.1

145224498 YP_001135176.1

337747991 YP_004642153.1

353184711 EHB50235.1

15827508 NP_301771.1

359772082 ZP_09275520.1

315502680 YP_004081567.1

302866123 YP_003834760.1

111023818 YP_706790.1

120403473 YP_953302.1

375094985 ZP_09741250.1

699157 AAA62922.1

356666940 EHI47011.1

229917171 YP_002885817.1

335038759 ZP_08531969.1

111027153 YP_709131.1

108799181 YP_639378.1 NCBI GI Accession Number NCBI RefSeq Accession Number

169826530 YP_001696688.1

291301396 YP_003512674.1

384100724 ZP_10001781.1

377567646 ZP_09796854.1

365870907 ZP_09410448.1

388821964 EIM44646.1

226366254 YP_002784037.1

297561771 YP_003680745.1

363420411 ZP_09308503.1

384566547 ZP_10013651.1

377561630 ZP_09791073.1

119717893 YP_924858.1

134098414 YP_001104075.1

169630067 YP_001703716.1

348171321 ZP_08878215.1

377562766 ZP_09792134.1

343924651 ZP_08764195.1

15806494 NP_295204.1

257056399 YP_003134231.1

312139468 YP_004006804.1

16081621 NP_393983.1

331696710 YP_004332949.1

319948480 ZP_08022614.1

311742292 ZP_07716101.1

296270232 YP_003652864.1

317121335 YP_004101338.1

354617379 ZP_09034810.1

354610192 ZP_09028148.1

359427086 ZP_09218161.1

333991062 YP_004523676.1

226306336 YP_002766296.1

229491306 ZP_04385132.1

256375690 YP_003099350.1

145594081 YP_001158378.1

111024859 YP_707279.1

385674401 ZP_10048329.1

386690160 ZP_10088922.1 NCBI GI Accession Number NCBI RefSeq Accession Number

295695923 YP_003589161.1

72162295 YP_289952.1

330466519 YP_004404262.1

378584109 ZP_09832664.1

383831229 ZP_09986318.1

296394322 YP_003659206.1

238063535 ZP_04608244.1

54654366 AAV37056.1

313902589 ZP_07835989.1

300781043 ZP_07090897.1

317508457 ZP_07966126.1

84496260 ZP_00995114.1

302525787 ZP_07278129.1

159037112 YP_001536365.1

325002058 ZP_08123170.1

376242999 YP_005133851.1

375291064 YP_005125604.1

227549138 ZP_03979187.1

326384415 ZP_08206095.1

383781997 YP_005466564.1

256395904 YP_003117468.1

376290601 YP_005162848.1

38233979 NP_939746.1

227504962 ZP_03935011.1

387982647 EIK56148.1

386852019 YP_006270032.1

317125032 YP_004099144.1

376248683 YP_005140627.1

375101462 ZP_09747725.1

375293270 YP_005127809.1

296139603 YP_003646846.1

152965553 YP_001361337.1

359421112 ZP_09213042.1

376293404 YP_005165078.1

19553103 NP_601105.1

145295803 YP_001138624.1

334564734 ZP_08517725.1 NCBI GI Accession Number NCBI RefSeq Accession Number

284031650 YP_003381581.1

227503493 ZP_03933542.1

377575250 ZP_09804244.1

344044034 EGV39715.1

306836259 ZP 07469241.1

311739576 ZP_07713411.1

255325114 ZP_05366220.1

354511692 EHE84598.1

296117770 ZP_06836354.1

325283524 YP_004256065.1

296130245 YP_003637495.1

288917125 ZP_06411495.1

226355654 YP_002785394.1

336117965 YP_004572733.1

326332270 ZP_08198550.1

225021282 ZP_03710474.1

237785637 YP_002906342.1

357588794 ZP_09127460.1

397729455 ZP_10496235.1

111017350 YP_700322.1

227833265 YP_002834972.1

337290914 YP_004629935.1

336320168 YP_004600136.1

387138794 YP_005694773.1

383314378 YP_005375233.1

379715495 YP_005303832.1

386740528 YP_006213708.1

339628143 YP_004719786.1

300858614 YP_003783597.1

375288794 YP_005123335.1

392400729 YP_006437329.1

70605865 YP_254735.1

386855711 YP_006259888.1

300784551 YP_003764842.1

86740042 YP_480442.1

384515826 YP_005710918.1

76801774 YP_326782.1 NCBI GI Accession Number NCBI RefSeq Accession Number

356664861 EHI44943.1

332798007 YP_004459507.1

358445417 ZP_09156027.1

260906569 ZP 05914891.1

117928587 YP_873138.1

167465230 ZP_02330319.1

388821007 EIM43781.1

334337905 YP_004543057.1

399577847 ZP_10771599.1

256833104 YP_003161831.1

332669795 YP_004452803.1

312196140 YP_004016201.1

385802483 YP_005838883.1

295394782 ZP_06804997.1

158316929 YP_001509437.1

379057271 ZP_09847797.1

340794530 YP_004759993.1

336177818 YP_004583193.1

110667069 YP_656880.1

195927607 3DTZ

94985810 YP_605174.1

292656006 YP_003535903.1

284041622 YP_003391962.1

222480603 YP_002566840.1

68536150 YP_250855.1

358462858 ZP_09172965.1

218289621 ZP 03493841.1

302522536 ZP_07274878.1

258511732 YP_003185166.1

389847397 YP_006349636.1

213964421 ZP_03392621.1

313125252 YP_004035516.1

227488766 ZP_03919082.1

297627503 YP_003689266.1

333373995 ZP_08465888.1

48477890 YP_023596.1

386359782 YP_006058027.1 NCBI GI Accession Number NCBI RefSeq Accession Number

172039722 YP_001799436.1

297567364 YP_003686336.1

111221538 YP_712332.1

269794165 YP_003313620.1

229819585 YP_002881111.1

269957406 YP_003327195.1

116619486 YP_821642.1

392943943 ZP_10309585.1

146303506 YP_001190822.1

46199654 YP_005321.1

381191171 ZP_09898682.1

291295788 YP_003507186.1

300710235 YP_003736049.1

55670428 1VDH

401679226 ZP_10811159.1

336326041 YP_004606007.1

330835234 YP_004409962.1

303230308 ZP_07317073.1

384440419 YP_005655143.1

303228654 ZP_07315479.1

302038380 YP_003798702.1

256825385 YP_003149345.1

374632137 ZP 09704511.1

284107237 ZP_06386378.1

298252251 ZP_06976054.1

367471078 ZP_09470737.1

320449344 YP_004201440.1

225872477 YP_002753932.1

218296104 ZP_03496873.1

269839067 YP_003323759.1

342214592 ZP_08707276.1

385652938 ZP_10047491.1

383621121 ZP_09947527.1

108803590 YP_643527.1

257052298 YP_003130131.1

257387306 YP_003177079.1

148271773 YP_001221334.1 NCBI GI Accession Number NCBI RefSeq Accession Number

383806533 ZP_09962095.1

345005866 YP_004808719.1

395236980 ZP_10415106.1

340344386 ZP_08667518.1

167043725 ABZ08417.1

258651774 YP_003200930.1

313893828 ZP_07827394.1

344210645 YP_004794965.1

386772469 ZP_10094847.1

163839968 YP_001624373.1

322368317 ZP_08042886.1

118430948 NP_147071.2

386875231 ZP_10117418.1

365906153 ZP_09443912.1

384135398 YP_005518112.1

161723231 YP_137518.2

55232393 AAV47812.1

352125886 ZP_08969712.1

269928683 YP_003321004.1

170783142 YP_001711476.1

298526148 ZP_07013557.1

380300719 ZP_09850412.1

257069785 YP_003156040.1

282849314 ZP_06258699.1

294793131 ZP_06758277.1

325963980 YP_004241886.1

88854545 ZP_01129212.1

257075934 ZP_05570295.1

365850812 ZP 09391271.1

221635966 YP_002523842.1

359778406 ZP_09281675.1

323358315 YP_004224711.1

269797286 YP_003311186.1

333976464 EGL77331.1

294794831 ZP_06759966.1

336254860 YP_004597967.1

161528023 YP_001581849.1 NCBI GI Accession Number NCBI RefSeq Accession Number

390958054 YP_006421811.1

220913236 YP_002488545.1

390565895 ZP_10246467.1

116671315 YP_832248.1

118576599 YP_876342.1

238019896 ZP_04600322.1

315283182 ZP_07871432.1

119960759 YP_948465.1

352114985 ZP_08963219.1

329764724 ZP_08256319.1

394761213 EJF43622.1

393796958 ZP_10380322.1

289581178 YP_003479644.1

400976087 ZP_10803318.1

300741889 ZP_07071910.1

259502145 ZP_05745047.1

393796863 ZP_10380227.1

159900062 YP_001546309.1

89100748 ZP_01173602.1

311111782 YP_003983004.1

255326984 ZP_05368060.1

320106349 YP_004181939.1

161486757 YP_591030.2

283457694 YP_003362280.1

94551032 ABF40956.1

284166731 YP_003405010.1

397774405 YP_006541951.1

322435228 YP_004217440.1

269217809 ZP_06161663.1

227496736 ZP_03927010.1

383810118 ZP_09965625.1

395203147 ZP_10394381.1

374312300 YP_005058730.1

397670494 YP_006512029.1

326772613 ZP_08231897.1

313806438 EFS44945.1

313793129 EFS41196.1 NCBI GI Accession Number NCBI RefSeq Accession Number

313802783 EFS44001.1

315082765 EFT54741.1

327334886 EGE76597.1

313810987 EFS48701.1

313772848 EFS38814.1

50841796 YP_055023.1

379978901 EIA12225.1

295129873 YP_003580536.1

353343976 EHB88289.1

315082092 EFT54068.1

340774183 EGR96670.1

350568884 ZP_08937282.1

343522997 ZP_08759962.1

340358532 ZP_08681049.1

308177933 YP_003917339.1

313821947 EFS59661.1

313765238 EFS36602.1

313828705 EFS66419.1

365973206 YP_004954765.1

365962026 YP_004943592.1

400293990 ZP_10795815.1

319760122 YP_004124061.1

327329287 EGE71047.1

386070848 YP_005985744.1

282853365 ZP_06262702.1

320532492 ZP_08033313.1

239918009 YP_002957567.1

342212563 ZP_08705288.1

365829316 ZP_09371052.1

293189110 ZP_06607836.1

329945534 ZP_08293272.1

289705148 ZP_06501552.1

325066698 ZP 08125371.1

154509628 ZP_02045270.1

399523435 ZP_10764076.1

184200497 YP_001854704.1

313814508 EFS52222.1 NCBI GI Accession Number NCBI RefSeq Accession Number

396585876 ZP_10486044.1

148655882 YP_001276087.1

378551105 ZP 09826321.1

365825602 ZP_09367556.1

156742004 YP_001432133.1

315604999 ZP_07880053.1

399527499 ZP_10767199.1

307635429 ADN79128.1

46202510 ZP_00208549.1

316983367 3Q08

224510506 2VXH

168203364 ACA21505.1

373252940 ZP_09541058.1

381398954 ZP_09924225.1

307635431 ADN79129.1

349806848 AEQ19297.1

196230396 ZP_03129258.1

397737933 ZP_10504580.1

389798300 ZP_10201323.1

352090357 ZP_08954468.1

389811160 ZP_10206102.1

171911071 ZP 02926541.1

255513657 EET89922.1

28493701 NP_787862.1

223935944 ZP_03627859.1

373482185 ZP_09572994.1

384915640 ZP_10015852.1

269986639 EEZ92920.1

335050425 ZP 08543391.1

290558840 EFD92232.1

290559836 EFD93159.1

87311298 ZP_01093420.1

301598730 3NN1

301598745 3NN4

301598740 3NN3

327542310 EGF28797.1

302036741 YP_003797063.1 NCBI GI Accession Number NCBI RefSeq Accession Number

149176472 ZP_01855085.1

161579027 NP_869234.2

189218926 YP_001939567.1

255018722 ZP_05290848.1

32446784 CAD76620.1

296124023 YP_003631801.1

283778293 YP_003369048.1

227876575 ZP_03994685.1

269977766 ZP_06184726.1

315657647 ZP_07910529.1

315654427 ZP_07907335.1

298345852 YP_003718539.1

256372606 YP_003110430.1

262384993 ACY64494.1

46948114 AAT07039.1

262385037 ACY64516.1

262384991 ACY64493.1

262384971 ACY64483.1

262385029 ACY64512.1

262384973 ACY64484.1

262385059 ACY64527.1

262385013 ACY64504.1

46948116 AAT07040.1

46948118 AAT07041.1

46948120 AAT07042.1

363805181 AEW31185.1

363805179 AEW31184.1

78217052 ABB36663.1

262385079 ACY64537.1

363805183 AEW31186.1

346987801 AE051751.1

78217078 ABB36674.1

78217063 ABB36667.1

78217076 ABB36673.1

78217080 ABB36675.1

78217071 ABB36671.1

262384985 ACY64490.1 NCBI GI Accession Number NCBI RefSeq Accession Number

262384943 ACY64469.1

262384965 ACY64480.1

262384995 ACY64495.1

262384967 ACY64481.1

262384969 ACY64482.1

262385017 ACY64506.1

262385031 ACY64513.1

346987805 AE051753.1

262384981 ACY64488.1

78217060 ABB36666.1

78217082 ABB36676.1

262384945 ACY64470.1

262385069 ACY64532.1

262384975 ACY64485.1

262384953 ACY64474.1

262384947 ACY64471.1

262385065 ACY64530.1

262385057 ACY64526.1

262384955 ACY64475.1

262385011 ACY64503.1

262384949 ACY64472.1

262385083 ACY64539.1

262385055 ACY64525.1

262385073 ACY64534.1

262384989 ACY64492.1

262385023 ACY64509.1

46948132 AAT07048.1

78217067 ABB36669.1

262385003 ACY64499.1

78217050 ABB36662.1

46948130 AAT07047.1

262384977 ACY64486.1

78217065 ABB36668.1

262384951 ACY64473.1

262384957 ACY64476.1

262385049 ACY64522.1

374346276 BAL46758.1 NCBI GI Accession Number NCBI RefSeq Accession Number

262385071 ACY64533.1

262385001 ACY64498.1

78217056 ABB36664.1

262385039 ACY64517.1

78217048 ABB36661.1

374346274 BAL46757.1

374346222 BAL46731.1

46948134 AAT07049.1

374346234 BAL46737.1

262384959 ACY64477.1

374346224 BAL46732.1

46948142 AAT07053.1

78217058 ABB36665.1

262385033 ACY64514.1

374346254 BAL46747.1

374346264 BAL46752.1

262384979 ACY64487.1

374346334 BAL46787.1

374346252 BAL46746.1

374346318 BAL46779.1

374346306 BAL46773.1

374346280 BAL46760.1

85815563 CAI54237.1

374346330 BAL46785.1

374346236 BAL46738.1

374346194 BAL46717.1

374346206 BAL46723.1

374346196 BAL46718.1

46948124 AAT07044.1

374346268 BAL46754.1

374346272 BAL46756.1

374346292 BAL46766.1

374346218 BAL46729.1

374346198 BAL46719.1

374346322 BAL46781.1

78217069 ABB36670.1

374346278 BAL46759.1 NCBI GI Accession Number NCBI RefSeq Accession Number

374346298 BAL46769.1

374346210 BAL46725.1

374346214 BAL46727.1

78217073 ABB36672.1

374346202 BAL46721.1

46948140 AAT07052.1

346987803 AE051752.1

374346288 BAL46764.1

46948138 AAT07051.1

46948128 AAT07046.1

374346204 BAL46722.1

46948136 AAT07050.1

374346244 BAL46742.1

46948126 AAT07045.1

374346332 BAL46786.1

374346308 BAL46774.1

254733624 ZP 05191341.1

89101109 ZP_01173946.1

374346226 BAL46733.1

374346230 BAL46735.1

124516141 EAY57649.1

374346192 BAL46716.1

294871860 XP_002766074.1

294941712 XP_002783202.1

206602939 EDZ39419.1

294873397 XP_002766607.1

294868340 XP_002765489.1

399026596 ZP_10728314.1

294868342 XP_002765490.1

392966205 ZP_10331624.1

46204585 ZP_00049779.2

251770823 EES51411.1

388565844 ZP_10152326.1

152983035 YP_001352597.1

54303991 CAH04648.1

149925487 ZP 01913751.1

385651069 ZP_10045622.1 NCBI GI Accession Number NCBI RefSeq Accession Number

383767747 YP_005446729.1

187928564 YP_001899051.1

88810506 ZP_01125763.1

162453252 YP_001615619.1

294868338 XP_002765488.1

85715952 ZP_01046929.1

384215662 YP_005606828.1

162453114 YP_001615481.1

222831855 EEE70332.1

37520735 NP_924112.1

294873395 XP_002766606.1

374346304 BAL46772.1

319760141 YP_004124080.1

383774186 YP_005453253.1

399020141 ZP_10722281.1

398822562 ZP_10580941.1

283778666 YP_003369421.1

290791109 ADD63334.1

386398282 ZP_10083060.1

383762862 YP_005441844.1

146340409 YP_001205457.1

75676626 YP_319047.1

339717590 3QPI

146283668 YP_001173821.1

374577875 ZP_09650971.1

172039718 YP_001799432.1

338971876 ZP_08627255.1

315283180 ZP_07871431.1

330015852 ZP_08308323.1

27382462 NP_773991.1

393163430 EJC63483.1

Chlorite dismutase homologs

[0033] In some embodiments, a chlorite dismutase polypeptide of the present disclosure is a homolog of any of the chlorite dismutase polypeptides listed in Table 1. In certain embodiments, a chlorite dismutase polypeptide of the present disclosure has an amino acid sequence that is at least 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to the amino acid sequence of a polypeptide having the RefSeq accession number YP_005026408.1, YP_285781.1, AAM92878.1, WP_014235269.1, AAT07043.1, WP 009867516.1, CAC14884.1, WP 013516316.1, ACA21503.1,

YP 004267835.1, EFH80711.1, YP 004178041.1, YP_004367213.1, YP_004058724.1, or YP_004172359.1 and has chlorite dismutase activity.

[0034] Homologs of the polypeptides described herein may also be used in the present disclosure. As used herein, "homology" refers to sequence similarity between a reference sequence and at least a fragment of a second sequence. Homologs may be identified by any method known in the art, preferably, by using the BLAST tool to compare a reference sequence to a single second sequence or fragment of a sequence or to a database of sequences. As described below, BLAST will compare sequences based upon percent identity and similarity.

[0035] The terms "identical" or percent "identity," in the context of two or more nucleic acids or polypeptide sequences, refer to two or more sequences or subsequences that are the same. Two sequences are "substantially identical" if two sequences have a specified percentage of amino acid residues or nucleotides that are the same (i.e., 29% identity, optionally 25%, 30%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 99% or 100% identity over a specified region, or, when not specified, over the entire sequence), when compared and aligned for maximum correspondence over a comparison window, or designated region as measured using one of the following sequence comparison algorithms or by manual alignment and visual inspection. Optionally, the identity exists over a region that is at least about 50 nucleotides (or 10 amino acids) in length, or more preferably over a region that is 100 to 500 or 1000 or more nucleotides (or 20, 50, 200, or more amino acids) in length.

[0036] Methods of alignment of sequences for comparison are well-known in the art. For example, the determination of percent sequence identity between any two sequences can be accomplished using a mathematical algorithm. Non-limiting examples of such mathematical algorithms are the algorithm of Myers and Miller (1988) CABIOS 4: 11 17; the local homology algorithm of Smith and Waterman (1981) J. Mol. Biol. 147: 195-7; the homology alignment algorithm of Needleman and Wunsch (1970) J. Mol. Biol. 48:443 453; the search-for- similarity-method of Pearson and Lipman (1988) Proc. Natl. Acad. Sci. 85:2444 2448; the algorithm of Karlin and Altschul (1990) Proc. Natl. Acad. Sci. USA 872264, modified as in Karlin and Altschul (1993) Proc. Natl. Acad. Sci. USA 90:5873 5877.

[0037] For sequence comparison, typically one sequence acts as a reference sequence, to which test sequences are compared. When using a sequence comparison algorithm, test and reference sequences are entered into a computer, subsequence coordinates are designated, if necessary, and sequence algorithm program parameters are designated. Default program parameters can be used, or alternative parameters can be designated. The sequence comparison algorithm then calculates the percent sequence identities for the test sequences relative to the reference sequence, based on the program parameters. When comparing two sequences for identity, it is not necessary that the sequences be contiguous, but any gap would carry with it a penalty that would reduce the overall percent identity. For blastn, the default parameters are Gap opening penalty=5 and Gap extension penalty=2. For blastp, the default parameters are Gap opening penalty=l 1 and Gap extension penalty=l.

[0038] A "comparison window," as used herein, includes reference to a segment of any one of the number of contiguous positions including, but not limited to from 20 to 600, usually about 50 to about 200, more usually about 100 to about 150 in which a sequence may be compared to a reference sequence of the same number of contiguous positions after the two sequences are optimally aligned. Methods of alignment of sequences for comparison are well known in the art. Optimal alignment of sequences for comparison can be conducted, e.g., by the local homology algorithm of Smith and Waterman (1981), by the homology alignment algorithm of Needleman and Wunsch (1970) J Mol Biol 48(3):443-453, by the search for similarity method of Pearson and Lipman (1988) Proc Natl Acad Sci USA 85(8):2444-2448, by computerized implementations of these algorithms (GAP, BESTFIT, FASTA, and TFASTA in the Wisconsin Genetics Software Package, Genetics Computer Group, 575 Science Dr., Madison, WI), or by manual alignment and visual inspection [see, e.g. , Brent et al., (2003) Current Protocols in Molecular Biology, John Wiley & Sons, Inc. (Ringbou Ed)].

[0039] To determine the sequence identity between a reference sequence and a query sequence {e.g. , in determining the sequence identity between a chlorite dismutase polypeptide of the present disclosure and another polypeptide), a dynamic programming algorithm may be used to find the optimal alignment between the sequences. Optimal alignments may be found for both global sequence alignments and local sequence alignments. Many examples of suitable dynamic programming algorithms for finding optimal alignments are known in the art. These may include, without limitation, Needleman-Wunsch (Needleman and Wunsch (1970) J. Mol. Biol. 48:443-453) and Smith- Waterman (Smith and Waterman (1981) J. Mol. Biol.

147:195-7).

[0040] Two examples of other algorithms that may be used for determining percent sequence identity and sequence similarity are the BLAST and BLAST 2.0 algorithms, which are described in Altschul et al. (1997) Nucleic Acids Res 25(17):3389-3402 and Altschul et al. (1990) J. Mol Biol 215(3)-403-410, respectively. Software for performing BLAST analyses is publicly available through the National Center for Biotechnology Information. This algorithm involves first identifying high scoring sequence pairs (HSPs) by identifying short words of length W in the query sequence, which either match or satisfy some positive-valued threshold score T when aligned with a word of the same length in a database sequence. T is referred to as the neighborhood word score threshold (Altschul et al., supra). These initial neighborhood word hits act as seeds for initiating searches to find longer HSPs containing them. The word hits are extended in both directions along each sequence for as far as the cumulative alignment score can be increased. Cumulative scores are calculated using, for nucleotide sequences, the parameters M (reward score for a pair of matching residues; always > 0) and N (penalty score for mismatching residues; always < 0). For amino acid sequences, a scoring matrix is used to calculate the cumulative score. Extension of the word hits in each direction are halted when: the cumulative alignment score falls off by the quantity X from its maximum achieved value; the cumulative score goes to zero or below, due to the accumulation of one or more negative- scoring residue alignments; or the end of either sequence is reached. The BLAST algorithm parameters W, T, and X determine the sensitivity and speed of the alignment. The BLASTN program (for nucleotide sequences) uses as defaults a wordlength (W) of 11 , an expectation (E) or 10, M=5, N=-4, and a comparison of both strands. For amino acid sequences, the BLASTP program uses as defaults a wordlength of 3, and expectation (E) of 10, and the BLOSUM62 scoring matrix [see Henikoff and Henikoff, (1992) Proc Natl Acad Sci USA 89(22): 10915- 10919] alignments (B) of 50, expectation (E) of 10, M=5, N=-4, and a comparison of both strands.

[0041] The BLAST algorithm also performs a statistical analysis of the similarity between two sequences (see, e.g., Karlin and Altschul, (1993) Proc Natl Acad Sci USA 90(12):5873- 5877). One measure of similarity provided by the BLAST algorithm is the smallest sum probability (P(N)), which provides an indication of the probability by which a match between two nucleotide or amino acid sequences would occur by chance. For example, a nucleic acid is considered similar to a reference sequence if the smallest sum probability in a comparison of the test nucleic acid to the reference nucleic acid is less than about 0.2, more preferably less than about 0.01, and most preferably less than about 0.001.

[0042] Other than percentage of sequence identity noted above, another indication that two nucleic acid sequences or polypeptides are substantially identical is that the polypeptide encoded by the first nucleic acid is immunologically cross-reactive with the antibodies raised against the polypeptide encoded by the second nucleic acid. Thus, a polypeptide is typically substantially identical to a second polypeptide, for example, where the two peptides differ only by conservative substitutions. Another indication that two nucleic acid sequences are substantially identical is that the two molecules or their complements hybridize to each other under stringent conditions. Yet another indication that two nucleic acid sequences are substantially identical is that the same primers can be used to amplify the sequence.

Microorganisms

Recombinantly engineered host microorganisms

[0043] Certain aspects of the present disclosure relate to culturing a cell that is

recombinantly engineered. As used herein, the terms "host," "host microorganism," "host cell," and "cell" are used interchangeably and refer to a cell of the present disclosure that is recombinantly engineered.

[0044] Any prokaryotic or eukaryotic cell may be used in the present disclosure so long as it remains viable after being recombinantly engineered (e.g., by being transformed with a sequence of nucleic acids). Preferably, the cell is not adversely affected by the transduction of the necessary nucleic acid sequences, the subsequent expression of the proteins or the resulting intermediates. In some embodiments, the recombinantly engineered cell is a bacterial, yeast, or fungal cell. The term "bacteria" refers to microorganisms within the domain of bacteria. Phyla within the domain or kingdom of bacteria include Acidobacteria, Actinobacteria, Aquificae, Armatimonadetes, Bacteroidetes, Caldiserica, Chlamydiae, Chlorobi, Chloroflexi,

Chrysiogenetes, Cyanobacteria, Deferribacteres, Deinococcus-thermus, Dictyoglomi,

Elusimicrobia, Fibrobacteres, Firmicutes, Fusobacteria, Gemmatimonadetes, Lentisphaerae, Nitrospira, Planet omycetes, Proteobacteria, Spirochaetes, Synergistetes, Tenericutes,

Thermodesulfobacteria, Thermotogae, and Verrucomicrobia. The term "fungal cells" refers to eukaryotic organisms within the kingdom of fungi. Phyla within the kingdom of fungi include Ascomycota, Basidiomycota, Blastocladiomycota, Chytridiomycota, Glomeromycota, Microsporidia, and Neocallimastigomycota. As used herein, the term "yeast" refers to any fungal cell within the phyla Ascomycota and Basidiomycota. Without being limited to these organisms, many types of yeast used in laboratory and commercial settings are part of the phylum Ascomycota.

[0045] In some embodiments, a cultured cell is used to generate a fermentation product. In certain embodiments, the cell is a bacterial, yeast, or fungal cell. Exemplary bacterial, yeast, or fungal cells include without limitation the following species: Saccharomyces sp.,

Saccharomyces cerevisiae, Saccharomyces monacensis, Saccharomyces bayanus,

Saccharomyces pastorianus, Saccharomyces carlsbergensis, Saccharomyces pombe,

Trichoderma reesei, Neurospora crassa, Neurospora sp., Kluyveromyces sp., Kluyveromyces marxiamus, Kluyveromyces lactis, Kluyveromyces fragilis, Pichia stipitis, Pichia pastoris, Pichia sp., Sporotrichum thermophile, Candida shehatae, Candida tropicalis, Neurospora crassa, Zymomonas mobilis, Clostridium sp., Clostridium saccharoperbutylacetonicum, Clostridium phytojermentans, Clostridium thermocellum, Clostridium beijerinckii, Clostridium acetobutylicum, Clostridium botulinum, Clostridium butyricum, Clostridium diolis,

Clostridium ljungdahlii, Clostridium aerotolerans, Clostridium cellulolyticum, Clostridium tyrobutyricum, Clostridium pasteurianum, Moorella thermoacetica, Escherichia coli,

Klebsiella oxytoca, Thermoanaerobacterium saccharolyticum, Yarrowia lipolytica, Yarrowia sp., Bacillus subtilis, and Bacillus sp.

[0046] In some embodiments, the cell is a Clostridium cell. As used herein, "Clostridium" may include any species of bacterium from the genus Clostridium. This genus encompasses a group of over 100 bacterial species that are Gram-positive and form a subset of the Firmicutes phylum of prokaryotes.

[0047] In some embodiments, the bacterial, yeast, or fungal cell does not endogenously express a chlorite dismutase. In other embodiments, the bacterial, yeast, or fungal cell may endogenously express a chlorite dismutase.

[0048] In some embodiments, the recombinantly engineered cell is an animal cell. In some embodiments, the recombinantly engineered animal cell is a mammalian or insect cell.

Examples of mammalian cells may include without limitation a human cell (e.g., a human embryonic kidney cell line such as a 293 cell, a cancer cell line such as a HeLa cell, a hybridoma, or any other human cell line), a non-human primate cell (e.g., a monkey kidney cell such as a CV1 cell or an African green monkey kidney cell such as a VERO cell), a mouse cell (e.g., a TM4 mouse Sertoli cell, hybridoma, myeloma, or other mouse cell line), a hamster cell {e.g., a Chinese hamster ovary (CHO) or baby hamster kidney cell (BHK)], a dog cell, a cat cell, a rat cell, a farm animal cell, a horse cell, a donkey cell, a cow cell, and so forth.

Examples of insect cells may include without limitation a Spodoptera frugiperda (e.g., an Sf9 cell) and a Drosophila cell (e.g., an S2 cell). In some embodiments, the recombinantly engineered cell is a plant cell (e.g., an algal cell or a transgenic plant cell). In some

embodiments, the mammalian or insect cell is a cancer cell. In some embodiments, the vertebrate, invertebrate, or plant cell is used to generate a product, such as a polypeptide (e.g., an antibody or a recombinantly produced polypeptide).

Recombinant engineering [0049] Certain aspects of the present disclosure relate to a cell recombinantly engineered to express a chlorite dismutase polypeptide.

[0050] "Recombinant nucleic acid" or "heterologous nucleic acid" or "recombinant polynucleotide" as used herein refers to a polymer of nucleic acids where at least one of the following is true: (a) the sequence of nucleic acids is foreign to (i.e., not naturally found in) a given host cell; (b) the sequence may be naturally found in a given host cell, but in an unnatural (e.g. , greater than expected) amount; or (c) the sequence of nucleic acids contains two or more subsequences that are not found in the same relationship to each other in nature. For example, regarding instance (c), a recombinant nucleic acid sequence will have two or more sequences from unrelated genes arranged to make a new functional nucleic acid.

Specifically, the present disclosure describes the introduction of an expression vector into a host cell, where the expression vector contains a nucleic acid sequence coding for a protein that is not normally found in a host cell or contains a nucleic acid coding for a protein that is normally found in a cell but is under the control of different regulatory sequences. With reference to the host cell's genome, then, the nucleic acid sequence that codes for the protein is recombinant. A polypeptide that is referred to as recombinant generally implies that it is encoded by a recombinant nucleic acid sequence in the host cell.

[0051] A "recombinant" polypeptide, protein, or enzyme of the present disclosure, is a polypeptide, protein, or enzyme that is encoded by a "recombinant nucleic acid" or

"heterologous nucleic acid" or "recombinant polynucleotide."

[0052] In some embodiments, the nucleic acids encoding the desired polypeptides in the host cell may be heterologous to the host cell or these nucleic acids may be endogenous to the host cell but are operatively linked to heterologous promoters and/or control regions which result in the higher expression of the nucleic acid(s) in the host cell. In certain embodiments, the host cell does not naturally produce the desired polypeptides, and contains heterologous nucleic acid constructs capable of expressing one or more genes necessary for producing those molecules.

[0053] "Recombinantly engineered," "recombinantly modified," "genetically engineered," or "genetically modified" refers to any recombinant DNA or RNA method used to create a host cell that expresses either a heterologous or endogenous polypeptide at elevated levels, at lowered levels, or in a mutated form. In other words, the cell has been transfected, transformed, or transduced with a recombinant polynucleotide molecule, and thereby been altered so as to cause the cell to alter expression of a desired protein. Methods and vectors for recombinantly engineering host cells are well known in the art; for example various techniques are illustrated in Current Protocols in Molecular Biology, Ausubel et al., eds. (Wiley & Sons, New York, 1988, and quarterly updates). Recombinant engineering techniques include, without limitation, expression vectors, and targeted homologous recombination and gene activation (see, for example, U.S. Pat. No. 5,272,071).

[0054] In certain embodiments, recombinant engineering may include the expression of a single gene (e.g. , a gene encoding a chlorite dismutase polypeptide), or it may include the expression of multiple genes encoding polypeptides, such as enzymes, of a biochemical pathway (e.g. , biochemical production of a commodity chemical).

[0055] In some embodiments, the host cell is recombinantly engineered to express a chlorite dismutase. In some embodiments, the host cell is further recombinantly engineered to express one or more proteins for the production of a fermentation product. The nucleic acid(s) encoding the chlorite dismutase and other recombinant polypeptide(s) may be exogenous to the host cell, or these nucleic acids may be endogenous to the host cell but operatively linked to heterologous promoters and/or control regions which result in higher or differentially regulated expression in a host cell. Any recombinant nucleic acids may be used in combination with any other nucleic acid that encodes a protein which aids in or enables the desired expression, stability, subcellular localization, or activity of a chlorite dismutase polypeptide or one or more proteins for the production of a fermentation product.

[0056] In some embodiments, the host cell does not endogenously express a chlorite dismutase. "Endogenous" as used herein with reference to a particular cell refers to a polynucleotide or a polypeptide that is naturally found in the cell, e.g. , a gene that was present in the cell when the cell was originally isolated from nature. "Exogenous" (used

interchangeably with "heterologous") as used herein with reference to a polynucleotide or a polypeptide and a particular cell refers to a polynucleotide or a polypeptide not found in that particular species of cell in nature, but instead that is encoded by one or more recombinant polynucleotides.

[0057] In some embodiments, the host cell further contains one or more proteins necessary for heme biosynthesis. In some embodiments, a host cell of the present disclosure may endogenously express one or more proteins necessary for heme biosynthesis. In some embodiments, the host cell is further recombinantly engineered to express one or more proteins necessary for heme biosynthesis. Without wishing to be bound to theory, it is thought that heterologous expression of one or more proteins necessary for heme biosynthesis may be advantageous for chlorite dismutase activity, since the active site of a chlorite dismutase contains a heme group. A protein necessary for heme biosynthesis may refer to any protein whose presence and/or activity is required for and/or promotes one or more steps of heme biosynthesis (e.g., one or more steps in a heme biosynthesis pathway) in a host cell. For example, in some embodiments, a heme biosynthesis pathway may include one or more of the chemical reactions depicted in FIG. 11 of the present disclosure. Heterologous expression of one or more proteins necessary for heme biosynthesis may refer to expression of the one or more proteins in a host cell that does not endogenously express them, or it may refer to expression of the one or more proteins at a different level or otherwise regulated in a different manner in a host cell that also has endogenous expression of the one or more proteins.

[0058] Many proteins necessary for heme biosynthesis are known in the art (for more detailed description, see, e.g., Heinemann, I.U., et al. (2008) Arch. Biochem. Biophys.

474(2):238-51). In some embodiments, the one or more proteins necessary for heme biosynthesis are selected from uroporphyrinogen III decarboxylase, coproporphyrinogen III oxidase, protoporphyrinogen IX oxidase, and ferrochelatase.

[0059] Uroporphyrinogen III decarboxylase as used herein may refer to any protein that catalyzes the conversion of uroporphyrinogen III to coproporphyrinogen III and carbon dioxide. The EC classification for uroporphyrinogen III decarboxylase is EC 4.1.1.37. In the present disclosure, any enzyme demonstrated or predicted to have the activity corresponding to EC classification 4.1.1.37 is considered to be a uroporphyrinogen III decarboxylase polypeptide. The GO term ID for uroporphyrinogen III decarboxylase is GO: 0004853. In the present disclosure, any protein demonstrated or predicted to have the cellular component, biological process, or molecular function corresponding to the GO term ID GO: 0004853 is considered to be a uroporphyrinogen III decarboxylase polypeptide. A non-limiting example of a uroporphyrinogen III decarboxylase polypeptide described in the literature is HemE (see Nishimura, K. et al. (1993) Gene 133(1):109-13 for further description). Uroporphyrinogen III decarboxylase polypeptides from a variety of host organisms may be used, including without limitation E. coli {e.g., as described by NCBI Reference Sequence NP_418425), B. subtilis {e.g., as described by NCBI Reference Sequence NP_388893), L. monocytogenes {e.g., as described by NCBI Reference Sequence CAD00290), S. epidermidis {e.g., as described by NCBI Reference Sequence AIR82849), and S. mutans {e.g., as described by NCBI Reference Sequence WP_024786000).

[0060] Coproporphyrinogen III oxidase (CPO) as used herein may refer to any protein that catalyzes the conversion of coproporphyrinogen III to protoporphyrinogen-IX and carbon dioxide. This reaction may be catalyzed by a CPO enzyme in an oxygen-dependent reaction that uses 0 2 and H(+) as substrates. Alternatively, the reaction may also be catalyzed by a CPO enzyme in an oxygen-independent reaction that uses NADPH and S-adenosyl-L- methionine (SAM) as substrates. The EC classification for coproporphyrinogen III oxidase is EC 1.3.3.3. In the present disclosure, any enzyme demonstrated or predicted to have the activity corresponding to EC classification 1.3.3.3 is considered to be a coproporphyrinogen III oxidase polypeptide. The GO term ID for coproporphyrinogen III oxidase is GO: 0004109. In the present disclosure, any protein demonstrated or predicted to have the cellular component, biological process, or molecular function corresponding to the GO term ID GO: 0004109 is considered to be a coproporphyrinogen III oxidase polypeptide. A non-limiting example of an oxygen-independent coproporphyrinogen III oxidase polypeptide described in the literature is HemN, and a non-limiting example of an oxygen-dependent coproporphyrinogen III oxidase polypeptide described in the literature is HemF (see Layer, G. et al. (1993) Biol. Chem.

386(10):971-80 for further description). Coproporphyrinogen III oxidase polypeptides from a variety of host organisms may be used, including without limitation E. coli {e.g., as described by NCBI Reference Sequence NP_416931), B. subtilis {e.g., as described by NCBI Reference Sequence YP_007534533), L. monocytogenes {e.g., as described by NCBI Reference Sequence CAC99554), S. epidermidis (e.g., as described by NCBI Reference Sequence NP_764825), and S. nutans (e.g., as described by NCBI Reference Sequence NP_721776).

[0061] Protoporphyrinogen IX oxidase as used herein may refer to any protein that catalyzes the conversion of protoporphyrinogen-IX to protoporphyrin-IX. This reaction may be catazlyed by a protoporphyrinogen IX oxidase in an oxygen-dependent or oxygen- independent mechanism. The EC classification for protoporphyrinogen IX oxidase is EC 1.3.3.4. In the present disclosure, any enzyme demonstrated or predicted to have the activity corresponding to EC classification 1.3.3.4 is considered to be a protoporphyrinogen IX oxidase polypeptide. The GO term ID for protoporphyrinogen IX oxidase is GO: 0070818. In the present disclosure, any protein demonstrated or predicted to have the cellular component, biological process, or molecular function corresponding to the GO term ID GO: GO: 0070818 is considered to be a protoporphyrinogen IX oxidase polypeptide. A non-limiting example of an oxygen-independent protoporphyrinogen IX oxidase polypeptide described in the literature is HemG (see Boynton, T.O. et al. (2009) Biochemistry 48(29):6705-l 1 for further description), and a non-limiting example of an oxygen-dependent protoporphyrinogen IX oxidase polypeptide is HemY (see Boynton, T.O. et al. (2011) Appl. Environ. Microbiol. 77(14):4795-801 for further description). Protoporphyrinogen IX oxidase polypeptides from a variety of host organisms may be used, including without limitation E. coli (e.g., as described by NCBI Reference Sequence NP_418292), K. pneumoniae (e.g., as described by NCBI Reference Sequence CED77855), and V. parahaemolyticus (e.g., as described by NCBI Reference Sequence NP_796412).

[0062] Ferrochelatase as used herein may refer to any protein that catalyzes the interconversion of protoporphyrin and protoheme. The EC classification for ferrochelatase is EC 4.99.1.1. In the present disclosure, any enzyme demonstrated or predicted to have the activity corresponding to EC classification 4.99.1.1 is considered to be a ferrochelatase polypeptide. The GO term ID for ferrochelatase is GO: 0004325. In the present disclosure, any protein demonstrated or predicted to have the cellular component, biological process, or molecular function corresponding to the GO term ID GO: GO: 0004325 is considered to be a ferrochelatase polypeptide. A non-limiting example of a ferrochelatase polypeptide described in the literature is HemH (see Frustaci, J.M. and O'Brian, M.R. (1993) Appl. Environ. Microbiol. 59(8):2347-51 for further description). Ferrochelatase polypeptides from a variety of host organisms may be used, including without limitation E. coli {e.g., as described by NCBI Reference Sequence NP_415008), B. subtilis {e.g., as described by NCBI Reference Sequence NP_388894), L. monocytogenes {e.g., as described by NCBI Reference Sequence NP_465735), S. epidermidis {e.g., as described by NCBI Reference Sequence AIR83582), S. mutans {e.g., as described by NCBI Reference Sequence WP_024781571), and K. pneumoniae {e.g., as described by NCBI Reference Sequence YP_005225503).

[0063] In some embodiments, chlorite dismutase is expressed under the control of a promoter. "Under the control" refers to a recombinant nucleic acid that is operably linked to a control sequence, or promoter. The term "operably linked" as used herein refers to a configuration in which a control sequence or promoter is placed at an appropriate position relative to the coding sequence of the nucleic acid sequence such that the control sequence or promoter directs the expression of a polypeptide.

[0064] "Promoter" is used herein to refer to any nucleic acid sequence that regulates the initiation of transcription for a particular polypeptide-encoding nucleic acid under its control. A promoter does not typically include nucleic acids that are transcribed, but it rather serves to coordinate the assembly of components that initiate the transcription of other nucleic acid sequences under its control. A promoter may further serve to limit this assembly and subsequent transcription to specific prerequisite conditions. Prerequisite conditions may include expression in response to one or more environmental, temporal, or developmental cues; these cues may be from outside stimuli or internal functions of the cell. Bacterial and fungal cells possess a multitude of proteins that sense external or internal conditions and initiate signaling cascades ending in the binding of proteins to specific promoters and subsequent initiation of transcription of nucleic acid(s) under the control of the promoters. When transcription of a nucleic acid(s) is actively occurring downstream of a promoter, the promoter can be said to "drive" expression of the nucleic acid(s). A promoter minimally includes the genetic elements necessary for the initiation of transcription, and may further include one or more genetic elements that serve to specify the prerequisite conditions for transcriptional initiation. A promoter may be encoded by the endogenous genome of a host cell, or it may be introduced as part of a recombinantly engineered polynucleotide. A promoter sequence may be taken from one host species and used to drive expression of a gene in a host cell of a different species. A promoter sequence may also be artificially designed for a particular mode of expression in a particular species, through random mutation or rational design. In recombinant engineering applications, specific promoters are used to express a recombinant gene under a desired set of physiological or temporal conditions or to modulate the amount of expression of a recombinant nucleic acid.

[0065] In some embodiments, chlorite dismutase is expressed under the control of a constitutive promoter. A constitutive promoter is defined herein as a promoter that drives the expression of nucleic acid(s) continuously and without interruption in response to internal or external cues. Constitutive promoters are commonly used in recombinant engineering to ensure continuous expression of desired recombinant nucleic acid(s). Constitutive promoters often result in a robust amount of nucleic acid expression, and, as such, are used in many recombinant engineering applications to achieve a high level of recombinant protein and enzymatic activity. These promoters are therefore suited for applications wherein a product derived from an enzymatic activity is purified, for example the production of a hydrocarbon or hydrocarbon derivative by one or more recombinant proteins.

[0066] Many constitutive promoters are known and characterized in the art. Exemplary bacterial constitutive promoters include without limitation the E. coli promoters P spc , P i a , PRNAi, P AII J PI an d P2 from rrnB, and the lambda phage promoter PL (Liang, S.T. et al. (1999). J Mol Biol 292(1): 19-37). Exemplary yeast constitutive promoters include without limitation the ADH1, GPD, TEF1, TEF2, and HXT7 promoters from S. cerevisiae (see, e.g., Sun, J. et al. (2012). Biotechnol Bioeng 109(8): 2082-92). Constitutive promoters functional in a wide variety of host bacterial and yeast cells are well known in the art.

[0067] In some embodiments, chlorite dismutase is expressed under the control of an inducible promoter. An inducible promoter is defined herein as a promoter that drives the expression of nucleic acid(s) selectively and reliably in response to a specific stimulus. An ideal inducible promoter will drive no nucleic acid expression in the absence of its specific stimulus but drive robust nucleic acid expression rapidly upon exposure to its specific stimulus. Additionally, some inducible promoters induce a graded level of expression that is tightly correlated with the amount of stimulus received. Stimuli for known inducible promoters include, for example, heat shock, exogenous compounds (e.g., a sugar, metal, drug, or phosphate), salts or osmotic shock, oxygen, and biological stimuli (e.g., a growth factor or pheromone).

[0068] Inducible promoters are often used in recombinant engineering applications to limit the expression of recombinant nucleic acid(s) to desired circumstances. For example, since high levels of recombinant protein expression may sometimes slow the growth of a host cell, the host cell may be grown in the absence of recombinant nucleic acid expression, and then the promoter may be induced when the host cells have reached a desired density. Many inducible promoters are known and characterized in the art. Exemplary bacterial inducible promoters include without limitation the E. coli promoters P lac , P^, P taC , Ρχ 7 , PBAD, and Pi ac uv 5 (Nocadello, S. and Swennen, E.F. (2012). Microb Cell Fact 11:3). Exemplary yeast inducible promoters include without limitation the GAL1, GAL10, PH05, PGK, and MFal promoters from S.

cerevisiae (Romanos, M.A. et al. (1992). Yeast 8:423-88). Inducible promoters functional in a wide variety of host bacterial and yeast cells are well known in the art.

[0069] In some embodiments, chlorite dismutase is expressed under the control of a yeast promoter. In addition to those listed above, examples of suitable yeast promoters include without limitation pTDH3, pTEFl, pRPL18, pRNR2, and pREVl. In some embodiments, chlorite dismutase is expressed by operably linking a chlorite dismutase coding sequence with a terminator sequence (e.g., the S. cerevisiae ADH1 terminator). In some embodiments, chlorite dismutase is expressed in yeast from a centromere-based plasmid. A centromere-based plasmid may include any plasmid bearing a functional centromere sequence (e.g., the S.

cerevisiae CEN6 sequence), which helps maintain the plasmid during cell division of the host cell. A yeast promoter of the present disclosure may refer to any promoter sequence that regulates the expression of a gene in any yeast of the present disclosure, including without limitation S. cerevisiae, Saccharomyces sp., Pichia pastoris, Pichia sp., Yarrowia lipolytica, Yarrowia sp., and any of the other yeasts described herein.

[0070] A recombinant polypeptide-encoding nucleic acid or plurality of polypeptide- encoding nucleic acids may be encoded by an expression vector. An expression vector contains one or more polypeptide-encoding nucleic acids, and it may further contain any desired elements that control the expression of the nucleic acid(s), as well as any elements that enable the replication and maintenance of the expression vector inside a given host cell. In some embodiments, one or more recombinant nucleic acids may be contained by the microorganism (e.g. , a chlorite dismutase polypeptide and one or more proteins for the production of a fermentation product). All of the recombinant nucleic acids may be present on a single expression vector, or they may be encoded by multiple expression vectors. As such, they may be controlled together by the same genetic elements or controlled separately by distinct genetic elements. Furthermore, the recombinant nucleic acid(s) may be maintained separately from the host genome, or the recombinant nucleic acid(s) may be integrated into the host genome. In addition, recombinant nucleic acid(s) may be introduced without an expression vector. For example, nucleic acids may be directly introduced into a bacterial or fungal host cell's genome by homologous recombination or recombineering. When more than one recombinant nucleic acid is present, they may all be maintained separately from the host genome, or they may all be integrated into the host genome, or they may exist in any combination of separate and integrated.

[0071] Methods to produce recombinant microorganisms by introducing recombinant nucleic acids/proteins, and vectors suitable for this purpose, are well known in the art. For example, various techniques are illustrated in Current Protocols in Molecular Biology, Ausubel et al., eds. (Wiley & Sons, New York, 1988, and quarterly updates). Methods for transferring expression vectors into host cells are well known in the art. Specific methods and vectors may differ depending upon the species of the desired host cell. For example, bacterial host cells may be transformed by heat shock, calcium chloride treatment, electroporation, liposomes, or phage infection. Yeast host cells may be transformed by lithium acetate treatment (may further include carrier DNA and PEG treatment) or electroporation. These methods are included for illustrative purposes and are in no way intended to be limiting or comprehensive. Routine experimentation through means well known in the art may be used to determine whether a particular expression vector or transformation method is suited for a given host cell.

Furthermore, reagents and vectors suitable for many different host microorganisms are commercially available and/or well known in the art. Cell Culture

[0072] Certain aspects of the present disclosure relate to methods of culturing a cell. As defined herein, "culturing" a cell refers to introducing an appropriate culture medium, under appropriate conditions, to promote the growth of a cell. Methods of culturing various types of cells are known in the art. Culturing may be performed using a liquid or solid growth medium. Culturing may be performed under aerobic or anaerobic conditions where aerobic, anoxic, or anaerobic conditions are preferred based on the requirements of the microorganism and desired metabolic state of the microorganism. In addition to oxygen levels, other important conditions may include, without limitation, temperature, pressure, light, pH, and cell density.

[0073] In some embodiments, a culture medium is provided. A "culture medium" or "growth medium" as used herein refers to a mixture of components that supports the growth of cells. In some embodiments, the culture medium may exist in a liquid or solid phase. A culture medium of the present disclosure can contain any nutrients required for growth of microorganisms. In certain embodiments, the culture medium may further include any compound used to reduce the growth rate of, kill, or otherwise inhibit additional contaminating microorganisms. In some embodiments, the compound is chlorite. In certain embodiments, the growth medium is treated with chlorite in an amount sufficient to reduce the growth rate, kill, or otherwise inhibit additional contaminating microorganisms. The growth medium may also contain any compound used to modulate the expression of a nucleic acid, such as one operably linked to an inducible promoter (for example, when using a yeast cell, galactose may be added into the growth medium to activate expression of a recombinant nucleic acid operably linked to a GAL1 or GAL10 promoter). In further embodiments, the culture medium may lack specific nutrients or components to limit the growth of contaminants, select for microorganisms with a particular auxotrophic marker, or induce or repress expression of a nucleic acid responsive to levels of a particular component.

[0074] In some embodiments, the methods of the present disclosure may include culturing a host cell under conditions sufficient for the production of a fermentation product. In certain embodiments, culturing a host cell under conditions sufficient for the production of a fermentation product entails culturing the cells in a suitable culture medium. Suitable culture media may differ among different microorganisms depending upon the biology of each microorganism. Selection of a culture medium, as well as selection of other parameters required for growth (e.g. , temperature, oxygen levels, pressure, etc.), suitable for a given microorganism based on the biology of the microorganism are well known in the art.

Examples of suitable culture media may include, without limitation, common commercially prepared media, such as Luria Bertani (LB) broth, Sabouraud Dextrose (SD) broth, or Yeast medium (YM) broth. In other embodiments, alternative defined or synthetic culture media may also be used.

[0075] In some embodiments, a host cell of the present disclosure that is recombinantly engineered to express a chlorite dismutase polypeptide is cultured under conditions where a chlorite dismutase polypeptide is expressed in the cell. As disclosed herein, conditions suitable for chlorite dismutase expression may refer, without limitation, to the nutrient composition, pH, temperature, oxygen level, and/or light exposure of the culture medium containing the host cell. In some embodiments, the conditions may allow for the chlorite dismutase polypeptide to be properly translated, processed, localized within the cell, and sufficiently stable and enzymatically active.

[0076] In some embodiments, a host cell of the present disclosure is cultured under micro- aerobic conditions. As used herein, "micro-aerobic conditions" may refer to any conditions for cell culture wherein the amount of oxygen is less than about 20% (the approximate amount of oxygen in the atmosphere). In some embodiments, the host cell is cultured in less than about 20% oxygen, less than about 19% oxygen, less than about 18% oxygen, less than about 17% oxygen, less than about 16% oxygen, less than about 15% oxygen, less than about 14% oxygen, less than about 13% oxygen, less than about 12% oxygen, less than about 11% oxygen, less than about 10% oxygen, less than about 9% oxygen, less than about 8% oxygen, less than about 7% oxygen, less than about 6% oxygen, less than about 5% oxygen, less than about 4% oxygen, less than about 3% oxygen, less than about 2% oxygen, or less than about 1 % oxygen. Decreasing the amount of available oxygen is known to result in cellular stress. Without wishing to be bound to theory, it is thought that the presence of oxygen may aid cells cultured under such stress in part by balancing co-factors to facilitate host cell growth, metabolism, polypeptide production, and/or polypeptide activity. As disclosed herein, chlorite dismutase polypeptides catalyze the reaction converting chlorite (CIO 2 ) into chloride (CI ) and oxygen (0 2 ). As such, and without wishing to be bound to theory, it is further thought that a chlorite dismutase polypeptide of the present disclosure may influence the growth or output of the host cell by enzymatically generating intracellular oxygen, particularly under micro-aerobic conditions and/or conditions under which oxygen levels influence the balance of co-factors. Without wishing to be bound to theory, expression of a chlorite dismutase polypeptide of the present disclosure may therefore be particularly advantageous for a host cell that experiences stress under micro-aerobic conditions, for example a yeast cell of the present disclosure.

[0077] Methods to identify conditions where a chlorite dismutase polypeptide is expressed in a host cell are well known in the art. These methods may include, for example, growing a cell expressing a chlorite dismutase under a specific set of conditions in the presence of chlorite, and comparing its growth under identical conditions and chlorite levels to a corresponding cell of the same species that lacks chlorite dismutase. In other embodiments, additional suitable conditions may be identified by directly assessing the abundance of chlorite dismutase RNA or protein through standard methods known in molecular biology (Current Protocols in Molecular Biology, Ausubel et al., eds. Wiley & Sons, New York, 1988, and quarterly updates).

[0078] In some embodiments, the chlorite dismutase is expressed under the control of a constitutive promoter. Accordingly, in certain embodiments, a host cell is cultured under conditions whereby the promoter is sufficiently active to produce the desired amount or activity of chlorite dismutase polypeptide. In some embodiments, this desired amount may be empirically determined, for example, by measuring the growth of a host cell expressing the chlorite dismutase as a function of chlorite concentration in the growth medium.

[0079] In some embodiments, the chlorite dismutase is expressed under the control of an inducible promoter. Thus, in certain embodiments, the host cell is cultured under appropriate conditions to allow activation of the inducible promoter and subsequent production of chlorite dismutase polypeptide. For example, if the inducible promoter is activated by the addition of a metal, the host cell may be cultured in a culture medium containing the requisite amount of a suitable metal-containing compound that will allow activation of the promoter and sufficient expression of an operably linked chlorite dismutase polypeptide.

Fermentation

[0080] Certain aspects of the present disclosure relate to a host cell that produces a fermentation product. "Fermentation" refers to the metabolic process by which

microorganisms convert a reduced carbon source into products including, without limitation, acids, alcohols, or gases without utilizing oxidative phosphorylation. Fermentation can either be an endogenous process in a host microorganism, or it may be an exogenous process that has been recombinantly engineered into a host microorganism. The enzymes that generate a fermentation product may be endogenous to the cell, or they may be exogenous proteins encoded by recombinantly expressed polynucleotides. One example of endogenous fermentation is the breakdown of glucose to yield ethanol and carbon dioxide. In some embodiments, a microorganism of the present disclosure contains one or more recombinant proteins necessary for the production of a fermentation product. In some embodiments, the necessary proteins may include, without limitation, one or more recombinant enzymes catalyzing any step in a chemical pathway that yields a desired product. In other embodiments, the one or more recombinant proteins may further include any protein that enhances the expression, stability, or activity of an enzyme, even if the protein itself does not possess catalytic activity.

[0081] In some embodiments, a host cell is cultured under conditions sufficient for the cell to produce a fermentation product. Conditions sufficient for a cell to produce a fermentation product are well known in the art and may be adapted depending upon the species of the cell, the desired product(s), or the scale of production. Fermentation may be executed on any scale of production, from a flask in a laboratory to large-scale industrial or commercial processing. Methods for fermentation on many scales are well known in the art (for example, see

Villadsen, J. et al. Bioreaction Engineering Principles. 3 rd ed. Springer; 2011). In some embodiments, fermentation methods have been adapted for diverse applications including, without limitation, biofuel production, beer production, winemaking, protein purification, and synthesis of pharmaceutical products. [0082] In some embodiments, the cell uses a carbon source as a substrate for fermentative metabolism. As used herein, a "carbon source" refers to a carbon-containing substrate or compound suitable for use by the microorganism for cell growth. Suitable carbon sources may include without limitation polymers, carbohydrates, acids, alcohols, aldehydes, ketones, amino acids, and peptides. Further examples of suitable carbon sources may include, without limitation, various monosaccharides such as glucose, arabinose, or xylose; oligosaccharides; disaccharides; polysaccharides; biomass polymers such as cellulose or hemicellulose;

cellodextrins, such as cellobiose, cellotriose, etc.; saturated or unsaturated fatty acids;

succinate; lactate; acetate; and ethanol, or mixtures of any of the preceding compounds. The carbon source can additionally be a product of photosynthesis, including, but not limited to glucose.

[0083] In some embodiments, a culture medium of the present disclosure may further include suitable minerals, salts, cofactors, buffers and other components known in the art that are suitable for the growth of the cells in culture and for the promotion of the enzymatic pathways that produce fermentation products. In other embodiments, the culture medium may be further supplemented with components that facilitate fermentation, including, without limitation, trace elements, growth factors, gases, chemical precursors, inducers, chelators, inhibitors, antibiotics and/or antimycotics, buffers, and antifoaming agents. In some embodiments, recombinantly engineered cells of the present disclosure are cultured under anaerobic conditions that promote fermentative metabolism.

[0084] A recombinantly engineered cell of the present disclosure may be grown by any culturing method known in the art, including, without limitation, batch culturing, fed-batch culturing, or continuous culturing. In some embodiments, a recombinantly engineered cell is grown in a bioreactor. The term "bioreactor" as used herein may refer to any vessel and its supporting components that contain the cell and its culture medium during fermentation. A bioreactor of the present disclosure may be of any size, shape, or design suitable for the particular application. In some embodiments, a suitable bioreactor includes, without limitation, a stirred-tank bioreactor. In some embodiments, a bioreactor of the present disclosure may contain one or more vessels, sensors for monitoring culture and microorganismal parameters, and control devices to alter the conditions and composition of the culture medium. Typically, bioreactors are closed to the outside environment, except for controlled inputs and outputs, to minimize introduction of additional contaminating microorganisms. In some embodiments, a recombinantly engineered cell of the present disclosure may be suspended in liquid culture medium within a bioreactor. In other embodiments, a recombinantly engineered cell may be fixed within the bioreactor. Suitable bioreactors for culturing fixed cells may include, without limitation, features such as plates, packed beds, moving beds, or membranes.

[0085] In some embodiments, the culture medium containing the cell is treated with chlorite prior to growing the cell in a bioreactor. In some embodiments, the chlorite treatment is carried out "off-line," or separate from fermentation. Examples of off-line treatment may include an organism recycle scheme, wherein a recombinantly engineered cell is removed from a bioreactor, treated with a chlorite solution (in a culture medium of the present disclosure, and while the cell is expressing a chlorite dismutase polypeptide of the present disclosure), and then re-introduced to a bioreactor for fermentation.

[0086] In some embodiments, the culture medium containing the cell is treated with chlorite while growing the cell in a bioreactor. In some embodiments, chlorite is added to the culture medium while the cell is fermenting in a bioreactor.

[0087] During fermentation, bioreactor conditions may be adjusted to affect the growth rate of the cultured host cell, for example by using continuous culture methods. Devices that continuously monitor and adjust particular parameters of the conditions in the bioreactor are well known in the art and include, without limitation, chemostats, auxostats, and turbidostats. Methods and devices for monitoring and controlling parameters such as temperature, pH, flow rates, dissolved gas levels, nutrient and waste levels, pressure, turbidity, agitation, and circulation are well known in the art (see, e.g., Villadsen, J. et al. Bioreaction Engineering

rd

Principles. 3 ed. Springer; 2011).

Fermentation products

[0088] Certain aspects of the present disclosure relate to cells recombinantly engineered to express a chlorite dismutase polypeptide that are cultured under conditions sufficient for the cell to produce a fermentation product. As used herein, "fermentation products" may include compounds endogenously produced by fermentative metabolism of the host cell, or they may include compounds produced by the activity of recombinantly encoded polypeptides.

Examples of suitable fermentation products include, without limitation, pharmaceutical compounds, commodity chemicals, plastics, biofuels, alcohols, oils, biodiesel, carbohydrates, amino acids, fatty acids and their esters, and precursors thereof. In some embodiments, the fermentation product of a recombinantly engineered cell may include hydrocarbons, hydrocarbon derivatives, ethanol, and butanol. "Hydrocarbon" as used herein refers to any compound solely composed of the elements carbon and hydrogen.

[0089] In some embodiments, cells of the present disclosure endogenously produce a fermentation product. Examples of suitable endogenous fermentation products include, without limitation, ethanol, lactic acid, and carbon dioxide. In some embodiments, recombinant engineering may enhance a cell' s production of an endogenous fermentation product.

[0090] In other embodiments, a recombinantly engineered cell of the present disclosure further contains the proteins necessary to produce a fermentation product. In certain embodiments, the recombinantly engineered cell is further engineered to express one or more proteins necessary for the production of a fermentation product. In some embodiments, the fermentation product may result from a biochemical pathway that is engineered into the cell by recombinant polypeptide(s). In other embodiments, the fermentation product may result from a biochemical pathway in the cell that includes both endogenous and recombinant polypeptides. Recombinant proteins may participate in any step of fermentative metabolism. For example, recombinant proteins may allow a cell to utilize a carbon source that is not endogenously used by the cell for fermentation, such as xylose or cellobiose. Recombinant proteins may also allow a cell to transform a carbon source into a fermentation product that is not endogenously produced by the cell, such as butanol.

[0091] In some embodiments, a cell is further recombinantly engineered to produce a hydrocarbon or hydrocarbon derivative. In certain embodiments, suitable hydrocarbon derivatives may include ethanol or butanol. Examples of recombinantly engineering a host cell to produce a fermentation product, including hydrocarbons, hydrocarbon derivatives, ethanol, and butanol, are well known in the art (Tang, W.L. and Zhao, H. (2009). Biotechnol J

4(12): 1725-39). In one non-limiting example, a set of recombinant nucleic acid encoding an n- butanol production pathway, including the Clostridium acetobutylicum genes adhE2, bed, crt, etfAB, hbd, and the E. coli atoB gene, can be introduced into recombinant cells to allow the cells to produce rc-butanol from rich culture medium (Atsumi, S. et al. (2008). Metab Eng 10:305-11). In some embodiments, oils, such as fatty acid esters, can be produced by recombinantly engineered cells. For example, cells can be recombinantly engineered to express pyruvate decarboxylase and alcohol dehydrogenase and to lack acyl-CoA synthetase genes. In some embodiments, oil fermentation products can be further reacted with alcohols to produce biodiesel (see, e.g., Tang, W.L. and Zhao, H. (2009). Biotechnol J 4(12): 1725-39).

[0092] In certain embodiments, a recombinantly engineered cell of the present disclosure may contain a deletion of one or more endogenous genes whose encoded protein's activities compete or otherwise interfere with the production of a desired fermentation product. In one non-limiting example, it has been shown that the removal of msgA, cydAB, cyoABCD, and cbdAB genes in E. coli, along with expression of a recombinant IdhL from Pediococcus acidilactici, allows the production of D- and L-lactate under specific conditions (Tang, W.L. and Zhao, H. (2009). Biotechnol J 4(12): 1725-39).

[0093] Fermentation products may be purified by any suitable method known in the art. In some embodiments, fermentation products may be purified continuously, e.g., from a continuous culture. In other embodiments, fermentation products may be purified separately from fermentation, e.g., from a batch or fed-batch culture.

Chlorite Treatment

Chlorite-containing solution

[0094] Certain aspects of the present disclosure relate to the use of chlorite in the culture medium to reduce the growth rate or kill one or more contaminating microorganisms. As used herein, "chlorite" or "chlorite ions" refers to any solution or compound that contains a chlorite anion (CIO 2 ). "Chlorite" of the present disclosure may include any salt or acid containing the chlorite anion. Additionally, "chlorite" of the present disclosure may also include any solution containing any compound that can break down into or otherwise generate chlorite. Examples of compounds that may generate chlorite include, without limitation, any compound that contains a chlorate (any compound containing the CIO 3 " anion), perchlorate (any compound containing the CIO 4 " anion), or chlorine dioxide (the non-ionic CIO 2 ). It is well known in the art that such compounds may be chemically interconverted. For example, upon contact with water, sanitizing solutions with chlorine dioxide can undergo several oxidation-reduction reactions that generate chlorite, (per)chlorate, and chloride anions (EPA Guidance Manual, Chapter 4: Chlorine dioxide). The term "(per)chlorate" as used herein refers to any solutions or compounds that contain a chlorate or perchlorate anion.

[0095] Solutions and methods for using chlorite treatment as a disinfectant are well known in the art (see for example Bichai, F. and Barbeau, B. (2006). Water Qual Res J Canada 41(4):375-82). One non-limiting example of a chlorite solution of the present disclosure is sodium chlorite, which is sold in various formulations including SANOVA® Base, a 25% solution of sodium chlorite salt, for disinfecting food products (Technical Evaluation Report for the USDA National Organic Program: Acidified Sodium Chlorite. July 21, 2008). In some embodiments, disinfecting solutions containing chlorite may further include other active or inert compound that aids or complements the disinfecting properties of the chlorite. One non- limiting example of this type of solution is acidified sodium chlorite, a disinfecting solution that contains chlorite and an acid, such as citric acid.

[0096] Another means known in the art for generating a chlorite solution of the present disclosure is by electrochemical generation (also known as a membrane electrolysis method). In this method, sodium chlorite is introduced into an electrochemical cell in an aqueous solution. Electrochemical energy applied to the cell catalyzes the formation of chlorine dioxide, which can be separated from the sodium chlorite and aqueous solution, for example by using a membrane. In some embodiments, chlorine dioxide may be generated, e.g., in the influent stream, by using the electrochemical method.

[0097] In some embodiments, a culture medium of the present disclosure is treated with chlorite of the present disclosure in an amount sufficient to reduce the growth rate or kill one or more contaminating microorganisms. In some embodiments, the amount used for treatment is dependent upon the parameters of the culture medium. For example, the volume, surrounding pH, and temperature of the culture medium may affect the amount sufficient to reduce the growth rate or kill one or more contaminating microorganisms. In some embodiments, the amount sufficient to reduce the growth rate or kill one or more contaminating microorganisms may further depend upon the pre-treatment growth rate, metabolic capabilities, pre-treatment amount, and specific species of the contaminating microorganisms. Methods for identifying the amount sufficient to reduce the growth rate or kill one or more contaminating

microorganisms are well known in the art. For example, well-known assays may be used, such as a kill curve plotting the cell density (a measure of growth) of a particular microorganism as a function of chlorite concentration in the culture medium for a particular time interval of interest.

[0098] In some embodiments, the amount of chlorite sufficient to reduce the growth rate, kill, or otherwise inhibit one or more contaminating microorganisms may include, without limitation, at least 1 μΜ, at least 5 μΜ, at least 10 μΜ, at least 20 μΜ, at least 30 μΜ, at least

40 μΜ, at least 50 μΜ, at least 60 μΜ, at least 70 μΜ, at least 80 μΜ, at least 90 μΜ, at least

100 μΜ, at least 110 μΜ, at least 120 μΜ, at least 130 μΜ, at least 140 μΜ, at least 150 μΜ, at least 160 μΜ, at least 170 μΜ, at least 180 μΜ, at least 190 μΜ, at least 200 μΜ, at least

210 μΜ, at least 220 μΜ, at least 230 μΜ, at least 240 μΜ, at least 250 μΜ, at least 260 μΜ, at least 270 μΜ, at least 280 μΜ, at least 290 μΜ, at least 300 μΜ, at least 310 μΜ, at least

320 μΜ, at least 330 μΜ, at least 340 μΜ, at least 350 μΜ, at least 360 μΜ, at least 370 μΜ, at least 380 μΜ, at least 390 μΜ, at least 400 μΜ, at least 410 μΜ, at least 420 μΜ, at least

430 μΜ, at least 440 μΜ, at least 450 μΜ, at least 460 μΜ, at least 470 μΜ, at least 480 μΜ, at least 490 μΜ, at least 500 μΜ, at least 510 μΜ, at least 520 μΜ, at least 530 μΜ, at least

540 μΜ, at least 550 μΜ, at least 560 μΜ, at least 570 μΜ, at least 580 μΜ, at least 590 μΜ, at least 600 μΜ, at least 610 μΜ, at least 620 μΜ, at least 630 μΜ, at least 640 μΜ, at least

650 μΜ, at least 660 μΜ, at least 670 μΜ, at least 680 μΜ, at least 690 μΜ, at least 700 μΜ, at least 710 μΜ, at least 720 μΜ, at least 730 μΜ, at least 740 μΜ, at least 750 μΜ, at least

760 μΜ, at least 770 μΜ, at least 780 μΜ, at least 790 μΜ, at least 800 μΜ, at least 810 μΜ, at least 820 μΜ, at least 830 μΜ, at least 840 μΜ, at least 850 μΜ, at least 860 μΜ, at least

870 μΜ, at least 880 μΜ, at least 890 μΜ, at least 900 μΜ, at least 910 μΜ, at least 920 μΜ, at least 930 μΜ, at least 940 μΜ, at least 950 μΜ, at least 960 μΜ, at least 970 μΜ, at least 980 μΜ, at least 990 μΜ, at least 1 niM, at least 1.5 niM, at least 2 niM, at least 2.5 niM, at least 3 niM, at least 3.5 niM, at least 4 niM, at least 4.5 niM, and at least 5 niM chlorite ion concentration. In some embodiments, the amount of chlorite sufficient to reduce the growth rate, kill, or otherwise inhibit one or more contaminating microorganisms is between about 10 μΜ and about 200 μΜ.

[0099] A solution containing chlorite of the present disclosure may be introduced to the culture medium at any time or stage of the culturing process, in the presence or absence of the recombinantly engineered cell. In certain embodiments, a culture medium is treated with chlorite prior to culturing a recombinantly engineered cell of the present disclosure. In some embodiments, the chlorite is added to the culture medium concurrently with the addition of a recombinantly engineered cell. In some embodiments, the chlorite is added to the culture medium intermittently during the culturing process. In some embodiments, the chlorite is produced continuously during culturing of a recombinantly engineered cell of the present disclosure. In some embodiments, the chlorite is continuously produced by electrochemical generation in the culture medium.

[0100] In some embodiments, a recombinantly engineered cell of the present disclosure is cultured in culture medium treated with chlorite under conditions whereby the chlorite dismutase polypeptide of the present disclosure is expressed. In certain preferred embodiments, the chlorite dismutase polypeptide is expressed and active when the cell is exposed to chlorite, thereby protecting the cell from the toxic effects of chlorite treatment.

Contaminating microorganisms

[0101] As used herein, "contaminating microorganisms" refers to any microorganism present in the growth or fermentation medium other than the host cell that is cultured intentionally in the medium. As it contains all of the nutrients required to support the growth of desired

microorganisms, culture medium may unintentionally serve to promote the growth of one or more contaminating microorganisms. Although hygienic process methods are designed to limit the exposure of the culture medium to contaminating microorganisms, in practice it is very difficult to ensure that one or more additional contaminating microorganisms will never be introduced into the culture medium, especially when cell culturing is performed on an industrial scale. Contaminating microorganisms are unwanted because they interfere with the growth and metabolism of the desired microorganisms through various means, such as by killing them, impairing their metabolism, competing with them for nutrients, or by secreting products that alter the metabolism of the fermentation microorganisms. They may further be unwanted because they chemically affect the growth medium or the desired fermentation product.

[0102] Examples of such "contaminating microorganisms" include, without limitation, prokaryotic microorganisms, bacteria, archaea, eukaryotic microorganisms, fungal cells, yeast, algae, and viruses, including bacteriophages. As used herein, the term "prokaryotic

microorganisms" may refer to any microorganism that lacks a membrane-bound nucleus.

Examples of prokaryotic microorganisms may include, without limitation, bacteria and archaea. As used herein, the term "bacteria" may refer to any microorganism within the domain of bacteria. As used herein, the term "archaea" may refer to any microorganism within the domain of archaea. As used herein, the term "eukaryotic microorganisms" may refer to any

microorganism within the domain of eukaryota, including, without limitation, fungal cells, yeast, and algae. The terms "fungal cells" and "yeast" are described in paragraphs [0045]-[0046] of the present disclosure. As used herein, the term "algae" may refer to any eukaryotic microorganism that contains chlorophyll, including for example phototrophic protists. As used herein, the term "bacteriophage" may refer to any virus that infects bacteria.

[0103] In some embodiments, the one or more contaminating microorganisms are from a bacterial species. In some embodiments, the contaminating bacteria may be members of a genus such as Lactobacillus, Clostridium, Pediococcus, Enterococcus, Acetobacter, and

Gluconobacter. Contaminating microorganisms may also include bacteria or fungi, and they may represent any single species or any mixture of species from either the kingdom of bacteria or fungi.

[0104] In some embodiments, the contaminating microorganism is a bacteriophage.

Bacteriophages are viruses that infect bacteria. Upon infecting a bacterial cell, they inject their DNA into the bacterium. After phage DNA entry into a bacterial cell, the phage can execute one of two pathways for propagation: the prophage or the lytic pathway. In the prophage pathway, the phage DNA is integrated into the bacterial chromosome and replicated along with it during cell division. In the lytic pathway, the phage uses bacterial cellular machinery to synthesize phage components (such as polypeptides and phage DNA) and eventually causes bacterial cell to lyse and thereby release viral particles to infect other bacterial cells. Phages in the prophage pathway may later be induced by environmental events to engage the lytic pathway (Alberts, B. et al. Molecular Biology of the Cell. 4 th ed. New York: Garland Science; 2002).

[0105] In some embodiments, the culture medium is treated with chlorite to reduce the growth rate, kill, or otherwise inhibit additional contaminating microorganisms. Due to the ability of bacteriophages to remain dormant during lysogeny inside bacteria (including bacteria expressing a chlorite dismutase polypeptide of the present disclosure), chlorite treatment may only be effective at killing or inactivating lytic bacteriophages. In the case of a contaminating bacteriophage, to reduce the growth rate, kill, or otherwise inhibit refers to reducing the incidence of bacterial infection and/or bacterial lysis by the bacteriophage.

[0106] The following examples are offered for illustrative purposes and to aid one of skill in better understanding the various embodiments of the disclosure. The following examples are not intended to limit the scope of the present disclosure in any way.

EXAMPLES

Example 1: Engineered chlorite resistance through expression of chlorite dismutase Introduction

[0107] Large-scale fermentation-based biofuel production is moving towards scaling bioprocesses beyond anything previously conceived. However, solvent producing processes, such as ethanol fermentation by yeast, or acetone-butanol-ethanol fermentation by Clostridium species, are renowned for their susceptibility to contamination and bacteriophage infection. Although disinfectants, such as hypochlorite, chlorite, or chlorine dioxide, can be added to process waters to prevent infection, these same disinfectants would also be bactericidal against the process organism. However, if the process organism is engineered to heterologously express the chlorite dismutase enzyme that dismutates chlorite into chloride and oxygen, then it would be protected from the bactericidal effects of any chlorite added as a suitable

disinfectant. [0108] The chlorite dismutase is a well-characterized enzyme that is primarily found in perchlorate reducing bacteria (PRB), where it is a primary component of the respiratory pathway of the organism (Coates, J. D. & Achenbach, L. A. Nat Rev Micro 2, 569-580 (2004)). PRBs couple the oxidation of reduced substrates (organic carbon, H 2 , H 2 S, or Fe(II)) to the reduction of perchlorate to generate energy for growth. The perchlorate is initially reduced by the perchlorate reductase enzyme (Per) into chlorite, which is subsequently dismutated into chloride and molecular oxygen by the chlorite dismutase (Cld) (FIG. 1).

[0109] Studies have identified the genes involved in this metabolism, demonstrated that this metabolism is the result of horizontal gene transfer, and by using comparative genomics have shown that this metabolism is encoded by a highly conserved perchlorate reduction genomic island (PRI) (FIG. 2) (Melnyk RA et al. (2011). Appl Environ Microbiol 77(20):7401-4).

Materials and Methods

Bacterial strains and plasmids

[0110] Azospira suillum PS (ATCC B AA-33/DSMZ 13638) was revived from a lab freezer stock and used as the wild-type strain for all genetic manipulations. Various E. coli strains were also used for cloning and conjugation purposes. Prior to all growth curves and genetic manipulations, all strains were streaked out from master freezer stocks to get single colonies.

Culture conditions and media

[0111] E. coli strains were grown in LB media. Kanamycin (Kan, 50 μg/mL) was used for selection and diaminopimelic acid (DAP, 0.3 mM) was used as a supplement to cultivate the auxotrophic strain WM3064. For routine culturing as well as growth assays, wild-type and mutant strains of PS were grown in ALP media. One liter of ALP media is composed of 0.49 g monobasic sodium phosphate dihydrate, 0.97 g dibasic anhydrous sodium phosphate, 0.1 g potassium chloride, 0.25 g ammonium chloride, 0.82 g sodium acetate, 2.0 g yeast extract, 7.6 g of a 60% w/w sodium lactate solution, 1.10 g sodium pyruvate and 10 mL of both vitamin mix and mineral mix. To make solid ALP media for plates, 15 g/L agar was added. Kanamycin was used for selection of PS mutants at a concentration of 50 μg/mL. For anaerobic growth of PS, ALP media was supplemented with either 5 mM sodium nitrate, 2.5 mM sodium perchlorate, or 5 niM of both sodium nitrate and sodium perchlorate. No growth was observed when no electron acceptor was added, confirming that PS is unable to grow via fermentation of the carbon sources present in ALP media. All strains of E. coli and PS were cultivated at 37°C. Anaerobic growth curves of PS for the transposon screen and characterization of strains was performed using a Spectramax 340PC384 plate reader in an anaerobic chamber (Coy Laboratory Products, Grass Lake, MI).

Results

[0112] A genetic system for studying the function of chlorite dismutase was established using the bacterial strain Azospira suillum strain PS (called "PS")- A deletion mutant was constructed, removing chlorite dismutase from this strain {Acid, based on the abbreviation for the chlorite dismutase gene, cld). Although this mutant has no growth defect when compared to the wild type strain PS when grown with oxygen as the sole electron acceptor, it is unable to grow with perchlorate or chlorate (FIG. 3).

[0113] However, the functional role of this enzyme goes beyond simply supporting a capacity to grow by perchlorate respiration by the host organism. The enzyme also plays a detoxification role for the host organism, protecting it against the bactericidal activity of chlorite in the environment. This is clearly shown by the significant increase in sensitivity to chlorite of the Acid mutant compared to the wild type strain PS when grown aerobically in the presence of 40 μΜ chlorite (FIG. 3). Under this condition, no growth is observed for the Acid mutant, while growth of the wild type strain is unimpaired in the presence or absence of chlorite.

[0114] To further demonstrate the protective effect of the chlorite dismutase to a host cell against chlorite, we recombinantly engineered Shewanella oneidensis strain MR-1 to

constitutively express the heterologous cld gene from Shewanella algae ACDC. When the aerobic growth of the engineered strain (ICC38) in the presence of chlorite was compared to that of the wild type strain containing an empty expression vector (strain RAM13), recombinant chlorite dismutase expression clearly promoted the growth of the host cell (FIG. 4). In this instance, 250 μΜ chlorite was sufficient to cause a significant reduction in the growth rate on the wild type strain, whereas the growth of the recombinantly engineered strain was unimpaired (FIG. 4). Even at chlorite concentrations as high as 1000 μΜ, the recombinantly engineered strain showed minimal growth impairment, whereas the growth of the wild type strain was completely inhibited (FIG. 4).

[0115] These results clearly demonstrate that not only does chlorite dismutase play an important role in the respiratory pathway of perchl orate, but it also plays an important detoxification role in host cells. Its expression in an engineered strain endowed the host cell with the ability to withstand toxic levels of chlorite present in the growth medium. Such

concentrations would prove inhibitory to more contaminating agents, such as bacteriophage, as it has recently been demonstrated that greater than 99% reduction in lytic phage particles can be achieved when the culture medium is treated with chlorite at a dose level of 15 mM (Bichai, F. and Barbeau, B. (2006). Water Qual Res J Canada 41(4):375-82).

Example 2: Chlorite dismutase expression protects cells from levels of chlorite toxic to contaminating microorganisms

Materials and Methods

P. chloritidismutans growth experiments

[0116] Pseudomonas chloritidismutans, a chlorate-reducing bacterium that expresses a native Cld protein (see, e.g., Wolterink, A.F., et al. (2002) Int. J. Syst. Evol. Microbiol. 52: 183- 90 and Mehboob, F., et al. (2009) FEMS Microbiol. Lett. 293:115-21), was grown in Luria Broth (LB). A sample of primary effluent from the East Bay Municipal Utility District water treatment plant was centrifuged at 4000 x g for 20 minutes to remove debris and passed sequentially through a 0.45 μπι filter and a 0.22 μπι filter to remove particles and microbes. The clarified sample was incubated with P. chloritidismutans and plated in LB top agar to detect plaques. A plaque was transferred to a 25 ml culture of P. chloritidismutans and incubated until cell lysis was observed (5-6 hours) before centrifugation and filtration to obtain a working phage stock.

[0117] For growth experiments, P. chloritidismutans was grown in LB medium to an OD 6 oo of 0.233 (selected from a dilution series made from an overnight culture). The culture was diluted to OD 6 oo = 0.02, split into aliquots, and treated with 60 μΜ chlorite, and/or 50 μΐ phage stock, as appropriate (control included no added components). Each condition was replicated in six wells of a microtiter plate incubated at 37°C. Optical density (600 nm) was recorded during growth in a plate reader.

Caulobacter crescentus growth experiments

[0118] Wild-type Caulobacter crescentus NA1000 (Marks, M.E., et al. (2010) J. Bacteriol. 192:3678-88) was grown in peptone-yeast extract medium (PYE; see Ely, B. (1991) Methods Enzymol. 204:372-84) supplemented with 0.1% (v/v) ferrous sulphate/chelate solution (Sigma #F0518). The plasmid pJS14 is a derivative of the broad-host-range cloning vector pBBRlMCS (Kovach, M.E., et al. (1994) Biotechniques 16:800-2), which confers resistance to

chloramphenicol. To express Cld from pJS14, the cld open reading frame from P.

chloritidismutans was placed behind a -430 base pair NcoI-EcoRI fragment of the xylX promoter from C. crescentus (Meisenzahl, A.C., et al. (1997) J. Bacteriol. 179:592-600). pJS14- /Xvc/J or the empty vector pJS14 was transformed into NA1000 with selection using 1 μg/ml chloramphenicol.

[0119] The two strains were grown separately in PYE/chloramphenicol/ferrous sulphate supplemented with 0.3% xylose to induce cld expression. Each culture also received 1 mM chlorite, 4 mM chlorite, or no additional treatment, and the increase in optical density (660 nm) was measured over time in a microtiter plate reader. Each strain/condition was assayed in six individual wells.

[0120] Caulobacter crescentus NA1000 harboring either pJS71 (KR260) or pJS14- /Xvc/J (KR3557) were grown in PYE medium supplemented with 0.1% ferrous sulphate/chelate (Sigma #F0518) and 0.3% xylose to induce cld expression. pJS71 is a derivative of the broad-host-range plasmid pBBRlMCS (Kovach, M.E., et al. (1994) Biotechniques 16:800-2), which confers resistance to spectinomycin. The two strains were inoculated together at a 1 : 1 ratio in

PYE/ferrous sulphate/xylose medium with or without 0.5 mM chlorite at 30°C. Samples were withdrawn from each co-culture at the indicated times, diluted, and plated on PYE medium supplemented with 1 μg/ml chloramphenicol or with 100 μg ml spectinomycin to enumerate survivors of each species.

Measurement of chlorite in culture [0121] KR260, KR3557, or a 1 : 1 mixture of the two strains were grown at 30°C in PYE medium supplemented with 0.1% ferrous sulphate/chelate, 0.3% xylose, and 0.5 mM chlorite. Samples were withdrawn from each culture at the indicated times, and the chlorite concentration was measured using the colorimetric assay described in EPA Method 327.

Expression vectors for chlorite dismutase or heme biosynthesis

[0122] The cld gene from Pseudomonas chloritidismutans DSM 13592 is cloned into a series of plasmids, such that each plasmid contains cld under the control of one promoter selected from pTDH3, pTEFl, pRPL18, pRNR2, and pREVl. Each of the plasmids contains a CEN6 element for stable transmission and complements a uracil auxotrophy with the URA3 gene. The host strain is BY4741 (MATa his3Al leu2A0 metl5A0 ura3A0).

[0123] heniE, hemN, hemG, and hemli genes from Escherichia coll are cloned into a plasmid under the control of a single promoter. The sequence of the ribosome binding site upstream of each gene may be varied to adjust expression levels of individual genes. The entire operon is integrated into the C. acetohutylic m chromosome of C. acetobutylicum strain ATCC 824 using the described technique for homologous recombination (Heap, J.T., et al. (2012) Nucleic Acids Res. 40:e59).

[0124] In addition to the above E. coll genes, homologous genes from. Klebsiella pneumonia, Vibrio parahemolyticus (particularly as these contain HemG rather than HemY), Bacillus sublllis, Lisleria monocytogenes, Staphylococcus epidermidis, Streptococcus mutans, or from Gram-positive firmicutes more closely related to C. acetobutylicum may also be tested.

Results

[0125] To demonstrate that chlorite dismutase expression may protect cells from levels of chlorite that inactivate contaminating agents such as bacteriophages, a naturally chlorate- reducing strain of Pseudomonas chloritidismutans was tested. This strain is known to express chlorite dismutase. An as-yet uncharacterized bacteriophage that attacks this strain was isolated from a MUD anaerobic sludge digester. [0126] The growth of this P. chloritidismutans strain was tested in the presence and absence of chlorite. As shown in FIG. 5, it was observed that 60 μΜ chlorite alone did not hinder the growth of P. chloritidismutans, nor did the heat-killed phage. Live phage blocked the growth of P. chloritidismutans, but importantly, the addition of chlorite and phage together resulted in a wild-type growth rate. This result demonstrates that the combination of Cld expression and chlorite addition can protect P. chloritidismutans from phage-mediated killing.

[0127] Because P. chloritidismutans growth was not completely inhibited by the

bacteriophage, and to test this approach in a bacterium that does not endogenously express chlorite dismutase, chlorite dismutase from A. suillum PS was expressed in the oligotrophic alpha-proteobacterium Caulobacter crescentus. The growth of wild-type Caulobacter was completely blocked in the presence of ImM or 4mM chlorite (FIG. 6). However, when chlorite dismutase expression by introduced into this Caulobacter strain on a medium-copy plasmid under the control of a xylose-inducible promoter, the strain was resistant to both ImM and 4mM chlorite, growing at the same rate in medium with ImM or 4mM chlorite as compared to control medium lacking chlorite (FIG. 7). Therefore, while both concentrations of chlorite killed C. crescentus cells harboring pJS14, the strain expressing Cld grew at nearly wild-type rates in either 1 mM or 4 mM chlorite, indicating that Cld expression protects C. crescentus from the toxic effects of chlorite. These results demonstrate that heterologous expression of chlorite dismutase in a cell that does not normally express the enzyme protects the growth of the cell from chlorite toxicity. It was noted that Caulobacter cells expressing chlorite dismutase did not grow well unless the medium was supplemented with chelated ferrous iron, suggesting the presence of iron may be required for activity of the heme-containing chlorite dismutase.

[0128] Next, the ability of chlorite to kill or inhibit the growth of non-chlorite dismutase- expressing contaminants was tested. Caulobacter strains KR260 (empty plasmid) and KR3557 (chlorite dismutase expression plasmid) were co-cultured in the presence of 0.5mM chlorite. In the co-culture, the presence of empty plasmid (containing a spectinomycin resistance marker) or chlorite dismutase expression plasmid (containing a chloramphenicol resistance marker) was not selected. Rather, both strains were co-cultured in PYE medium without selection, samples were taken, and the number of surviving colonies that were resistant to spectinomycin (KR260) or chloramphenicol (KR3557) were counted at the indicated times (FIG. 8). [0129] In the culture without chlorite, KR260 and KR3557 grew and divided at similar rates, while in the culture containing chlorite, KR3557 grew at a slightly reduced rate, and the growth of KR260 was completely blocked (FIG. 8). This experiment demonstrates that in the presence of chlorite, Cld-expressing bacteria can be selectively spared, while the growth of bacteria lacking Cld is greatly inhibited.

[0130] Because the above experiments were performed in batch culture, the concentration of chlorite present in the medium was examined over time to determine how quickly added chlorite is cleared from cultures of chlorite dismutase-expressing or non-expressing strains. Control KR260 and chlorite dismutase-expressing strain KR3557 were cultured individually and together in a starting concentration of 500μΜ chlorite (FIG. 9). As shown in FIG. 9, the rate of chlorite disappearance was similar in the three cultures, even though one culture (KR260) contained no organisms with Cld activity. Also, the chlorite did not immediately react to form other products upon addition to the culture medium, as -0.3 mM chlorite was still present in each culture after a period of six hours.

[0131] These results demonstrate that chlorite dismutase expression protects host cells from levels of chlorite that are sufficient to inactivate contaminating microorganisms, such as bacteriophages and non-chlorite dismutase-expressing bacteria.

Example 3: Vectors for expression of chlorite dismutase

[0132] The previous Example demonstrates that chlorite dismutase expression may be used as a tool to reduce the growth of or kill contaminating microorganisms, including bacteriophages and bacterial strains. In order to apply this discovery to a variety of process organisms of interest, there is a need for expression vectors to express chlorite dismutase and related enzymes at appropriate levels.

[0133] To meet this need, a set of centromere-based plasmids designed to express chlorite dismutase at different levels in Saccharomyces cerevisiae is designed. As depicted in FIG. 10, all of these expression plasmids contain a promoter, chlorite dismutase coding sequence coupled to the ADH1 terminator, a selectable marker (e.g., URA3), and a centromere sequence (e.g., CEN6). Exemplary promoters include pTDH3, pTEFl, pRPL18, pRNR2, and pREVl. Each plasmid is transformed into S. cerevisiae strains BY4741 and BY4742, as well as industrially relevant strains. For each transformant, chlorite sensitivity is measured under aerobic and anaerobic conditions, and cell extracts are tested for chlorite dismutase expression by Western blotting.

[0134] Chlorite sensitivity in unmodified yeasts and in those expressing Cld are measured first in aerobic batch culture with concentrations of chlorite up to 20 mM. Growth is measured as an increase in optical density (600 nni). If a strain expressing Cld is found to have increased tolerance to one or more concentrations of chlorite, the growth of the same unmodified and Cld- expressing strains in anaerobic fermentation conditions will be measured, with and without added chlorite. To distinguish between blocked growth and cell killing, colony- forming units on plates lacking chlorite are measured during each of the growth experiments described above.

[0135] Chlorite dismutase expression is also tested in industrial fermenting strains, such as Clostridium strains {e.g., C. acetobutylicum) that perform acetone-butanol-ethanol fermentation. However, Clostridium strains are obligate fermenters that do not naturally produce cytochromes or heme. An exemplary pathway for heme biosynthesis is provided in FIG. 11. C.

acetobutylicum contains genes encoding a partial heme biosynthesis pathway, covering reactions 1-3 in FIG. 11. This partial pathway leads to the production of the heme intermediate uroporphyrinogen III, which is subsequently converted to siroheme or corrinoids.

[0136] Clostridium acetobutylicum ATCC 824 contains the genes encoding HetnB, HemC, and He D, which together convert aminolevulinic acid to uroporphyrinogen III. However, C. acetobutylicum lacks genes for the enzymes that convert uroporphyrinogen III to protoheme IX. The first step in the pathway, catalyzed by HemE, converts uroporphyrinogen III to

coproporphyrinogen III and carbon dioxide. The second step, catalyzed either by HemN or HemF, converts coproporphyrinogen III to protoporphyrinogen IX and carbon dioxide. The third step, catalyzed by HemG or HemY, converts protoporphyrinogen IX to propoporphyrin IX. The final step, catalyzed by Heml , adds iron to protoporphyrin IX to form protoheme IX. Step 2 can be catalyzed by an oxygen-dependent enzyme (HemF) or an oxygen-independent enzyme (HemN). Step 3 can also be catalyzed by oxygen- dependent (HemY) or oxygen -inde endent (HemG) enzymes. The oxygen-independent enzyme in each case may be tested first, in keeping with the fermentative lifestyle of C. acetobutylicum.

[0137] To express a functional, heme-containing chlorite dismutase in Clostridium, enzymes that catalyze reactions 4-7 in FIG. 11 are expressed. As described above, step 4 is catalyzed by uroporphyrinogen III decarboxylase, encoded by hemE. Step 5 is catalyzed by

coproporphyrinogen III oxidase (CPO). The C. acetobutylicum genome appears to encode two copies of the oxygen-independent CPO hemN, although their expression and function in C. acetobutylicum has not been tested. Step 6 is catalyzed by protoporphyrinogen IX oxidase, which is present in oxygen-dependent and -independent (e.g., hemG) forms. Step 7 is catalyzed by ferrochelatase (e.g., hemH).

[0138] Constructs for expressing these enzymes are generated and integrated into C.

acetobutylicum as described above. As sources for the genes in these pathways, the hemE, hemN, hemG, and hernH genes from Escherichia coii are tested first. Without wishing to be bound to theory, it is thought that these enzymes have coevolved in the same organism and thus may work better together than enzymes from disparate sources. However, genes from other Gram- negative organisms such as Klebsiella pneumoniae and Vibrio parahemolyticus

(particularly as these contain HemG rather than HemY), or from Gram-positive firmicutes more closely related to C. acetobutylicum may also be used. Non-limiting examples of firmicute species containing some or ail of the relevant genes include Bacillus suhtilis, Listeria monocytogenes, Staphylococcus epidermidis, and Streptococcus mutatis, as described herein.

[0139] Recombinant C. acetobutylicum strain(s) are grown and tested under anaerobic conditions. The presence of heme, porphyrins, and/or heme pathway intermediates in C.

acetobutylicum strains expressing a heme biosynthesis pathway is measured in extracts using two methods: reverse-phase chromatography followed by thin-layer chromatography (Hansson M. and Hederstedt L. (1994) .). Bacterid. 176:5962-70), and high-performance liquid

chromatography (Franken ACW et al, (2013) Appl. Microbiol. Biotechnol. 97:9773-85).

Production of chlorite dismutase protein in C. acetobutylicum strains expressing a heme biosynthesis pathway is monitored using a chlorite dismutase antibody. Heme bound to chlorite dismutase expressed in C. acetobutylicum strains expressing a heme biosynthesis pathway is detected and quantified (Nygaard TK et al. (2006) BMC Microbiol. 6:82). Methods will be used to determine if the strain(s) perform solvenlogenic fermentation in the presence of added chlorite.

[0140] Tolerance to chlorite in the culture medium is tested by growing a C. acetobutylicum strain expressing a heme biosynthesis pathway and a chlorite dismutase in varying

concentrations of chlorite (as described above), as compared to growth in culture medium lacking chlorite. These growth rates are compared to the growth rate of a C. acetobutylicum strain lacking one or more components of the heme biosynthesis pathway and/or the chlorite dismutase {e.g., a strain that lacks the appropriate coding sequence, or a strain in which expression of the appropriate coding sequence has not been induced) grown in culture medium with the same concentrations of chlorite. ABE fermentation in a C. acetobutylicum strain expressing a heme biosynthesis pathway and a chlorite dismutase grown in a cell culture medium with varying concentrations of chlorite is compared with ABE fermentation of the same strain in cell culture medium lacking chlorite. ABE fermentation of this strain is also compared with the ABE fermentation of a C. acetobutylicum strain lacking one or more components of the heme biosynthesis pathway and/or the chlorite dismutase {e.g., a strain that lacks the appropriate coding sequence, or a strain in which expression of the appropriate coding sequence has not been induced) grown in culture medium with the same concentrations of chlorite.