Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2023 Feb 1.
Published in final edited form as: Curr Opin Microbiol. 2021 Oct 22;65:1–7. doi: 10.1016/j.mib.2021.09.007

Animal models for dissecting Vibrio cholerae intestinal pathogenesis and immunity

Brandon Sit a,b, Bolutife Fakoya a,b, Matthew K Waldor a,b,c,d,*
PMCID: PMC8792189  NIHMSID: NIHMS1743345  PMID: 34695646

Abstract

The human diarrheal disease cholera is caused by the bacterium Vibrio cholerae. Efforts to develop animal models that closely mimic cholera to study the pathogenesis of this disease began >125 years ago. Here, we review currently used non-surgical, oral inoculation-based animal models for investigation of V. cholerae intestinal colonization and disease and highlight recent discoveries that have illuminated mechanisms of cholera pathogenesis and immunity, particularly in the area of how V. cholerae interacts with the gut microbiome to influence infection. The emergence of high-throughput tools for studies of pathogen-host interactions, along with continued advances in host genetic engineering and manipulation in animal models of V. cholerae will deepen understanding of cholera pathogenesis, uncovering knowledge important for control of this globally important bacterial pathogen.

Keywords: Cholera, Vibrio cholerae, animal models

Introduction

The Gram-negative bacterium Vibrio cholerae causes cholera, a severe human diarrheal illness that remains endemic in >50 countries [1]. The ongoing 7th cholera pandemic is estimated to cause >3 million cases annually and is caused by ‘variant’ El Tor V. cholerae O1 strains that are continually evolving [2]. The secretory diarrhea that is the hallmark of cholera is largely explained by the actions of cholera toxin (CT) [1], an AB5-type toxin secreted by V. cholerae in the small intestine (SI), the site where the pathogen primarily colonizes the human intestine and causes disease [3]. While many pathogen factors facilitate SI colonization, a bacterial surface appendage - toxin-coregulated pilus (TCP) - is V. cholerae’s chief colonization factor [3]. The mechanisms by which TCP enables colonization are incompletely understood.

The self-limiting nature of cholera has prompted the development of a controlled human infection model (CHIM), where V. cholerae is orally administered to volunteers [4]. CHIM studies have revealed the requirements for CT for disease, TCP’s critical role in intestinal colonization, enabled investigations of immunity to V. cholerae, and demonstrated the relative efficacy of killed and live oral cholera vaccines (OCVs) [4,5]. However, CHIM work is limited by several logistical and ethical considerations. Many mechanistic questions regarding V. cholerae-host interactions in the SI cannot be readily answered in this human model. CHIM studies also cannot currently be conducted in cholera-endemic countries, potentially limiting the applicability of conclusions from volunteer studies in non-endemic regions [6]. Instead, efforts to understand V. cholerae pathogenesis have led to the development of over a dozen different animal models for this pathogen. Historically, many different medium and large mammalian species ranging from guinea pigs to non-human primates have been used, but these suffer some of the same limitations as the CHIM (reviewed in [7]). The field is now largely driven by small mammal and non-mammalian/invertebrate models.

The utility of any given animal model can be gauged by the extent to which it recapitulates human disease. In our view, it is useful to stratify currently used models (Table 1) by how well they reflect two critical aspects of human cholera pathogenesis - TCP-dependent SI colonization and CT-dependent diarrheal illness. Although models that lack either or both criteria can yield important information, the direct applicability of this knowledge to understanding cholera is unclear. In this review, we use this framework to present and discuss current oral, non-surgical infection models of V. cholerae. We illustrate how new discoveries from the last several years across different models illuminate themes in V. cholerae research and identify open areas of investigation for deepening understanding of cholera pathogenesis and immunity.

Table 1.

Major non-surgical oral infection animal models for V. cholerae.

TCP-dependent colonization CT-dependent diarrhea ST coloniza tion** Genetic host manipulation Throughput/accessibility Application(s)***
Mammalian
M. musculus (infant mice) + + + ++ ++ Investigation of putative colonization factors
M. musculus (germfree/strept omycin-treated adult mice) ++ ++ Adaptive immunity/vaccination, commensal-pathogen interactions
O. cuniculus (infant rabbits) + + + + (rare) + High-throughput and “omics” approaches
H. sapiens (human volunteers) + + + Clinical-stage investigations
Non-mammalian and Invertebrate
D. melanogaster (fruit flies) * N/A +++ +++ Investigations of host defense and metabolism
C. elegans (nematodes) N/A +++ +++ Identification of putative accessory V. cholerae toxins
D. rerio (zebrafish) N/A +++ +++ Tissue imaging and commensal- pathogen interactions
*

CT-dependent mortality

**

SI as the primary site of pathogen colonization

***

Applications are not unique to models and indicate a representative use of the species

Scale: − (lowest) to +++ (highest) (columns with only +/− indicate presence or absence/unknown)

Insights into V. cholerae pathogenesis from animal models

The acuity of cholera is well-captured by short-term (~1 day) animal infection models. Conventionally reared, specific-pathogen free (SPF) adult mice (Mus musculus) are commonly used to study bacterial pathogens but are resistant to intestinal colonization by orally-inoculated V. cholerae, likely because of colonization resistance from the resident murine gut microbiota. Streptomycin-treated adult mice can be colonized by V. cholerae, but the pathogen is found almost exclusively in the colon and not the SI; moreover, intestinal colonization appears to be TCP-independent, and the animals do not develop diarrhea [8,9]. Alternate adult mouse models for oral V. cholerae infection (clindamycin or ketamine pretreatment) that may surmount these obstacles have been reported, but have yet to be widely adopted [10,11].

The dominant model of V. cholerae pathogenesis is the 3–5-day-old infant mouse, where oral gavage of V. cholerae leads to TCP-dependent SI (but not colon) colonization and in some variations, CT-dependent diarrheal illness within ~16 hours [3,12]. Infant mice are a logistically simple system for addressing many targeted questions about the pathogen. This model has enabled limited-throughput studies of within-host virulence regulation, such as the use of engineered V. cholerae to identify in vivo-activated and -repressed loci, and the use of clinical V. cholerae isolates to understand how ongoing genetic variation in 7th pandemic strains impacts pathogenicity [1216]. Advanced tissue imaging of the infant mouse SI has revealed the sub-intestinal localization patterns of V. cholerae during infection, and spatiotemporal control of virulence gene regulation [18,19]. The most common use of this model has been to validate suspected colonization factors with competitive index studies, where a mutant strain is competed against a WT strain in vivo [3]. Competition studies have recently highlighted the contributions of fatty acid and carbon metabolism, cell wall maintenance and V. cholerae LPS modifications to intestinal colonization [9,2023].

A key limitation of infant mice is the relative scarcity of biological material for high-throughput, hypothesis-generating studies such as transcriptomics and whole-genome functional genetic screens. The infant rabbit (Oryctolagus cuniculus) model of cholera, which exhibits rapid CT-dependent lethal diarrheal illness along with TCP-dependent SI colonization, has filled this niche in V. cholerae animal studies [24]. Up to 1010 in vivo V. cholerae can be rapidly collected from the ~1 mL of diarrheal fluid that accumulates in the infant rabbit cecum, providing a valuable reagent for high-throughput analyses. The chemical composition of this fluid is highly similar to that of human choleric diarrhea [24]. Infant rabbit studies have revealed the genetic landscape of colonization factors in V. cholerae through transposon-insertion sequencing screens, which are largely limited by bottlenecks in infant mice [2528]. Infant rabbits have enabled the acquisition of high-resolution in vivo V. cholerae RNA-seq, metabolomic, and proteomic datasets, as well as insights into V. cholerae population dynamics during infection [2931]. A recent transcriptomic study in infant rabbits revealed novel roles for CT in the shaping of the pathogen’s nutritional microenvironment [32]. This and other studies have illustrated the value of combining animal models for complementary purposes - discovery experiments in infant rabbits and targeted competition assays in infant mice. Combined approaches offer considerable practical advantages, as infant rabbits are relatively expensive and large-scale experiments with these animals are technically challenging.

The most commonly used oral non-mammalian species for studying V. cholerae are fruit flies (Drosophila melanogaster), zebrafish (Danio rerio), and nematodes (Caenorhabditis elegans). Although flies can exhibit CT-dependent mortality, infection and host killing in these models is otherwise neither CT- nor TCP-dependent [3335]. Despite these important limitations, these models offer increased throughput, lowered cost, and ease of host genetic manipulation relative to infant mice. As the Drosophila intestinal immune system bears similarity to that of mammals, flies have been used to investigate host responses to V. cholerae, including pathogen manipulation of host lipid and carbon metabolism and how V. cholerae quorum sensing controls host outcomes [3639]. Studies in C. elegans have identified putative accessory V. cholerae virulence factors [35,40], and work in D. rerio aided by tissue imaging has been informative regarding V. cholerae’s interaction with gut symbionts and the intestinal epithelium [4143]. However, as most non-mammalian studies remain to be validated in either human or small mammal cholera models, their applicability to our understanding of cholera is limited.

An emerging theme in animal-based investigations of V. cholerae is the pathogen’s relationship with the intestinal microbiome. One of V. cholerae’s primary modes of interaction with other microbes is its type 6 secretion system (T6SS), an apparatus that mediates interbacterial antagonism [44]. Recent studies across several models using defined intestinal microbial communities have demonstrated the importance of the T6SS in mediating intestinal colonization, and dissected its role in creating colonizable niches for V. cholerae [25,41,42,4547]. Investigations in infant and adult murine models with altered gut microbiota have been complemented by efforts to identify microbiome changes in humans exposed to V. cholerae [4850]. Intestinal microbes that can promote or inhibit V. cholerae intestinal colonization in mice have been identified, and reconstitution of germ-free (GF) mice with these microbes offers an in vivo system in which to investigate interbacterial interactions (recently reviewed in [51]). The mechanisms underlying these phenotypes remain to be extensively characterized, but several appear to involve regulation of V. cholerae virulence programs by commensal-derived metabolites like secondary bile acids [10,50,52]. An important caveat to these findings is that V. cholerae intestinal colonization occurs primarily in the colon in GF mice. V. cholerae’s intricate relationship with intestinal microbes has also been hinted at by studies in infant rabbits that demonstrated V. cholerae is conjugation-competent in the intestine, and that pre-colonization with an avirulent V. cholerae live vaccine strain or V. cholerae-targeting phages can limit subsequent infection [28,47,53].

Insights into immunity to V. cholerae from animal models

Samples from cholera patients and volunteers in CHIM studies have been instrumental in the investigation of immune responses to V. cholerae. Observations that cholera bestows protection against future infection inspired development of currently used killed OCVs. However, there are significant limitations associated with killed OCVs, including their limited efficacy in young children; furthermore, many gaps remain in our understanding of the scope and mechanisms of protective immunity to V. cholerae [54]. Most questions in OCV development and immunity to cholera concern long-term adaptive immunity, but infant mammals and invertebrates have immature (or absent) adaptive immune systems. As such, adult mice have become the model of choice for studies of adaptive immunity to V. cholerae. Although adult mice have well-characterized and easily manipulated immune systems, mimicking OCV vaccination or natural infection by the oral route in this model is constrained by several critical limitations described above. These models exhibit no discernable illness, TCP-independent colonization, and colonic-biased pathogen localization, and GF mice are additionally confounded by sustained (months-years) V. cholerae colonization [8,55]. CHIM studies have shown that TCP-dependent intestinal colonization is critical for immunogenicity of live V. cholerae [5], and interpretation of immune responses observed in adult mice is complicated since the colon lacks immune features of the SI such as Peyer’s patches. Despite these shortcomings, viable alternatives to adult mice for studies of adaptive immune responses to V. cholerae are currently lacking.

Orally-infected adult mice can be used to profile diverse immune responses to V. cholerae. Many assays used in humans that report on known correlates of protection against cholera, such as antigen-specific ELISAs for CT and TCP, and the vibriocidal antibody titer assay, can be directly applied to murine samples. Immunological metrics can be read out from serum and fecal samples over time from the same mouse, enabling kinetics of immunity to be dissected [56]. Genetically modified adult mice could be used to explore the necessity and sufficiency of host factors in the development of immunity to V. cholerae, but these investigations have been rare [57].

Sustained V. cholerae colonization in GF adult mice precludes their rechallenge with virulent V. cholerae, a critical measure of vaccine efficacy. However, since colonization is transient in antibiotic-treated mice, rechallenge after clearance is possible [8,58]. While pulmonary inoculation of V. cholerae to circumvent obstacles associated with oral infection has been described, the translational significance of observations with this route of infection are unclear [59]. It is instead possible to use infant mice as proxy hosts to interrogate immunity in vaccinated dams, as well as to use V. cholerae pre-mixed with serum from immunized mice as an infant mouse challenge inoculum. These approaches, combined with readouts of immunogenicity, have collectively proven useful to test the protective efficacy of cutting-edge cholera vaccine candidates, including new live OCV strains, outer membrane vesicle-based formulations, and transcutaneous conjugate vaccines [23,56,60,61]. These studies underscore the importance of combining appropriate models (adult immunization with infant challenge) in the investigation of cholera vaccines and immunity. Standardization and consolidation of immunization schema and efficacy measures will aid these efforts [23].

The throughput and ease of host manipulation in mouse models makes them well-suited for targeted investigations of hypotheses derived from observations of human immune responses. For example, clinical investigations have suggested that immune responses to the V. cholerae O-antigen are the protective factor against reinfection [54]. Infant mice have been used by multiple groups to test and ultimately support this hypothesis with a variety of strategies, including direct isolation and in vivo testing of human-derived monoclonal antibodies [62,63]. A transgenic mouse line expressing a V. cholerae core/lipid A-targeted monoclonal antibody was developed to evaluate the sufficiency of this target for protection from intestinal colonization [64]. These examples demonstrate how hypotheses from human studies can be mechanistically evaluated using engineered animals. Similarly, the emerging concept that the intestinal microbiome shapes human immune responses to V. cholerae [65] is starting to be addressed using defined human microbial communities in GF mice. For example, prebiotic and microbial treatments in a GF model of CT-based vaccination were recently shown to enhance immune responses to this V. cholerae antigen [66]. Insights from mouse models may not only provide explanations for observations regarding human immunity but offer useful platforms to test interventions for future translation back to the clinic.

Conclusions

Despite the long history of studies of cholera pathogenesis, many questions regarding V. cholerae-host interactions remain (Table 2). Metchnikoff recognized the importance of ‘favorizing microbes’ in promoting experimental cholera in 1894 [67], but elucidation of the tripartite host-pathogen-commensal interaction axis during infection is just beginning. Surprisingly, despite the use of infant and adult mice to study V. cholerae for decades, experiments with genetically modified hosts are almost entirely lacking in the study of both pathogenesis and immunity. Although human loci conferring susceptibility to cholera have been identified [68,69], there are relatively few studies that use genetically engineered mice to investigate host innate defense against V. cholerae [7072]. Such studies will be valuable because cholera can be one of the most rapidly fatal acute infections and innate rather than adaptive responses may determine outcomes. Defining the innate immune pathways that are modulated by V. cholerae infection, and whether these responses are beneficial or detrimental to the host, should be feasible in engineered newborn mice despite the immaturity of their adaptive immune systems.

Table 2.

Future questions for animal models in the study of V. cholerae.

Pathogenesis and host response • Why is V. cholerae primarily a small intestinal pathogen?
• Why is TCP required for V. cholerae pathogenesis?
• How does CT modulate host responses beyond diarrhoeagenesis?
• How does V. cholerae intestinal colonization modify intestinal epithelial function?
• What are the molecular pathways that underlie the host -induced ‘hyperinfectious’ transmission state?
Microbiome- V. cholerae • How do the murine and human gut microbiomes prevent V. cholerae intestinal colonization?
• Why does V. cholerae display colonic tropism in mice lacking intestinal microbes?
• How does the gut microbiome control immune responses to V. cholerae and OCVs?
Innate Immunity • What pathways of innate immunity contribute to protection against or resolution of cholera?
• What is the role of the inflammasome and/or pyroptosis at the intestinal epithelial surface in defense against cholera?
• What is the function of suspected innate immune cholera susceptibility loci such as LPLUNC1?
Adaptive Immunity • Can a TCP-dependent animal model of adaptive immunity be developed?
• What are the pathways of adaptive immunity and cellular populations that contribute to protection against cholera? Do they differ in naturally-infected versus vaccinated hosts?
• Why do young children have poor memory responses to OCV immunization?
• Can non-mucosal immunization routes confer long term immunity to cholera?

Another key open area of investigation is V. cholerae host-to-host transmission. There is no controlled V. cholerae animal model of fecal-oral transmission. Approximate models of transmission such as transfer of infected intestinal homogenates or diarrheal fluid gavage have revealed a transient host-priming (“hyperinfectious”) phenotype of host-derived V. cholerae [73,74]. The mechanism(s) underlying this phenomenon, which could be critical to understanding the explosive kinetics of cholera outbreaks, remains unclear. Adaptation of new barcoding frameworks in future studies of transmission in animals will facilitate understanding of how particular regions of the intestine, including the colon, contribute to host-priming [75]. It is likely that studies involving host-, pathogen-, commensal-, and environment-targeted permutations across differing animal models will be needed to understand the biology of cholera transmission.

The power of animal models for understanding V. cholerae is heightened by the broad and accessible genetic toolkit for this bacterium, including multiplex genetic engineering and numerous reporter systems. The continued integration of data from different models and implementation of cutting-edge genome-scale techniques from other fields, such as single-cell sequencing and transcriptomics, will be crucial to construct a holistic view of cholera pathogenesis and to reveal novel therapeutic opportunities against V. cholerae and other bacterial pathogens.

Highlights.

  • Mammalian, non-mammalian, and invertebrate species are currently used to model Vibrio cholerae pathogenicity.

  • Models vary considerably in their recapitulation of V. cholerae-host interactions in the human intestine.

  • Recent work in animals has revealed molecular details of the host-V. cholerae-microbiome axis.

  • Open areas for investigation include intestinal innate defense against V. cholerae and determinants of cholera transmission.

Acknowledgements

We acknowledge the numerous and invaluable contributions to the field by groups whose work we did not have the space to cite and discuss in this review. Research in the Waldor Lab is funded by the National Institutes of Health (R01-AI-042347), the Wellcome Trust (218443/Z/19/Z) and the Howard Hughes Medical Institute.

Footnotes

Declaration of interests

The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper.

Declaration of Interest: none

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

References

  • 1.Clemens JD, Nair GB, Ahmed T, Qadri F, Holmgren J: Cholera. Lancet (London, England) 2017, 6736:1–11. [DOI] [PubMed] [Google Scholar]
  • 2.Ali M, Nelson AR, Lopez AL, Sack DA: Updated global burden of cholera in endemic countries. PLoS Negl Trop Dis 2015, 9:1–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Ritchie JM, Waldor MK: Vibrio cholerae interactions with the gastrointestinal tract: lessons from animal studies. Curr Top Microbiol Immunol 2009, 337:37–59. [DOI] [PubMed] [Google Scholar]
  • 4.Shirley D-AT, McArthur MA: The utility of human challenge studies in vaccine development: lessons learned from cholera. Vaccine (Auckland, NZ) 2011, 2011:3–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Herrington DA, Hall RH, Losonsky G, Mekalanos JJ, Taylor RK, Levine MM: Toxin, toxin-coregulated pili, and the toxR regulon are essential for Vibrio cholerae pathogenesis in humans. J Exp Med 1988, 168:1487–92. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Darton TC, Blohmke CJ, Moorthy VS, Altmann DM, Hayden FG, Clutterbuck EA, Levine MM, Hill AVS, Pollard AJ: Design, recruitment, and microbiological considerations in human challenge studies. Lancet Infect Dis 2015, 15:840–51. [DOI] [PubMed] [Google Scholar]
  • 7.Richardson SH: Animal Models in Cholera Research. In Vibrio cholerae and Cholera: Molecular to Global Perspectives. Edited by Wachsmuth K, Blake P, Olsvik O. American Society for Microbiology; 1994:203–226. [Google Scholar]
  • 8.Nygren E, Li B-L, Holmgren J, Attridge SR: Establishment of an adult mouse model for direct evaluation of the efficacy of vaccines against Vibrio cholerae. Infect Immun 2009, 77:3475–84. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Wang H, Xing X, Wang J, Pang B, Liu M, Larios-Valencia J, Liu T, Liu G, Xie S, Hao G, et al. : Hypermutation-induced in vivo oxidative stress resistance enhances Vibrio cholerae host adaptation. PLoS Pathog 2018, 14:e1007413. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.You JS, Yong JH, Kim GH, Moon S, Nam KT, Ryu JH, Yoon MY, Yoon SS: Commensal-derived metabolites govern Vibrio cholerae pathogenesis in host intestine. Microbiome 2019, 7:132. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Olivier V, Queen J, Satchell KJF: Successful small intestine colonization of adult mice by Vibrio cholerae requires ketamine anesthesia and accessory toxins. PLoS One 2009, 4:e7352. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Angelichio MJ, Spector J, Waldor MK, Camilli A: Vibrio cholerae intestinal population dynamics in the suckling mouse model of infection. Infect Immun 1999, 67:3733–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Lee SH, Butler SM, Camilli A: Selection for in vivo regulators of bacterial virulence. Proc Natl Acad Sci U S A 2001, 98:6889–6894. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Cakar F, Zingl FG, Moisi M, Reidl J, Schild S: In vivo repressed genes of Vibrio cholerae reveal inverse requirements of an H+/Cl− transporter along the gastrointestinal passage. Proc Natl Acad Sci U S A 2018, 115:E2376–E2385. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Son MS, Megli CJ, Kovacikova G, Qadri F, Taylor RK: Characterization of Vibrio cholerae O1 El Tor biotype variant clinical isolates from Bangladesh and Haiti, including a molecular genetic analysis of virulence genes. J Clin Microbiol 2011, 49:3739–49. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Satchell KJF, Jones CJ, Wong J, Queen J, Agarwal S, Yildiz FH: Phenotypic Analysis Reveals that the 2010 Haiti Cholera Epidemic Is Linked to a Hypervirulent Strain. Infect Immun 2016, 84:2473–81. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Ghosh P, Sinha R, Samanta P, Saha DR, Koley H, Dutta S, Okamoto K, Ghosh A, Ramamurthy T, Mukhopadhyay AK: Haitian Variant Vibrio cholerae O1 Strains Manifest Higher Virulence in Animal Models. Front Microbiol 2019, 10:111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Millet YA, Alvarez D, Ringgaard S, von Andrian UH, Davis BM, Waldor MK: Insights into Vibrio cholerae intestinal colonization from monitoring fluorescently labeled bacteria. PLoS Pathog 2014, 10:e1004405. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19. Gallego-Hernandez AL, DePas WH, Park JH, Teschler JK, Hartmann R, Jeckel H, Drescher K, Beyhan S, Newman DK, Yildiz FH: Upregulation of virulence genes promotes Vibrio cholerae biofilm hyperinfectivity. Proc Natl Acad Sci U S A 2020, 117:11010–11017.32355001 * This study used tissue clearing approaches to image intestinal V. cholerae in infant mice, providing insights into how virulence gene upregulation may contribute to hyper-infectivity.
  • 20.Yang S, Xi D, Wang X, Li Y, Li Y, Yan J, Cao B: Vibrio cholerae VC1741 (PsrA) enhances the colonization of the pathogen in infant mice intestines in the presence of the long-chain fatty acid, oleic acid. Microb Pathog 2020, 147:104443. [DOI] [PubMed] [Google Scholar]
  • 21.Hayes CA, Dalia TN, Dalia AB: Systematic genetic dissection of PTS in Vibrio cholerae uncovers a novel glucose transporter and a limited role for PTS during infection of a mammalian host. Mol Microbiol 2017, 00:1–39. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Fleurie A, Zoued A, Alvarez L, Hines KM, Cava F, Xu L, Davis BM, Waldor MK: A Vibrio cholerae BolA-Like Protein Is Required for Proper Cell Shape and Cell Envelope Integrity. MBio 2019, 10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23. Sit B, Fakoya B, Zhang T, Billings G, Waldor MK: Dissecting serotype-specific contributions to live oral cholera vaccine efficacy. Proc Natl Acad Sci U S A 2021, 118:1–9. ** This study developed a framework for evaluating live cholera vaccine candidates in a GF adult mouse model combined with infant mouse challenges, and showed how O-antigen modifications could influence OCV efficacy.
  • 24.Ritchie JM, Rui H, Bronson RT, Waldor MK: Back to the future: studying cholera pathogenesis using infant rabbits. MBio 2010, 1:1–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Fu Y, Waldor MK, Mekalanos JJ: Tn-Seq analysis of Vibrio cholerae intestinal colonization reveals a role for T6SS-mediated antibacterial activity in the host. Cell Host Microbe 2013, 14:652–63. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Kamp HD, Patimalla-Dipali B, Lazinski DW, Wallace-Gadsden F, Camilli A: Gene fitness landscapes of Vibrio cholerae at important stages of its life cycle. PLoS Pathog 2013, 9:e1003800. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Pritchard JR, Chao MC, Abel S, Davis BM, Baranowski C, Zhang YJ, Rubin EJ, Waldor MK: ARTIST: High-Resolution Genome-Wide Assessment of Fitness Using Transposon-Insertion Sequencing. PLoS Genet 2014, 10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28. Hubbard TP, Billings G, Dörr T, Sit B, Warr AR, Kuehl CJ, Kim M, Delgado F, Mekalanos JJ, Lewnard JA, et al. : A live vaccine rapidly protects against cholera in an infant rabbit model. Sci Transl Med 2018, 10. * This study demonstrated pre-colonization of infant rabbits by attenuated V. cholerae could block subsequent infection.
  • 29.Mandlik A, Livny J, Robins WP, Ritchie JM, Mekalanos JJ, Waldor MK: RNA-Seq-based monitoring of infection-linked changes in Vibrio cholerae gene expression. Cell Host Microbe 2011, 10:165–74. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Abel S, Abel Zur Wiesch P, Chang H-H, Davis BM, Lipsitch M, Waldor MK: Sequence tag-based analysis of microbial population dynamics. Nat Methods 2015, 12:223–226. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31. Zoued A, Zhang H, Zhang T, Giorgio RT, Kuehl CJ, Fakoya B, Sit B, Waldor MK: Proteomic analysis of the pathogen-host interface in experimental cholera. Nat Chem Biol 2021, in press. ** This study profiled the host-V. cholerae surface interaction proteome during infant rabbit and infant mouse infection, revealing new host secreted factors that may bind and surveil intestinal microbes.
  • 32. Rivera-Chávez F, Mekalanos JJ: Cholera toxin promotes pathogen acquisition of host-derived nutrients. Nature 2019, 572:244–248.31367037 ** This study combined infant rabbit and infant mouse studies to reveal a novel role for CT in modulating V. cholerae metabolism during infection.
  • 33.Blow NS, Salomon RN, Garrity K, Reveillaud I, Kopin A, Jackson FR, Watnick PI: Vibrio cholerae infection of Drosophila melanogaster mimics the human disease cholera. PLoS Pathog 2005, 1:0092–0098. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Runft DL, Mitchell KC, Abuaita BH, Allen JP, Bajer S, Ginsburg K, Neely MN, Withey JH: Zebrafish as a natural host model for Vibrio cholerae colonization and transmission. Appl Environ Microbiol 2014, 80:1710–1717. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Vaitkevicius K, Lindmark B, Ou G, Song T, Toma C, Iwanaga M, Zhu J, Andersson A, Hammarström M-LL, Tuck S, et al. : A Vibrio cholerae protease needed for killing of Caenorhabditis elegans has a role in protection from natural predator grazing. Proc Natl Acad Sci U S A 2006, 103:9280–9285. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Hang S, Purdy AE, Robins WP, Wang Z, Mandal M, Chang S, Mekalanos JJ, Watnick PI: The acetate switch of an intestinal pathogen disrupts host insulin signaling and lipid metabolism. Cell Host Microbe 2014, 16:592–604. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Vanhove AS, Hang S, Vijayakumar V, Wong ACN, Asara JM, Watnick PI: Vibrio cholerae ensures function of host proteins required for virulence through consumption of luminal methionine sulfoxide. PLoS Pathog 2017, 13:1–26. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Davoodi S, Foley E: Host-Microbe-Pathogen Interactions: A Review of Vibrio cholerae Pathogenesis in Drosophila. Front Immunol 2020, 10:1–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39. Kamareddine L, Wong ACN, Vanhove AS, Hang S, Purdy AE, Kierek-Pearson K, Asara JM, Ali A, Morris JG, Watnick PI: Activation of Vibrio cholerae quorum sensing promotes survival of an arthropod host. Nat Microbiol 2018, 3:243–252.29180725 * This study used fruit fly infections to demonstrate the differential contributions of quorum sensing to V. cholerae pathogenesis.
  • 40.Dongre M, Singh B, Aung KM, Larsson P, Miftakhova R, Persson K, Askarian F, Johannessen M, von Hofsten J, Persson JL, et al. : Flagella-mediated secretion of a novel Vibrio cholerae cytotoxin affecting both vertebrate and invertebrate hosts. Commun Biol 2018, 1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Logan SL, Thomas J, Yan J, Baker RP, Shields DS, Xavier JB, Hammer BK, Parthasarathy R: The Vibrio cholerae type VI secretion system can modulate host intestinal mechanics to displace gut bacterial symbionts. Proc Natl Acad Sci U S A 2018, 115:E3779–E3787. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Breen P, Winters AD, Theis KR, Withey JH: The Vibrio cholerae T6SS is Dispensable for Colonization but Affects Pathogenesis and the Structure of Zebrafish Intestinal Microbiome. Infect Immun 2021, doi: 10.1128/IAI.00151-21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Breen P, Winters AD, Theis KR, Withey JH: Vibrio cholerae Infection Induces Strain Specific Modulation of the Zebrafish Intestinal Microbiome. Infect Immun 2021, doi: 10.1128/IAI.00157-21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Pukatzki S, Ma AT, Sturtevant D, Krastins B, Sarracino D, Nelson WC, Heidelberg JF, Mekalanos JJ: Identification of a conserved bacterial protein secretion system in Vibrio cholerae using the Dictyostelium host model system. Proc Natl Acad Sci U S A 2006, 103:1528–1533. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Fast D, Kostiuk B, Foley E, Pukatzki S: Commensal pathogen competition impacts host viability. Proc Natl Acad Sci U S A 2018, 115:7099–7104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46. Zhao W, Caro F, Robins W, Mekalanos JJ: Antagonism toward the intestinal microbiota and its effect on Vibrio cholerae virulence. Science 2018, 359:210–213.29326272 * This study used infant mice to investigate the role of the V. cholerae T6SS in mediating pathogen competition with the intestinal microbiota.
  • 47.Fu Y, Ho BT, Mekalanos JJ: Tracking Vibrio cholerae Cell-Cell Interactions during Infection Reveals Bacterial Population Dynamics within Intestinal Microenvironments. Cell Host Microbe 2018, doi: 10.1016/j.chom.2017.12.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Midani FS, Weil AA, Chowdhury F, Begum YA, Khan AI, Debela MD, Durand HK, Reese AT, Nimmagadda SN, Silverman JD, et al. : Human Gut Microbiota Predicts Susceptibility to Vibrio cholerae Infection. J Infect Dis 2018, 218:645–653. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Levade I, Saber MM, Midani FS, Chowdhury F, Khan AI, Begum YA, Ryan ET, David LA, Calderwood SB, Harris JB, et al. : Predicting Vibrio cholerae Infection and Disease Severity Using Metagenomics in a Prospective Cohort Study. J Infect Dis 2020, 223:342–351. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Hsiao A, Ahmed AMS, Subramanian S, Griffin NW, Drewry LL, Petri WA, Haque R, Ahmed T, Gordon JI: Members of the human gut microbiota involved in recovery from Vibrio cholerae infection. Nature 2014, 515:423–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Cho JY, Liu R, Macbeth JC, Hsiao A: The Interface of Vibrio cholerae and the Gut Microbiome. Gut Microbes 2021, 13:1937015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52. Alavi S, Mitchell JD, Cho JY, Liu R, Macbeth JC, Hsiao A: Interpersonal Gut Microbiome Variation Drives Susceptibility and Resistance to Cholera Infection. Cell 2020, 181:1533–1546.e13.32631492 ** This study used gnotobiotic adult and infant mice to demonstrate how variations in intestinal microbiota across humans could explain susceptibility to V. cholerae infection, via commensal-mediated regulation of V. cholerae virulence genes.
  • 53.Yen M, Cairns LS, Camilli A: A cocktail of three virulent bacteriophages prevents Vibrio cholerae infection in animal models. Nat Commun 2017, 8:1–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Harris JB: Cholera: Immunity and Prospects in Vaccine Development. J Infect Dis 2018, 218:S141–S146. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Sasaki S, Ohnishi N, Suzuki R, Adachi K, Miyashita M, Shimamura T, Tazume S, Maeda R, Takahashi T: The Relation between the Persistence of El Tor Vibrio in the Intestines of Germfree Mice and So-called Coproantibody. J Infect Dis 1970, 121:S124–S131. [DOI] [PubMed] [Google Scholar]
  • 56.Sit B, Zhang T, Fakoya B, Akter A, Biswas R, Ryan ET, Waldor MK: Oral immunization with a probiotic cholera vaccine induces broad protective immunity against Vibrio cholerae colonization and disease in mice. PLoS Negl Trop Dis 2019, 13:e0007417. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Wade WF, King RG, Grandjean C, Wade TK, Justement LB: Murine marginal zone B cells play a role in Vibrio cholerae LPS antibody responses. Pathog Dis 2014, 70:153–157. [DOI] [PubMed] [Google Scholar]
  • 58.Fakoya B, Sit B, Waldor MK: Transient Intestinal Colonization by a Live-Attenuated Oral Cholera Vaccine Induces Protective Immune Responses in Streptomycin-Treated Mice. J Bacteriol 2020, 202. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Lee EY, Lee S, Rho S, Kim J-O, Choi SK, Lee YJ, Park JY, Song M, Yang JS: Immunogenicity of a bivalent killed thimerosal-free oral cholera vaccine, Euvichol, in an animal model. Clin Exp Vaccine Res 2018, 7:104–110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Leitner DR, Feichter S, Schild-Prüfert K, Rechberger GN, Reidl J, Schild S: Lipopolysaccharide modifications of a cholera vaccine candidate based on outer membrane vesicles reduce endotoxicity and reveal the major protective antigen. Infect Immun 2013, 81:2379–2393. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Sayeed MA, Bufano MK, Xu P, Eckhoff G, Charles RC, Alam MM, Sultana T, Rashu MR, Berger A, Gonzalez-Escobedo G, et al. : A Cholera Conjugate Vaccine Containing O-specific Polysaccharide (OSP) of V. cholerae O1 Inaba and Recombinant Fragment of Tetanus Toxin Heavy Chain (OSP:rTTHc) Induces Serum, Memory and Lamina Proprial Responses against OSP and Is Protective in Mice. PLoS Negl Trop Dis 2015, 9:e0003881. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62. Charles RC, Kelly M, Tam JM, Akter A, Hossain M, Islam K, Biswas R, Kamruzzaman M, Chowdhury F, Khan AI, et al. : Humans Surviving Cholera Develop Antibodies against Vibrio cholerae O-Specific Polysaccharide That Inhibit Pathogen Motility. MBio 2020, 11:1–13. * This study used infant mice to demonstrate that monoclonal anti-OSP antibodies isolated from cholera patients could block V. cholerae motility in vivo.
  • 63.Wang Z, Lazinski DW, Camilli A: Immunity provided by an outer membrane vesicle cholera vaccine is due to O-antigenspecific antibodies inhibiting bacterial motility. Infect Immun 2017, 85:1–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Baranova DE, Chen L, Destrempes M, Meade H, Mantis NJ: Passive Immunity to Vibrio cholerae O1 Afforded by a Human Monoclonal IgA1 Antibody Expressed in Milk. Pathog Immun 2020, 5:89–116. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Chac D, Bhuiyan TR, Saha A, Alam MM, Salma U, Jahan N, Chowdhury F, Khan AI, Ryan ET, LaRocque R, et al. : The gut microbiota and development of Vibrio cholerae-specific long-term memory B cells in adults after whole-cell killed oral cholera vaccine. Infect Immun 2021, doi: 10.1128/IAI.00217-21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66. Di Luccia B, Ahern PP, Griffin NW, Cheng J, Guruge JL, Byrne AE, Rodionov DA, Leyn SA, Osterman AL, Ahmed T, et al. : Combined Prebiotic and Microbial Intervention Improves Oral Cholera Vaccination Responses in a Mouse Model of Childhood Undernutrition. Cell Host Microbe 2020, 27:899–908.e5.32348782 ** This study used gnotobiotic adult mice to test how nutritional modulation of the gut microbiota influenced immune responses to CT-based vaccination.
  • 67.Metchnikoff E: Recherches sur le cholera et les vibrions. VI. Receptivite des jeunes lapins pour le cholera intestinal. Ann Inst Pasteur (Paris) 1894, 8:557–568. [Google Scholar]
  • 68.LaRocque RC, Sabeti P, Duggal P, Chowdhury F, Khan AI, Lebrun LM, Harris JB, Ryan ET, Qadri F, Calderwood SB: A variant in long palate, lung and nasal epithelium clone 1 is associated with cholera in a Bangladeshi population. Genes Immun 2009, 10:267–272. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Karlsson EK, Harris JB, Tabrizi S, Rahman A, Shlyakhter I, Patterson N, O’Dushlaine C, Schaffner SF, Gupta S, Chowdhury F, et al. : Natural Selection in a Bangladeshi Population from the Cholera-Endemic Ganges River Delta. Sci Transl Med 2013, 5:192ra86. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Sasabe J, Miyoshi Y, Rakoff-Nahoum S, Zhang T, Mita M, Davis BM, Hamase K, Waldor MK, Fujii N, Saito T, et al. : Interplay between microbial d-amino acids and host d-amino acid oxidase modifies murine mucosal defence and gut microbiota. Nat Microbiol 2016, 1:16125. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Hsiao AAA, Liu Z, Joelsson A, Zhu J: Vibrio cholerae virulence regulator-coordinated evasion of host immunity. Proc Natl Acad Sci U S A 2006, 103:14542–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Alisson-Silva F, Liu JZ, Diaz SL, Deng L, Gareau MG, Marchelletta R, Chen X, Nizet V, Varki N, Barrett KE, et al. : Human evolutionary loss of epithelial Neu5Gc expression and species-specific susceptibility to cholera. PLoS Pathog 2018, 14:1–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Merrell DS, Butler SM, Qadri F, Dolganov NA, Alam A, Cohen MB, Calderwood SB, Schoolnik GK, Camilli A: Host-induced epidemic spread of the cholera bacterium. Nature 2002, 417:642–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Alam A, Larocque RC, Harris JB, Vanderspurt C, Ryan ET, Qadri F, Calderwood SB: Hyperinfectivity of human-passaged Vibrio cholerae can be modeled by growth in the infant mouse. Infect Immun 2005, 73:6674–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Hullahalli K, Pritchard JR, Waldor MK: Refined Quantification of Infection Bottlenecks and Pathogen Dissemination with STAMPR. mSystems 2021, 6:e0088721. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES