Review
Animal models to study the pathogenesis of human and animal Clostridium perfringens infections

https://doi.org/10.1016/j.vetmic.2015.02.013Get rights and content

Highlights

  • Gas gangrene in humans was mainly elucidated using a mouse model coupled with genetic studies.

  • A chicken model was used to understand type A-mediated necrotic enteritis in poultry.

  • Food poisoning and necrotic enteritis were studied using mainly rabbits and mice.

  • C. perfringens type D infection has been studied using models in mice, rats, sheep, goats and cattle.

  • Molecular Koch's postulates have been fulfilled using animal models for most of these diseases.

Abstract

The most common animal models used to study Clostridium perfringens infections in humans and animals are reviewed here. The classical C. perfringens-mediated histotoxic disease of humans is clostridial myonecrosis or gas gangrene and the use of a mouse myonecrosis model coupled with genetic studies has contributed greatly to our understanding of disease pathogenesis. Similarly, the use of a chicken model has enhanced our understanding of type A-mediated necrotic enteritis in poultry and has led to the identification of NetB as the primary toxin involved in disease. C. perfringens type A food poisoning is a highly prevalent bacterial illness in the USA and elsewhere. Rabbits and mice are the species most commonly used to study the action of enterotoxin, the causative toxin. Other animal models used to study the effect of this toxin are rats, non-human primates, sheep and cattle. In rabbits and mice, CPE produces severe necrosis of the small intestinal epithelium along with fluid accumulation. C. perfringens type D infection has been studied by inoculating epsilon toxin (ETX) intravenously into mice, rats, sheep, goats and cattle, and by intraduodenal inoculation of whole cultures of this microorganism in mice, sheep, goats and cattle. Molecular Koch's postulates have been fulfilled for enterotoxigenic C. perfringens type A in rabbits and mice, for C. perfringens type A necrotic enteritis and gas gangrene in chickens and mice, respectively, for C. perfringens type C in mice, rabbits and goats, and for C. perfringens type D in mice, sheep and goats.

Introduction

Clostridium perfringens, an anaerobic, spore-forming Gram-positive rod, can produce ∼17 toxins (McClane et al., 2006, Li et al., 2013). Four of these toxins, alpha (CPA), beta (CPB), epsilon (ETX), and iota (ITX) are used to classify this microorganism into five toxinotypes A, B, C, D and E. Most C. perfringens isolates produce, in addition to at least one of the typing toxins, other toxins including, but not limited to, enterotoxin (CPE), beta2 toxin (CPB2), NetB and TpeL (McClane et al., 2006, Li et al., 2013).

The different toxinotypes of C. perfringens produce a wide variety of diseases in both humans and animals, ranging from C. perfringens type A gas gangrene to several enterotoxemias and enteritis syndromes. All of these diseases are mediated by one or more toxins of C. perfringens (Uzal et al., 2014).

Several animal models have been used to study the role of the different toxins of C. perfringens in the pathogenesis of the infections produced by this microorganism (McDonel, 1980, Sayeed et al., 2008, Garcia et al., 2013, Li et al., 2013, Uzal et al., 2014). In particular, over the past few years, some of these animal models have been used to fulfill molecular Koch's postulates for various diseases (Awad et al., 1995, Sarker et al., 1999, McClane et al., 2006, Keyburn et al., 2008, Sayeed et al., 2008, Garcia et al., 2013). We review here the information published on the main animal models used to study the pathogenesis of C. perfringens infections, with special emphasis on those used to fulfill molecular Koch's postulates.

Section snippets

Gas gangrene

Gas gangrene, or clostridial myonecrosis, is an invasive, anaerobic infection of muscle and is characterized by extensive tissue necrosis and the production of gas (MacLennan, 1962). In humans, this infection can be divided into two types: spontaneous gangrene and traumatic gangrene. The former type is commonly caused by Clostridium septicum, while approximately 80% of the latter is caused by C. perfringens. However, other clostridia have also been associated with clostridial myonecrosis (

C. perfringens type C

C. perfringens type C isolates must produce, at the minimum, CPA and CPB (McClane et al., 2006). These strains are responsible for highly lethal enteric diseases and enterotoxemias in humans (enteritis necroticans) and in many other mammalian species. Type C disease is mainly characterized by necrotizing enteritis or enterocolitis and systemic disease. It is currently accepted that most clinical manifestations and lesions observed in patients with type C disease are a direct consequence of the

C. perfringens type D

C. perfringens type D is responsible for a highly lethal enterotoxemia in sheep, goats and other ruminants. Type D isolates produce CPA and ETX, but several toxinotype D isolates also produce several other toxins (McClane et al., 2006).

ETX is produced in the form of a relatively inactive prototoxin, which becomes fully activated when a string of 14 amino acids from the C-terminus are proteolytically removed (Minami et al., 1997). Activation of ETX in the host intestine is mediated by serine

Concluding remarks

Although significant progress has been made over the last few decades toward the understanding of C. perfringens infections in humans and animals, it was not until relatively recently that the development of reverse genetics, combined with the use of several animal models, allowed researchers to determine the importance of individual toxins in the pathogenesis and virulence of different toxinotypes and strains of this microorganism. In addition, although several animal models have been

Conflict of interest

None.

Acknowledgements

This work was supported by Public Health Service grants R37 AI019844 and AI056177 from the National Institute of Allergy and Infectious Diseases. Grant CE0562063 from the Australian Research Council, Project Grant GNT10695985 from the Australian National Health and Medical Research Council and Grant 1.1.2 from the Poultry Cooperative Research Centre (Australia). We thank Ms. S. Fitisemanu for excellent secretarial help.

References (125)

  • J.P. Garcia et al.

    The effect of Clostridium perfringens type C strain CN3685 and its isogenic beta toxin null mutant in goats

    Vet. Microbiol.

    (2012)
  • J. Katahira et al.

    Clostridium perfringens enterotoxin utilizes two structurally related membrane proteins as functional receptors in vivo

    J. Biol. Chem.

    (1997)
  • J.L. McReynolds et al.

    Evaluation of immunosuppressants and dietary mechanisms in an experimental disease model for necrotic entertitis

    Poult. Sci.

    (2004)
  • J. Miclard et al.

    Clostridium perfringens beta-toxin targets endothelial cells in necrotizing enteritis in piglets

    Vet. Microbiol.

    (2009)
  • M. Nagahama et al.

    Distribution of labeled Clostridium perfringens epsilon toxin in mice

    Toxicon

    (1991)
  • L. Niilo

    Effect on calves of the intravenous injection of the enterotoxin of Clostridium welchii type A

    J. Comp. Pathol.

    (1973)
  • U. Singh et al.

    CaCo-2 cells treated with Clostridium perfringens enterotoxin form multiple large complex species, one of which contains the tight junction protein occluding

    J. Biol. Chem.

    (2000)
  • U. Singh et al.

    Comparative biochemical and immunochemical studies reveal differences in the effects of Clostridium perfringens enterotoxin on polarized CaCo-2 cells versus Vero cells

    J. Biol. Chem.

    (2001)
  • J.A. Smyth et al.

    Disease producing capability of netB positive isolates of C. perfringens recovered from normal chickens and a cow, and netB positive and negative isolates from chickens with necrotic entertitis

    Vet. Microbiol.

    (2010)
  • A. Soler-Jover et al.

    Distribution of Clostridium perfringens epsilon toxin in the brains of acutely intoxicated mice and its effect upon glial cells

    Toxicon

    (2007)
  • W.A. Altemeier et al.

    Toxoid immunization in experimental gas Gangrene – a preliminary report

    Ann. Surg.

    (1947)
  • C.B. Annett et al.

    Necrotic enteritis: effect of barley, wheat and corn diets on proliferation of Clostridium perfringens type A

    Avian Pathol.

    (2002)
  • A.R. Armstrong et al.

    Chemotherapy and experimental gas gangrene

    Can. Med. Assoc. J.

    (1941)
  • D. Autheman et al.

    Clostridium perfringens beta-toxin induces necrostatin-inhibitable, calpain-dependent necrosis in primary porcine endothelial cells

    PLOS ONE

    (2013)
  • M.M. Awad et al.

    Virulence studies on chromosomal a-toxin and q-toxin mutants constructed by allelic exchange provide genetic evidence for the essential role of a-toxin in Clostridium perfringens-mediated gas gangrene

    Mol. Microbiol.

    (1995)
  • M.M. Awad et al.

    Synergistic effects of alpha-toxin and perfringolysin O in Clostridium perfringens-mediated gas gangrene

    Infect. Immun.

    (2001)
  • T.L. Bannam et al.

    Necrotic enteritis-derived Clostridium perfringens with three closely related three independently conjugative toxin and antibiotic resistance plasmids

    mBio

    (2011)
  • T.E. Blackwell et al.

    Differences in signs and lesions in sheep and goats with enterotoxemia induced by intraduodenal infusion of Clostridium perfringens type D

    Am. J. Vet. Res.

    (1991)
  • J. Bos et al.

    Fatal necrotizing colitis following a foodborne outbreak of enterotoxigenic Clostridium perfringens type A infection

    Clin. Infect. Dis.

    (2005)
  • A.E. Bryant et al.

    Clostridial myonecrosis: new insights in pathogenesis and management

    Curr. Infect. Dis. Rep.

    (2010)
  • J.A. Caserta et al.

    Development and application of a mouse intestinal loop model to study the in vivo action of Clostridium perfringens enterotoxin

    Infect. Immun.

    (2011)
  • G. Chakrabarti et al.

    Death pathways activated in CaCo-2 cells by Clostridium perfringens enterotoxin

    Infect. Immun.

    (2003)
  • G. Chakrabarti et al.

    The importance of calcium influx, calpain, and calmodulin for the activation of CaCo-2 cell death pathways by Clostridium perfringens enterotoxin

    Cell. Microbiol.

    (2005)
  • A. Chakravorty et al.

    The cysteine protease alpha-clostripain is not essential for the pathogenesis of Clostridium perfringens-mediated myonecrosis

    PLoS ONE

    (2011)
  • A. Chakravorty et al.

    Opioid analgesics stop the development of clostridial gas gangrene

    J. Infect. Dis.

    (2014)
  • E. Chen et al.

    Management of gas gangrene in Wenchuan earthquake victims

    J. Huazhong Univ. Sci. Technol.

    (2011)
  • M. Chiarezza et al.

    The NanI and NanJ sialidases of Clostridium perfringens are not essential for virulence

    Infect. Immun.

    (2009)
  • J.R. Czeczulin et al.

    Cloning, nucleotide sequencing, and expression of the Clostridium perfringens enterotoxin gene in Escherichia coli

    Infect. Immun.

    (1993)
  • S.S. Diab et al.

    Pathology of Clostridium perfringens type C enterotoxemia in horses

    Vet. Pathol.

    (2012)
  • J. Dorca-Arévalo et al.

    Correlation between in vitro cytotoxicity and in vivo lethal activity in mice of epsilon toxin mutants from Clostridium perfringens

    PLOS ONE

    (2014)
  • C.L. Duncan et al.

    Ileal loop fluid accumulation and production of diarrhea in rabbits by cell-free products of Clostridium perfringens

    J. Bacteriol.

    (1969)
  • D.M. Ellemor et al.

    Use of genetically manipulated strains of Clostridium perfringens reveals both alpha-toxin and theta-toxin are required for vascular leukostasis to occur in experimental gas gangrene

    Infect. Immun.

    (1999)
  • D.G. Evans

    The protective properties of the alpha antitoxin and antihyaluronidase occurring in C. welchii type A antiserum

    J. Pathol. Bacteriol.

    (1943)
  • J.M. Farrant et al.

    Pigbel-like syndrome in a vegetarian in Oxford

    Gut

    (1996)
  • H.I. Field et al.

    The experimental reproduction of enterotoxaemia in piglets

    J. Hyg.

    (1959)
  • E.J. Filho et al.

    Clinicopathologic features of experimental Clostridium perfringens type D enterotoxemia in cattle

    Vet. Pathol.

    (2009)
  • J.W. Finnie

    Pathogenesis of brain damage produced in sheep by Clostridium perfringens type D epsilon toxin: a review

    Aust. Vet. J.

    (2003)
  • J.W. Finnie et al.

    Aquaporin-4 in acute cerebral edema produced by Clostridium perfringens type D epsilon toxin

    Vet. Pathol.

    (2008)
  • D.J. Fisher et al.

    Dissecting the contributions of Clostridium perfringens type C toxins to lethality in the mouse intravenous injection model

    Infect. Immun.

    (2006)
  • J.C. Freedman et al.

    Proteolytic processing and activation of Clostridium perfringens epsilon toxin by caprine small intestinal contents

    mBio

    (2014)
  • Cited by (0)

    View full text