Next Article in Journal
Advances in Research on Bacterial Oxidation of Mn(II): A Visualized Bibliometric Analysis Based on CiteSpace
Previous Article in Journal
The Effects of Aspirin Intervention on Inflammation-Associated Lingual Bacteria: A Pilot Study from a Randomized Clinical Trial
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Clostridium perfringens in the Intestine: Innocent Bystander or Serious Threat?

1
State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730000, China
2
Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou 730046, China
3
State Key Laboratory of Grassland Agro-Ecosystems, College of Pastoral Agriculture Science and Technology, Lanzhou University, Lanzhou 730020, China
*
Author to whom correspondence should be addressed.
Microorganisms 2024, 12(8), 1610; https://doi.org/10.3390/microorganisms12081610
Submission received: 2 July 2024 / Revised: 31 July 2024 / Accepted: 2 August 2024 / Published: 7 August 2024
(This article belongs to the Section Public Health Microbiology)

Abstract

:
The Clostridium perfringens epidemic threatens biosecurity and causes significant economic losses. C. perfringens infections are linked to more than one hundred million cases of food poisoning annually, and 8–60% of susceptible animals are vulnerable to infection, resulting in an economic loss of more than 6 hundred million USD. The enzymes and toxins (>20 species) produced by C. perfringens play a role in intestinal colonization, immunological evasion, intestinal micro-ecosystem imbalance, and intestinal mucosal disruption, all influencing host health. In recent decades, there has been an increase in drug resistance in C. perfringens due to antibiotic misuse and bacterial evolution. At the same time, traditional control interventions have proven ineffective, highlighting the urgent need to develop and implement new strategies and approaches to improve intervention targeting. Therefore, an in-depth understanding of the spatial and temporal evolutionary characteristics, transmission routes, colonization dynamics, and pathogenic mechanisms of C. perfringens will aid in the development of optimal therapeutic strategies and vaccines for C. perfringens management. Here, we review the global epidemiology of C. perfringens, as well as the molecular features and roles of various virulence factors in C. perfringens pathogenicity. In addition, we emphasize measures to prevent and control this zoonotic disease to reduce the transmission and infection of C. perfringens.

1. Introduction

Clostridium perfringens, a Gram-positive bacillus, is ubiquitous in the environment as well as in the normal intestinal flora of humans and animals. C. perfringens, was first isolated and identified by William H. Welch in 1892 by necropsy (Figure 1), and is an intestinal conditionally pathogenic organism that becomes pathogenic when immunity is compromised, is an enteric conditionally pathogenic bacterium that becomes pathogenic when immunity is compromised. C. perfringens, with its rapid doubling speed and efficient resource utilization, coupled with its potent toxin production and environmental adaptability, can quickly colonize hosts and cause severe infections. These characteristics pose a serious threat to human health and global livestock (8–60% of susceptible animals are vulnerable to infection). Avian necrotizing enterocolitis causes a staggering global economic loss approximated at around 6 hundred million USD annually [1]. Antibiotic-associated diarrhea has been associated with a variety of bacterial infection factors, including C. perfringens, Clostridium difficile, Staphylococcus aureus, Candida albicans, and Klebsiella oxytoca [2]. Both C. perfringens and Clostridioides difficile can cause intestinal dysbiosis after antibiotic use. After the use of antibiotics such as β-lactams, β-lactamase inhibitors, and cephalosporins, C. perfringens produces enterotoxins, that cause antibiotic-associated diarrhea, in contrast, the use of antibiotics like fluoroquinolones, cephalosporins, and clindamycin, leads to Clostridioides difficile releasing toxins A and B, which also result in antibiotic-associated diarrhea [3]. New evidence suggests that antibiotic-associated diarrhea occurs in 5% to 30% of patients receiving antibiotics, with C. perfringens type A causing up to 15% of AAD cases and type F associated with 2% to 15% of AAD cases [3,4,5].
C. perfringens is classified into A–G strains and is known to cause various diseases in humans and animals, including gas gangrene, necrotizing enterocolitis, multiple sclerosis, antibiotic-associated diarrhea, and others (Figure 2). These pathogenic effects are linked to the production of exotoxins (e.g., α-toxin, β-toxin, enterotoxin, ε-toxin, ι-toxin, etc.) and enzymes (e.g., salivary lyase, phospholipase, hexosaminidase, among others) (Figure 2). Notably, C. perfringens infections cause a significant morbidity and lethality rate [6,7,8,9]. C. perfringens colonizes the intestinal tract, alters the intestinal environment, disrupts the intestinal microecological balance, and causes an inflammatory response, and its toxins attack multiple target organs, including the intestinal tract, kidneys, lungs, and heart, further damaging organs such as the kidneys, lungs, and heart [10]. In addition, C. perfingens salivary lyase, by promoting the adhesion and colonization of C. perfringens in the intestinal tract and the combination of toxins and intestinal cells, provides nutrition for C. perfringens, destroying the intestinal microecological balance and host immune system [11].
It is reasonable to assume that the primary treatment of C. perfringens disease is the precise elimination of C. perfringens colonization of the intestinal tract, with antibiotics now serving as the cornerstone. However, studies have demonstrated that prolonged antibiotic usage may disrupt the intestinal flora, decrease intestinal short-chain fatty acid concentrations, alter carbohydrate and bile acid uptake, and increase the pathogenicity of C. perfringens [12,13]. In addition, when the use of ionophore for the prevention and treatment of coccidiosis is discontinued, the bacterial flora in the intestinal tract of poultry may become unbalanced, leading to a rapid proliferation of C. perfringens, which can trigger dysbiosis. Meanwhile, C. perfringens acquire antibiotic resistance over time through genetic mutation, horizontal gene transfer, genetic resistance loading, and natural selection. Nearly 1.27 million individuals worldwide succumbed to antibiotic resistance in 2019, and global GDP and trade losses due to antibiotic resistance are projected to be as high as $8.5 billion and $2.3 billion, respectively, by 2025 [14,15]. Therefore, the solution to C. perfringens disease lies in the production of organic acids, enzymes, prebiotic oligosaccharides, and microecological agents at the multidisciplinary intersection of environment, chemistry, and medicine (Figure 2 and Figure 3).
Overall, this work examines the spatial and temporal evolutionary features of C. perfringens, as well as the pathogenic mechanisms of enzymes and major toxins produced by C. perfringens. Furthermore, we critically review the therapeutic perspectives of antibiotics, vaccines, and other biologics against C. perfringens to provide the most up-to-date research-based solutions to researchers, physicians, students, and the general.

1.1. C. perfringens Threatens Global Biological Public Safety

1.1.1. Humans

Foodborne illness, bacteremia, and antibiotic-associated diarrhea (antibiotic-associated diarrhea, AAD) caused by C. perfringens are widespread worldwide, including the United Kingdom, the United States, Canada, and China, where there is a high prevalence of infection and cross-contamination through vectors such as raw meat, raw milk, and spices. Emerging evidence indicates that C. perfringens cause roughly 13% of foodborne food poisoning, 5–20% of AAD, and a portion of non-foodborne diarrhea [12,16] (Table 1). C. perfringens-related foodborne infections were predominantly reported in the Eastern region of the UK (2012–2014). These infections were particularly prevalent in communal settings, such as nursing homes. Alarmingly, this resulted in approximately 25 fatalities per year from C. perfringens [17,18]. Food poisoning caused by C. perfringens infection affects roughly 1 million individuals in the United States, about 900,000 people in England and approximately 16,000 people in Canada (2013) each year [19,20,21] (Table 1). Studies have shown that C. difficile is a major cause of ADD, and the incidence of C. difficile AAD is approximately four times higher than that of C. perfringens AAD [22]. In Iran, approximately 13.3% of patients diagnosed with antibiotic-associated diarrhea tested positive for C. perfringens infection, with type A being the most prevalent (Table 1). The prevalent type in central China (2018–2019) was F, accounting for 49.5% of cases [23,24]. C. perfringens is transmitted and cross-infected by various carriers, including raw meat and spices. For instance, the prevalence of C. perfringens in raw poultry products sold in different countries, including the United States, Japan, India, and Canada, varies significantly between samples, ranging from 6% to 97% [25]. The examination of raw meat samples in a slaughterhouse region in Shaanxi, China (2018–2019) revealed that 21.2% were isolated as type A C. perfringens (with 80.8% prevalence), and 19.2% as type D C. perfringen [26]. The detection rate in Kazakhstan was 28%, with a predominance of type A [27]. In China, cross-infection of C. perfringens during rearing and milking on dairy farms is present, with a prevalence of 22.3% [28]. C. perfringens spores can be found in spices in the United States, making them a vector for foodborne transmission of C. perfringens [29].

1.1.2. Animal

C. perfringens may infect horses, sheep, goats, cattle, pigs, chickens, and other animals through their digestive tracts. C. perfringens type A is the most common, but types B, C, D, E, F, and G are also present in small amounts [31,32,33,34]. The prevalence of C. perfringens was roughly 8% of healthy horses in Switzerland (2013–2016) [35] (Table 2). The prevalence of enterotoxemia in sheep and goats was 45.2% in Samsun governorate, Turkey (2014–2015) [36]. The prevalence of C. perfringens was 17.5% lambs in Kalubiya and Menofia governorates, Egypt (2021–2022) [37] (Table 2). The prevalence of C. perfringens in India (2000–2021) was detected among different animal species as follows: pigs (60%) > sheep (56.3%) > goats (38.7%) > cattle (35%) [38] (Table 2). C. perfringens was detected in poultry farms in Weifang, Guangdong, Taian, and Pingyin at a remarkably high rate of 38.42% [36]. The isolation rate of C. perfringens in sheep in Gansu (2019–2020) was 14.7% (Table 2). Furthermore, in commercial farms in Anhui, Guangdong, Guangxi, and Fujian provinces (2020), the isolation rate of C. perfringens was notably higher, reaching 44.173% [39,40] (Table 2).

1.1.3. Economy

C. perfringens, a globally widespread bacterium, can induce various illnesses with significant global economic consequences. C. perfringens-related food poisoning costs the United States economy roughly $ thirty billion, with type A accounting for approximately $500 million and type F accounting for approximately $310 million [41,42,43]. In the United Kingdom, the cost of treating necrotizing small bowel colitis in preterm infants caused by C. perfringens is approximately £130,000 per year [44]. Avian necrotizing enterocolitis causes a global economic loss of nearly $600 million each year [45].

2. Virulence Mechanisms

The pathogenic mechanism of C. perfringens includes intestinal colonization and proliferation, immune escape, toxin production, imbalance in the intestinal micro-ecosystem, disruption of the intestinal mucosa, and the formation of gas-producing pods, adversely impacting the host organism. Overall, the main pathogenic factors are multi-enzymes and toxins (α, β, ε, ι and others).

2.1. Enzymes

The intestinal mucus layer serves as a crucial physiological structure on the intestinal mucosa, acting as the primary line of defense for natural immunity. This barrier primarily comprises highly glycosylated mucins, which trap microorganisms, particulate matter, and other harmful substances, effectively protecting the intestinal mucosa from direct aggression. C. perfringens, an intestinal microorganism, can produce salivary lyase (NanH, NanI and NanJ, etc.), hexosaminidase, galactosidase, fucoidan, phospholipase, and other enzymes [46]. Different C. perfringens strains secrete different salivary lyase enzymes that remove terminal salivary acids from the intestinal mucus layer, resulting in mucin degradation [46]. In addition, C. perfringens potentially utilizes salivary acid as a carbon source. This metabolic trait facilitates the colonization of the intestinal tract by C. perfringens and enhances the toxic effects of its toxins on host cells [11,47].

2.2. Toxin

2.2.1. Alpha Toxin

C. perfringens alpha toxin (CPA) is a member of the phospholipase C family. The CPA has two structural domains: an N domain with nine tightly stacked α-helices and a C domain with eight antiparallel β-intercalated structures [48]. All types of C. perfringens secrete CPA. CPA inhibits granulopoiesis and erythroid differentiation while increasing vascular permeability and hemolysis [49,50]. CPA creates an anaerobic environment for C. perfringens growth by activating phosphatidylinositol turnover and thromboxane A2 synthesis, causing aortic constriction and reduced blood supply to the tissues [51,52]. Moreover, CPA inhibits erythropoiesis by blocking erythroid lineage differentiation, causing erythrocyte lysis and hemolysis, thereby providing an anaerobic environment for C. perfringens growth [53]. Furthermore, CPA activates the c-Jun N-terminal kinase (JNK) signaling pathway (Figure 4A). This stimulation increases the secretion of colony-stimulating factor (G-CSF). However, it concurrently suppresses the expression of colony-stimulating factor receptor (G-CSFR) (Figure 4A). This dual effect influences G-CSF-mediated granulocyte production and release, impacting the immune response and the ability of the body to fight infection [54]. More, CPA binds to the ganglioside receptor GM1a and is able to promote the degradation of sphingolipids and phosphatidylcholine in the cytoplasmic membrane, causing a pro-inflammatory response and exacerbating tissue infection aggravation [21].

2.2.2. Beta Toxin

Unlike CPA, C. perfringens beta toxin (CPB) is a pore-forming toxin with a capsid, rim, and stem domain and produced by C. perfringens types B and C (Figure 4B) [55]. The CPB toxin disrupts the integrity of cell membranes by altering intracellular backbone proteins, signaling, and triggering inflammatory responses, among other pathways that lead to damage and inflammation in intestinal epithelial cells. Notably, endothelial cells and platelets are the target cells for CPB, and the CD31 protein, expressed explicitly on endothelial cells and platelets, functions as a CPB receptor (Figure 4A,B) [56,57]. CD31-deficient C57BL/6 mice demonstrated resistance to CPB [58]. CPB interaction with endothelial cells causes intestinal vascular necrosis and massive hemorrhage in the lamina propria and submucosa, primarily as small intestinal epithelial hemorrhage and necrosis [59]. CPB inhibits primary hemostasis by binding to the platelet plasma membrane, inhibits platelet aggregation, and impairs the ability of platelets to be activated by thrombin. (Figure 4A) [60]. Furthermore, the ability of CPB to bind to endothelial cells causes endothelial cell damage, increased vascular permeability and inflammatory response, localized tissue damage, as well as the exposure of subendothelial tissues and the initiation of endogenous coagulation, resulting in sustained bleeding from damaged vessels.

2.2.3. Epsilon Toxin

Epsilon toxin (ETX) is a seven-pore toxin produced by C. perfringens type B that causes cell membrane permeability, resulting in cell lysis and death (Figure 4A). Proteolytic enzymes convert the initially less active 32.9 kDa prototoxin (P-ETX) to the 27 kDa active toxin within the intestine [61,62,63]. The ETX specifically targets lymphocytes, blood cells, and oligodendrocytes. ETX preferentially binds to lymphocytes with high levels of myelin and lymphocyte protein (MAL) and causes cell death, with the degree of this binding determined by the duration and concentration of ETX exposure [64,65]. ETX-mediated hemolysis in human erythrocytes is intricately linked to MAL and P2 receptors, specifically P2Y13 and P2X7. Erythrocytes contract in response to ETX, which exposes phosphatidylserine to the erythrocyte membrane. This process is accompanied by increased erythrocyte sphingolipid content, increased intracellular concentrations of calcium, sodium, and chloride ions, and a decrease in potassium ion concentrations. The ensuing intra- and extracellular ionic imbalance promotes hemolysis [66,67,68].

2.2.4. Iota Toxin

Iota toxin (CPI) is a bacterial toxin consisting of two components: Ia and Ib produced by C. perfringens type E. (Figure 4B) [69]. Ia is an ADP-ribosylase that catalyzes ADP-ribosylation within the cell, disrupting cellular protein synthesis and energy metabolism. This process impairs cellular protein synthesis and energy metabolism. On the other hand, Ib, a membrane-associated component, binds to the cell membrane, causing intracellular sphingolipid accumulation. This disrupts the integrity and function of the cell membrane. The synergistic action of these two molecules is required to initiate the toxic effects [70]. The lipolysis-stimulated lipoprotein receptor (LSR) is the cellular receptor for Ib. Ib binds to amino acids 10–15 at the N-terminus of the LSR to generate the heptameric Ib, which subsequently binds to Ia (Figure 4B). As a result, the heptameric Ib binds to Ia, consequently increasing the inward flow of calcium ions and triggering the release of lysosomal proteases such as acid sphingomyelinase and histolytic protease. Acid sphingomyelinase hydrolyzes sphingolipids from the cell membrane, converting them into choline phosphate and ceramides, thereby promoting rapid endocytosis of CPIs [69,71,72].

2.2.5. Enterotoxins and Necrotizing Enterocolitis B-Like Toxins

C. perfringens enterotoxin (CPE) consists primarily of an NH2-terminal region and a C-terminal structural domain (cCPE), which is formed only during spore production and is produced by C. perfringens F [73]. The toxin interacts with claudin receptors (such as Claudin-4) on the surface of intestinal epithelial cells. The binding of the c-CPE region of CPE to the receptor on the surface of intestinal epithelial cells induces dissociation of the tight junctions between cells. This process widens the gaps between the layers of epithelial cells, increasing the permeability of the epithelial cell barrier. As a result, molecules can readily flow through the cell gaps, compromising the integrity of the epithelial cell barrier [74,75,76,77]. The N-terminal structural domains reassemble, forming cation-selective β-barrel pores. These pores allow ions such as Ca2+ and small molecules to traverse the cell membrane. This process impairs cell membrane integrity, resulting in damage and, ultimately, death of intestinal epithelial cells [78,79,80].
Necrotizing Enterocolitis B-like toxin (NetB) is a 33 kDa pore-forming toxin. It interacts with cholesterol in cell membranes on the surface of susceptible cells, leading to the formation of pores of approximately 1.6–1.8 nm in diameter. The formation of these pores disrupts the cell membrane, causing an increased influx of calcium ions into the cell. This imbalance in intra- and extracellular ion homeostasis subsequently activates the apoptotic pathway in the host cell (Figure 4B) [81,82,83]. The precise pathogenic mechanism of the NetB toxin is currently being investigated.

3. Prevention and Treatment of C. perfringens

3.1. Vaccines

Recombinant vaccines for C. perfringens are used in animals, including pigs, cattle, sheep, goats, and horses. Additionally, oral bait pellet vaccines expressing toxins CPA, ETX, CPB1, and CPB2 have been developed using recombinant Lactobacillus lactis [84,85]. In countries such as the United Kingdom, Canada and China, appropriate vaccines have been developed and relevant property rights registered for the prevention of C. perfringens (Table 3). Most C. perfringens vaccines available on the market today are inactivated, subunit, and live attenuated. However, these vaccines have significant drawbacks. These include the need for multiple injections, specificity to particular toxins, and the lack of vaccines targeting enteric diseases [86]. To address these challenges, the integration of bioinformatics analysis, proteomics, and vaccinology can be employed to design more effective multi-epitope vaccines for the future development of C. perfringens vaccines [87,88]. Moreover, phage display technology can be used to produce phage display vaccines on a large scale within a short period and at relatively low cost. It can also facilitate the development of more efficient adjuvants. A path towards a more comprehensive, safer, and effective C. perfringens vaccine may necessitate the investigation of diverse vaccine routes, fostering interdisciplinary collaborations.

3.2. Antibiotics

Since the discovery of penicillin by Fleming in 1928, antibiotics, including azithromycin, ampicillin, penicillin, florfenicol, and metronidazole, have been widely employed for treating C. perfringens infections. However, the antibiotic resistance genes of C. perfringens have undergone horizontal transfer (transformation, transduction, conjugation) and vertical transmission in natural environments, C. perfringens is now resistant to ampicillin, streptomycin, tetracycline, penicillin, antimicrobial peptides, and other antibiotics [89,90]. Studies have shown that the transfer of clostridial plasmids (Tcp) plasmid in C. perfringens encodes antibiotic resistance genes, including chloramphenicol, clindamycin, erythromycin, bacitracin and lincomycin [21]. In addition, the tetracycline resistance genes, tetA(P) and tetB(P), encode proteins that mediate the mechanisms of active tetracycline efflux and ribosome-protected tetracycline resistance, respectively [91]. Target site modification, efflux pumps and drug inactivation confer resistance to macrolides. The erm gene of C. perfringens encodes a 23S rRNA methylase that mediates target site modification and confers resistance to macrolides. The highest antibiotic resistance rate of C. perfringens isolated from chicken and pork sold in Vietnam was tetracycline (91.30%), followed by clindamycin (43.48%) [92]. Whereas in China, C. perfringens resistance to clindamycin was as high as 49.66% in samples isolated from human sources [16]. Antibiotic resistance is a significant global health concern, accounting for approximately1.27 million (2019) deaths. The cumulative economic cost of antibiotic resistance is estimated at $a trillion, and the number of deaths is projected to increase to 20.3 million by 2024 [15,93]. Hence, given the resistance of C. perfringens to multiple antibiotics, there is a critical need for research and development into alternative therapeutic strategies. Exploring avenues such as microecological agents, herbal products, phages, and other innovative approaches has become imperative. Furthermore, by diversifying therapeutic modalities, there is a potential to reduce reliance on conventional antibiotics, lowering the risk of resistance transmission.

3.3. Probiotics

Probiotics in microecological preparations modulate intestinal microecological balance by increasing lipid metabolism, competitive inhibition, production of antimicrobial compounds, and stimulation of host immune system development, thereby protecting the host from C. perfringens invasion [94]. Some probiotics (i.e., Bacillus subtilis, Lactobacillus, Bacillus velezensis and Bacillus polymyxa) have been proven to prevent C. perfringens [95,96,97]. Lactobacillus, for example, produces antimicrobial compounds, such as bacteriocins. These compounds induce bacterial rupture and disrupt bacterial cell wall synthesis by increasing the permeability of the bacterial inner membrane [98]. Bacillus amyloliquefaciens also improves the gut microbiota following infection with C. perfringens [99].

3.4. Alternative Treatment

Plant extracts, herbal products, phage therapy, and fecal flora transplantation effectively treat C. perfringens infections [100]. Phytic acid has been demonstrated to inhibit the germination and growth of C. perfringens spores, making it a promising alternative food preservative for reducing C. perfringens contamination in various food products [101]. Arginine significantly attenuates α toxin-induced reduction in serum immunoglobulin IgA and IgG levels and intestinal morphological damage [102]. Organic acids can modify intestinal pH and other environmental factors, keep normal intestinal flora in balance, and alter the intestinal microenvironment, lowering the prevalence of C. perfringens infections [103]. Furthermore, yam was found to alleviate ampicillin-induced antibiotic-associated diarrhea by increasing the levels of the probiotics Bifidobacterium bifidum and Lactobacillus lactis and promoting the restoration of bacterial community diversity [4]. Phage therapy is now widely used to manage multidrug-resistant bacteria, and C. perfringens-specific phages have been identifieds [104]. Moreover, fecal flora transplantation involves introducing the gut microbiota from a healthy donor into the gut of a patient to restore balanced and normal gut flora. Fecal flora transplantation has been investigated as a potential treatment for Clostridium difficile infections [105].

4. Conclusions and Prospects

C. perfringens is a common member of the human gut microbiota, but its role as a harmless commensal or a potential pathogen remains controversial. Under normal conditions, C. perfringens maintains intestinal homeostasis together with other microorganisms. However, when the host immune function is reduced, the intestinal flora is dysbiotic, or when broad-spectrum antibiotics are used, C. perfringens proliferates and produces toxins, leading to enterotoxemia, ADD, and food poisoning. Therefore, C. perfringens may be either a harmless bystander in the gut or a serious threat under specific conditions, and further study of its behavioral mechanisms is essential for the prevention and control of related diseases. Based on the principle of “one world, one health”, managing this pathogen and promoting the investigation in this field warrants the following: (1) Explore in-depth the pathogenic mechanism. C. perfringens can produce various toxins and enzymes, but only a subset has been examined, leaving a lack of comprehensive understanding. Utilizing high-throughput sequencing, multi-omics approaches, and microbial culture technologies for in-depth research on the pathogenic mechanism of C. perfringens exotoxins and enzymes will establish a more robust scientific foundation for treating C. perfringens-related diseases. (2) Reducing the spread and infection of C. perfringens. Implement biosecurity measures to boost immunity and actively develop novel vaccines. (3) Implement personalized precision medicine. Developing multi-faceted and multi-pathway therapy is critical for precisely managing C. perfringens infections and reducing antibiotic resistance. This involves exploring resistance-reducing alternatives such as microecological agents, phage therapy, and bacterial colony transplantation.

Author Contributions

X.B. and Y.J. conceived the review concept. X.B. developed the original draft. X.B., Y.J., X.N., Y.G., W.L., Y.L., Y.W. and J.Z. reviewed and approved the final review. All authors have read and agreed to the published version of the manuscript.

Funding

This research was supported by the National Key R&D Program (2022YFC2304000).

Acknowledgments

We are grateful to BioRender for providing the image gallery and to the anonymous reviewers for their constructive comments and suggestions, which greatly improved this paper.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Orrell, K.E.; Melnyk, R.A. Large Clostridial Toxins: Mechanisms and Roles in Disease. Microbiol. Mol. Biol. Rev. 2021, 85. [Google Scholar] [CrossRef] [PubMed]
  2. Bergogne-Bérézin, E. Treatment and Prevention of Antibiotic Associated Diarrhea. Int. J. Antimicrob. Agents 2000, 16, 521–526. [Google Scholar] [CrossRef] [PubMed]
  3. Larcombe, S.; Hutton, M.L.; Lyras, D. Involvement of Bacteria Other Than Clostridium difficile in Antibiotic-Associated Diarrhoea. Trends Microbiol. 2016, 24, 463–476. [Google Scholar] [CrossRef] [PubMed]
  4. Zhang, N.; Liang, T.; Jin, Q.; Shen, C.; Zhang, Y.; Jing, P. Chinese Yam (Dioscorea opposita Thunb.) Alleviates Antibiotic-Associated Diarrhea, Modifies Intestinal Microbiota, and Increases the Level of Short-Chain Fatty Acids in Mice. Food Res. Int. 2019, 122, 191–198. [Google Scholar] [CrossRef] [PubMed]
  5. Alimolaei, M.; Afzali, S. Prevalence of Clostridium perfringens Toxinotypes in Antibiotic-Associated Diarrheal (AAD) Patients in Iranian Hospitals; Can Toxinotype D Serve as a Possible Zoonotic Agent for Humans? Acta Trop. 2023, 247, 107002. [Google Scholar] [CrossRef] [PubMed]
  6. Shrestha, A.; Uzal, F.A.; McClane, B.A. Enterotoxic Clostridia: Clostridium perfringens Enteric Diseases. Microbiol. Spectr. 2018, 6. [Google Scholar] [CrossRef] [PubMed]
  7. Finnie, J.W.; Navarro, M.A.; Uzal, F.A. Pathogenesis and Diagnostic Features of Brain and Ophthalmic Damage Produced by Clostridium perfringens Type D Epsilon Toxin. J. Vet. Diagn. Investig. 2020, 32, 282–286. [Google Scholar] [CrossRef] [PubMed]
  8. Fancher, C.A.; Thames, H.T.; Colvin, M.G.; Zhang, L.; Nuthalapati, N.; Kiess, A.; Dinh, T.T.N.; Sukumaran, A.T. Research Note: Prevalence and Molecular Characteristics of Clostridium perfringens in “No Antibiotics Ever” Broiler Farms. Poult. Sci. 2021, 100, 101414. [Google Scholar] [CrossRef] [PubMed]
  9. Rumah, K.R.; Linden, J.; Fischetti, V.A.; Vartanian, T. Isolation of Clostridium perfringens Type B in an Individual at First Clinical Presentation of Multiple Sclerosis Provides Clues for Environmental Triggers of the Disease. PLoS ONE 2013, 8, e76359. [Google Scholar] [CrossRef]
  10. Chen, J.; Ma, M.; Uzal, F.A.; McClane, B.A. Host Cell-Induced Signaling Causes Clostridium perfringens to Upregulate Production of Toxins Important for Intestinal Infections. Gut Microbes 2014, 5, 96–107. [Google Scholar] [CrossRef]
  11. Wang, Y.-H. Sialidases From Clostridium perfringens and Their Inhibitors. Front. Cell. Infect. Microbiol. 2019, 9, 462. [Google Scholar] [CrossRef] [PubMed]
  12. Bendary, M.M.; Abd El-Hamid, M.I.; El-Tarabili, R.M.; Hefny, A.A.; Algendy, R.M.; Elzohairy, N.A.; Ghoneim, M.M.; Al-Sanea, M.M.; Nahari, M.H.; Moustafa, W.H. Clostridium perfringens Associated with Foodborne Infections of Animal Origins: Insights into Prevalence, Antimicrobial Resistance, Toxin Genes Profiles, and Toxinotypes. Biology 2022, 11, 551. [Google Scholar] [CrossRef]
  13. Yan, Z.; Liu, Z.; Ma, Y.; Yang, Z.; Liu, G.; Fang, J. Effects of Lactobacillus Plantarum and Weissella Viridescens on the Gut Microbiota and Serum Metabolites of Mice with Antibiotic-Associated Diarrhea. Nutrients 2023, 15, 4603. [Google Scholar] [CrossRef] [PubMed]
  14. Ahmed, S.A.; Barış, E.; Go, D.S.; Lofgren, H.; Osorio-Rodarte, I.; Thierfelder, K. Assessing the Global Poverty Effects of Antimicrobial Resistance. World Dev. 2018, 111, 148–160. [Google Scholar] [CrossRef]
  15. Murray, C.J.L.; Ikuta, K.S.; Sharara, F.; Swetschinski, L.; Robles Aguilar, G.; Gray, A.; Han, C.; Bisignano, C.; Rao, P.; Wool, E.; et al. Global Burden of Bacterial Antimicrobial Resistance in 2019: A Systematic Analysis. Lancet 2022, 399, 629–655. [Google Scholar] [CrossRef] [PubMed]
  16. Zhong, J.X.; Zheng, H.R.; Wang, Y.Y.; Bai, L.L.; Du, X.L.; Wu, Y.; Lu, J.X. Molecular Characteristics and Phylogenetic Analysis of Clostridium perfringens from Different Regions in China, from 2013 to 2021. Front. Microbiol. 2023, 14, 1195083. [Google Scholar] [CrossRef] [PubMed]
  17. Holland, D.; Thomson, L.; Mahmoudzadeh, N.; Khaled, A. Estimating Deaths from Foodborne Disease in the UK for 11 Key Pathogens. BMJ Open Gastroenterol. 2020, 7, e000377. [Google Scholar] [CrossRef] [PubMed]
  18. Dolan, G.P.; Foster, K.; Lawler, J.; Amar, C.; Swift, C.; Aird, H.; Gorton, R. An Epidemiological Review of Gastrointestinal Outbreaks Associated with Clostridium perfringens, North East of England, 2012–2014. Epidemiol. Infect. 2016, 144, 1386–1393. [Google Scholar] [CrossRef]
  19. Rajkovic, A.; Jovanovic, J.; Monteiro, S.; Decleer, M.; Andjelkovic, M.; Foubert, A.; Beloglazova, N.; Tsilla, V.; Sas, B.; Madder, A.; et al. Detection of Toxins Involved in Foodborne Diseases Caused by Gram-Positive Bacteria. Compr. Rev. Food Sci. Food Saf. 2020, 19, 1605–1657. [Google Scholar] [CrossRef]
  20. Thomas, M.; Murray, R. Estimating the Burden of Food-Borne Illness in Canada. Can. Commun. Dis. Rep. 2014, 40, 299–302. [Google Scholar] [CrossRef]
  21. Camargo, A.; Rámirez, J.D.; Kiu, R.; Hall, L.J.; Muñoz, M. Unveiling the Pathogenic Mechanisms of Clostridium perfringens Toxins and Virulence Factors. Emerg. Microbes Infect. 2024, 13, 2341968. [Google Scholar] [CrossRef] [PubMed]
  22. Asha, N.J.; Tompkins, D.; Wilcox, M.H. Comparative Analysis of Prevalence, Risk Factors, and Molecular Epidemiology of Antibiotic-Associated Diarrhea Due to Clostridium Difficile, Clostridium perfringens, and Staphylococcus Aureus. J. Clin. Microbiol. 2006, 44, 2785–2791. [Google Scholar] [CrossRef] [PubMed]
  23. Wang, B.; Dong, W.; Ma, L.; Dong, Y.; Wang, S.; Yuan, Y.; Ma, Q.; Xu, J.; Yan, W.; Nan, J.; et al. Prevalence and Genetic Diversity of Clostridium Perfringen Isolates in Hospitalized Diarrheal Patients from Central Chin. Infect. Drug Resist. 2021, 14, 4783–4793. [Google Scholar] [CrossRef] [PubMed]
  24. Sarasoja, M.; Nilson, B.; Wide, D.; Lindberg, Å.; Torisson, G.; Holm, K. Epidemiology, Aetiology and Clinical Characteristics of Clostridial Bacteraemia: A 6-Year Population-Based Observational Study of 386 Patients. Eur. J. Clin. Microbiol. Infect. Dis. 2022, 41, 1305–1314. [Google Scholar] [CrossRef] [PubMed]
  25. Gaucher, M.-L.; Thibodeau, A.; Fravalo, P.; Archambault, M.; Arsenault, J.; Fournaise, S.; Letellier, A.; Quessy, S. Broiler Chicken Carcasses and Their Associated Abattoirs as a Source of Enterotoxigenic Clostridium perfringens: Prevalence and Critical Steps for Contamination. AIMS Microbiol. 2018, 4, 439–454. [Google Scholar] [CrossRef]
  26. Jiang, Y.; Ma, Y.; Liu, Q.; Li, T.; Li, Y.; Guo, K.; Zhang, Y. Tracing Clostridium perfringens Strains from Beef Processing of Slaughter House by Pulsed-Field Gel Electrophoresis, and the Distribution and Toxinotype of Isolates in Shaanxi Province, China. Food Microbiol. 2022, 101, 103887. [Google Scholar] [CrossRef] [PubMed]
  27. Issimov, A.; Baibatyrov, T.; Tayeva, A.; Kenenbay, S.; Abzhanova, S.; Shambulova, G.; Kuzembayeva, G.; Kozhakhiyeva, M.; Brel-Kisseleva, I.; Safronova, O.; et al. Prevalence of Clostridium perfringens and Detection of Its Toxins in Meat Products in Selected Areas of West Kazakhstan. Agriculture 2022, 12, 1357. [Google Scholar] [CrossRef]
  28. Jiang, Y.; Pan, Y.; Yin, J. Prevalence, Toxin-Genotype Distribution, and Transmission of Clostridium perfringens from the Breeding and Milking Process of Dairy Farms. Food Microbiol. 2024, 120, 104485. [Google Scholar] [CrossRef] [PubMed]
  29. Lee, C.A.; Labbé, R. Distribution of Enterotoxin- and Epsilon-Positive Clostridium perfringens Spores in U.S. Retail Spices. J. Food Prot. 2018, 81, 394–399. [Google Scholar] [CrossRef]
  30. Azimirad, M.; Gholami, F.; Yadegar, A.; Knight, D.R.; Shamloei, S.; Aghdaei, H.A.; Zali, M.R. Prevalence and Characterization of Clostridium perfringens Toxinotypes among Patients with Antibiotic-Associated Diarrhea in Iran. Sci. Rep. 2019, 9, 7792. [Google Scholar] [CrossRef]
  31. Santana, J.A.; Ferreira, A.C.D.A.; Souza, M.D.C.C.D.; Moreira, M.A.S.; Lima, M.C.; Cruz, D.S.G.; Lobato, F.C.F.; Silva, R.O.S. Isolation and Genotyping of Clostridium perfringens from Goats in Minas Gerais, Brazil. Ciênc. Rural 2018, 48, e20180101. [Google Scholar] [CrossRef]
  32. Hayati, M.; Tahamtan, Y. Toxin Typing of Clostridium perfringens Associated with Enterotoxaemia in Sheep in Fars Province. Arch. Razi Inst. 2021, 76, 691–697. [Google Scholar] [CrossRef]
  33. Khan, M.U.Z.; Humza, M.; Yang, S.; Alvi, M.A.; Iqbal, M.Z.; Zain-ul-Fatima, H.; Khalid, S.; Munir, T.; Cai, J. Occurrence and Toxicogenetic Profiling of Clostridium perfringens in Buffalo and Cattle: An Update from Pakistan. Toxins 2021, 13, 212. [Google Scholar] [CrossRef] [PubMed]
  34. Wang, X.; Ning, C.; Ji, C.; Li, Y.; Li, J.; Meng, Q.; Qiao, J.; Wang, L.; Cai, K.; Zhang, J.; et al. Antimicrobial Resistance Profiling and Molecular Typing of Ruminant-Borne Isolates of Clostridium perfringens from Xinjiang, China. J. Glob. Antimicrob. Resist. 2021, 27, 41–45. [Google Scholar] [CrossRef]
  35. Schoster, A.; Kunz, T.; Lauper, M.; Graubner, C.; Schmitt, S.; Weese, J.S. Prevalence of Clostridium difficile and Clostridium perfringens in Swiss Horses with and without Gastrointestinal Disease and Microbiota Composition in Relation to Clostridium Difficile Shedding. Vet. Microbiol. 2019, 239, 108433. [Google Scholar] [CrossRef]
  36. Xu, W.; Wang, H.; Liu, L.; Miao, Z.; Huo, Y.; Zhong, Z. Prevalence and characterization of Clostridium perfringens isolated from different chicken farms in China. Anaerobe 2021, 72, 102467. [Google Scholar] [CrossRef]
  37. Moustafa, S.; Zakaria, I.; Moustafa, A.; AboSakaya, R.; Selim, A. Bacteriological and Serological Investigation of Clostridium perfringens in Lambs. Sci. Rep. 2022, 12, 19715. [Google Scholar] [CrossRef]
  38. Yadav, J.P.; Kaur, S.; Dhaka, P.; Vijay, D.; Bedi, J.S. Prevalence, Molecular Characterization, and Antimicrobial Resistance Profile of Clostridium perfringens from India: A Scoping Review. Anaerobe 2022, 77, 102639. [Google Scholar] [CrossRef] [PubMed]
  39. Wu, K.; Feng, H.; Ma, J.; Wang, B.; Feng, J.; Zhang, H.; Jiang, Y.; Li, R.; Wang, J.; Yang, Z. Prevalence, Toxin-Typing and Antimicrobial Susceptibility of Clostridium perfringens in Sheep with Different Feeding Modes from Gansu and Qinghai Provinces, China. Anaerobe 2022, 73, 102516. [Google Scholar] [CrossRef]
  40. Mohiuddin, M.; Song, Z.; Liao, S.; Qi, N.; Li, J.; Lv, M.; Lin, X.; Cai, H.; Hu, J.; Liu, S.; et al. Animal Model Studies, Antibiotic Resistance and Toxin Gene Profile of NE Reproducing Clostridium perfringens Type A and Type G Strains Isolated from Commercial Poultry Farms in China. Microorganisms 2023, 11, 622. [Google Scholar] [CrossRef]
  41. Li, J.; Pradhan, A.; McClane, B.A. NanJ Is the Major Sialidase for Clostridium perfringens Type F Food Poisoning Strain 01E809. Infect. Immun. 2023, 91, e00053-23. [Google Scholar] [CrossRef] [PubMed]
  42. Suzuki, H.; Hosomi, K.; Nasu, A.; Kondoh, M.; Kunisawa, J. Development of Adjuvant-Free Bivalent Food Poisoning Vaccine by Augmenting the Antigenicity of Clostridium perfringens Enterotoxin. Front. Immunol. 2018, 9, 403835. [Google Scholar] [CrossRef]
  43. Li, J.; Uzal, F.A.; McClane, B.A. Clostridium perfringens Sialidases: Potential Contributors to Intestinal Pathogenesis and Therapeutic Targets. Toxins 2016, 8, 341. [Google Scholar] [CrossRef] [PubMed]
  44. Liu, Y.; Lei, Y.-X.; Li, J.-W.; Ma, Y.-Z.; Wang, X.-Y.; Meng, F.-H.; Wu, Y.-J.; Wang, N.; Liang, J.; Zhao, C.-Q.; et al. G Protein-Coupled Receptor 120 Mediates Host Defense against Clostridium perfringens Infection through Regulating NOD-like Receptor Family Pyrin Domain-Containing 3 Inflammasome Activation. J. Agric. Food Chem. 2023, 71, 7119–7130. [Google Scholar] [CrossRef]
  45. Dittoe, D.K.; Johnson, C.N.; Byrd, J.A.; Ricke, S.C.; Piva, A.; Grilli, E.; Swaggerty, C.L. Impact of a Blend of Microencapsulated Organic Acids and Botanicals on the Microbiome of Commercial Broiler Breeders under Clinical Necrotic Enteritis. Animals 2023, 13, 1627. [Google Scholar] [CrossRef]
  46. Low, K.E.; Smith, S.P.; Abbott, D.W.; Boraston, A.B. The Glycoconjugate-Degrading Enzymes of Clostridium perfringens: Tailored Catalysts for Breaching the Intestinal Mucus Barrier. Glycobiology 2021, 31, 681–690. [Google Scholar] [CrossRef] [PubMed]
  47. Van Damme, L.; Callens, C.; Dargatz, M.; Flügel, M.; Hark, S.; Thiemann, F.; Pelzer, S.; Ducatelle, R.; Van Immerseel, F.; Goossens, E. NanI Sialidase Contributes to Toxin Expression and Host Cell Binding of Clostridium perfringens Type G Strain CP56 in Vitro. Vet. Microbiol. 2022, 266, 109371. [Google Scholar] [CrossRef]
  48. Guo, S.; Liu, D.; Zhang, B.; Li, Z.; Li, Y.; Ding, B.; Guo, Y. Two Lactobacillus Species Inhibit the Growth and α-Toxin Production of Clostridium perfringens and Induced Proinflammatory Factors in Chicken Intestinal Epithelial Cells in Vitro. Front. Microbiol. 2017, 8, 2081. [Google Scholar] [CrossRef] [PubMed]
  49. Cao, A.; Chi, H.; Shi, J.; Sun, R.; Du, K.; Song, Y.; Zhu, M.; Zhang, L.; Huang, J. Visual Detection of Clostridium perfringens Alpha Toxin by Combining Nanometer Microspheres with Smart Phones. Microorganisms 2020, 8, 1865. [Google Scholar] [CrossRef]
  50. Chinen, K. Sudden Death Caused by Clostridium perfringens Sepsis Presenting as Massive Intravascular Hemolysis. Autops. Case Rep. 2020, 10, e2020185. [Google Scholar] [CrossRef]
  51. Liu, S.; Yang, X.; Zhang, H.; Zhang, J.; Zhou, Y.; Wang, T.; Hu, N.; Deng, X.; Bai, X.; Wang, J. Amentoflavone Attenuates Clostridium perfringens Gas Gangrene by Targeting Alpha-Toxin and Perfringolysin O. Front. Pharmacol. 2020, 11, 179. [Google Scholar] [CrossRef] [PubMed]
  52. Takagishi, T.; Takehara, M.; Seike, S.; Miyamoto, K.; Kobayashi, K.; Nagahama, M. Clostridium perfringens α-Toxin Impairs Erythropoiesis by Inhibition of Erythroid Differentiation. Sci. Rep. 2017, 7, 5217. [Google Scholar] [CrossRef] [PubMed]
  53. Suzaki, A.; Ohtani, K.; Komine-Aizawa, S.; Matsumoto, A.; Kamiya, S.; Hayakawa, S. Pathogenic Characterization of Clostridium perfringens Strains Isolated From Patients With Massive Intravascular Hemolysis. Front. Microbiol. 2021, 12, 713509. [Google Scholar] [CrossRef]
  54. Takehara, M.; Seike, S.; Sonobe, Y.; Bandou, H.; Yokoyama, S.; Takagishi, T.; Miyamoto, K.; Kobayashi, K.; Nagahama, M. Clostridium perfringens α-Toxin Impairs Granulocyte Colony-Stimulating Factor Receptor-Mediated Granulocyte Production While Triggering Septic Shock. Commun. Biol. 2019, 2, 45. [Google Scholar] [CrossRef]
  55. Bruggisser, J.; Iacovache, I.; Musson, S.C.; Degiacomi, M.T.; Posthaus, H.; Zuber, B. Cryo-EM Structure of the Octameric Pore of Clostridium perfringens β-Toxin. EMBO Rep. 2022, 23, e54856. [Google Scholar] [CrossRef] [PubMed]
  56. Lencer, W.I. Everything Illuminated—Clostridium perfringens β-Toxin. Cell Host Microbe 2020, 28, 5–6. [Google Scholar] [CrossRef]
  57. Tarek, B.; Bruggisser, J.; Cattalani, F.; Posthaus, H. Platelet Endothelial Cell Adhesion Molecule 1 (CD31) Is Essential for Clostridium perfringens Beta-Toxin Mediated Cytotoxicity in Human Endothelial and Monocytic Cells. Toxins 2021, 13, 893. [Google Scholar] [CrossRef]
  58. Bruggisser, J.; Tarek, B.; Wyder, M.; Müller, P.; von Ballmoos, C.; Witz, G.; Enzmann, G.; Deutsch, U.; Engelhardt, B.; Posthaus, H. CD31 (PECAM-1) Serves as the Endothelial Cell-Specific Receptor of Clostridium perfringens β-Toxin. Cell Host Microbe 2020, 28, 69–78.e6. [Google Scholar] [CrossRef]
  59. Navarro, M.A.; McClane, B.A.; Uzal, F.A. Mechanisms of Action and Cell Death Associated with Clostridium perfringens Toxins. Toxins 2018, 10, 212. [Google Scholar] [CrossRef]
  60. Thiel, A.; Mogel, H.; Bruggisser, J.; Baumann, A.; Wyder, M.; Stoffel, M.H.; Summerfield, A.; Posthaus, H. Effect of Clostridium perfringens β-Toxin on Platelets. Toxins 2017, 9, 336. [Google Scholar] [CrossRef]
  61. Finnie, J.W.; Uzal, F.A. Pathology and Pathogenesis of Brain Lesions Produced by Clostridium perfringens Type D Epsilon Toxin. Int. J. Mol. Sci. 2022, 23, 9050. [Google Scholar] [CrossRef] [PubMed]
  62. Savva, C.G.; Clark, A.R.; Naylor, C.E.; Popoff, M.R.; Moss, D.S.; Basak, A.K.; Titball, R.W.; Bokori-Brown, M. The Pore Structure of Clostridium perfringens Epsilon Toxin. Nat. Commun. 2019, 10, 2641. [Google Scholar] [CrossRef] [PubMed]
  63. Ma, Y.; Sannino, D.; Linden, J.R.; Haigh, S.; Zhao, B.; Grigg, J.B.; Zumbo, P.; Dündar, F.; Butler, D.; Profaci, C.P.; et al. Epsilon Toxin-Producing Clostridium perfringens Colonize the Multiple Sclerosis Gut Microbiome Overcoming CNS Immune Privilege. J. Clin. Investig. 2023, 133, e163239. [Google Scholar] [CrossRef]
  64. Shetty, S.V.; Mazzucco, M.R.; Winokur, P.; Haigh, S.V.; Rumah, K.R.; Fischetti, V.A.; Vartanian, T.; Linden, J.R. Clostridium perfringens Epsilon Toxin Binds to and Kills Primary Human Lymphocytes. Toxins 2023, 15, 423. [Google Scholar] [CrossRef] [PubMed]
  65. Morcrette, H.; Bokori-Brown, M.; Ong, S.; Bennett, L.; Wren, B.W.; Lewis, N.; Titball, R.W. Clostridium perfringens Epsilon Toxin Vaccine Candidate Lacking Toxicity to Cells Expressing Myelin and Lymphocyte Protein. NPJ Vaccines 2019, 4, 32. [Google Scholar] [CrossRef] [PubMed]
  66. Geng, Z.; Huang, J.; Kang, L.; Gao, S.; Yuan, Y.; Li, Y.; Wang, J.; Xin, W.; Wang, J. Clostridium perfringens Epsilon Toxin Binds to Erythrocyte MAL Receptors and Triggers Phosphatidylserine Exposure. J. Cell. Mol. Med. 2020, 24, 7341–7352. [Google Scholar] [CrossRef] [PubMed]
  67. Gao, J.; Xin, W.; Huang, J.; Ji, B.; Gao, S.; Chen, L.; Kang, L.; Yang, H.; Shen, X.; Zhao, B.; et al. Hemolysis in Human Erythrocytes by Clostridium perfringens Epsilon Toxin Requires Activation of P2 Receptors. Virulence 2018, 9, 1601–1614. [Google Scholar] [CrossRef]
  68. Ji, B.; Huang, J.; Zou, K.; Liu, M.; Pei, Y.; Huang, J.; Wang, Y.; Wang, J.; Zhou, R.; Xin, W.; et al. Direct Visualization of the Dynamic Process of Epsilon Toxin on Hemolysis. Small Methods 2023, 7, e2300028. [Google Scholar] [CrossRef] [PubMed]
  69. Nagahama, M.; Kobayashi, K.; Takehara, M. Cathepsin Release from Lysosomes Promotes Endocytosis of Clostridium perfringens Iota-Toxin. Toxins 2021, 13, 721. [Google Scholar] [CrossRef]
  70. Takehara, M.; Takagishi, T.; Seike, S.; Oda, M.; Sakaguchi, Y.; Hisatsune, J.; Ochi, S.; Kobayashi, K.; Nagahama, M. Cellular Entry of Clostridium perfringens Iota-Toxin and Clostridium Botulinum C2 Toxin. Toxins 2017, 9, 247. [Google Scholar] [CrossRef]
  71. Nagahama, M.; Takehara, M.; Kobayashi, K. Interaction of Clostridium perfringens Iota Toxin and Lipolysis-Stimulated Lipoprotein Receptor (LSR). Toxins 2018, 10, 405. [Google Scholar] [CrossRef] [PubMed]
  72. Nagahama, M.; Takehara, M.; Miyamoto, K.; Ishidoh, K.; Kobayashi, K. Acid Sphingomyelinase Promotes Cellular Internalization of Clostridium perfringens Iota-Toxin. Toxins 2018, 10, 209. [Google Scholar] [CrossRef] [PubMed]
  73. Mehdizadeh Gohari, I.; Li, J.; Navarro, M.A.; Mendonça, F.S.; Uzal, F.A.; McClane, B.A. Identification of Orphan Histidine Kinases That Impact Sporulation and Enterotoxin Production by Clostridium perfringens Type F Strain SM101 in a Pathophysiologically-Relevant Ex Vivo Mouse Intestinal Contents Model. PLoS Pathog. 2023, 19, e1011429. [Google Scholar] [CrossRef] [PubMed]
  74. Banga, A.R.; Odiase, P.; Rachakonda, K.; Garg, A.P.; Adunyah, S.E.; Rachakonda, G. Application of C-Terminal Clostridium perfringens Enterotoxin in Treatment of Brain Metastasis from Breast Cancer. Cancers 2022, 14, 4309. [Google Scholar] [CrossRef] [PubMed]
  75. Ogbu, C.P.; Roy, S.; Vecchio, A.J. Disruption of Claudin-Made Tight Junction Barriers by Clostridium perfringens Enterotoxin: Insights from Structural Biology. Cells 2022, 11, 903. [Google Scholar] [CrossRef] [PubMed]
  76. Vecchio, A.J.; Rathnayake, S.S.; Stroud, R.M. Structural Basis for Clostridium perfringens Enterotoxin Targeting of Claudins at Tight Junctions in Mammalian Gut. Proc. Natl. Acad. Sci. USA 2021, 118, e2024651118. [Google Scholar] [CrossRef] [PubMed]
  77. Saitoh, Y.; Suzuki, H.; Tani, K.; Nishikawa, K.; Irie, K.; Ogura, Y.; Tamura, A.; Tsukita, S.; Fujiyoshi, Y. Structural insight into tight junction disassembly by Clostridium perfringens enterotoxin. Science 2015, 347, 775–778. [Google Scholar] [CrossRef] [PubMed]
  78. Orlando, B.J.; Dominik, P.K.; Roy, S.; Ogbu, C.P.; Erramilli, S.K.; Kossiakoff, A.A.; Vecchio, A.J. Development, Structure, and Mechanism of Synthetic Antibodies That Target Claudin and Clostridium perfringens Enterotoxin Complexes. J. Biol. Chem. 2022, 298, 102357. [Google Scholar] [CrossRef] [PubMed]
  79. Benz, R.; Popoff, M.R. Clostridium perfringens Enterotoxin: The Toxin Forms Highly Cation-Selective Channels in Lipid Bilayers. Toxins 2018, 10, 341. [Google Scholar] [CrossRef]
  80. Mehdizadeh Gohari, I.; Li, J.; Navarro, M.; Uzal, F.; McClane, B. Effects of Claudin-1 on the Action of Clostridium perfringens Enterotoxin in Caco-2 Cells. Toxins 2019, 11, 582. [Google Scholar] [CrossRef]
  81. Lee, K.-W.; Lillehoj, H.S. Role of Clostridium perfringens Necrotic Enteritis B-like Toxin in Disease Pathogenesis. Vaccines 2021, 10, 61. [Google Scholar] [CrossRef] [PubMed]
  82. Fathima, S.; Hakeem, W.G.A.; Shanmugasundaram, R.; Selvaraj, R.K. Necrotic Enteritis in Broiler Chickens: A Review on the Pathogen, Pathogenesis, and Prevention. Microorganisms 2022, 10, 1958. [Google Scholar] [CrossRef] [PubMed]
  83. Hoseini, Z.S.; Hajizade, A.; Razmyar, J.; Ahmadian, G.; Arpanaei, A. Mesoporous Silica Nanoparticles-Based Formulations of a Chimeric Proteinous Vaccine Candidate against Necrotic Enteritis Disease. Mater. Sci. Eng. C 2021, 128, 112316. [Google Scholar] [CrossRef] [PubMed]
  84. Freitas, N.F.Q.R.; Barbosa, J.D.; Otaka, D.Y.; Ferreira, M.R.A.; Rodrigues, R.R.; Moreira Jr, C.; Conceição, F.R.; Salvarani, F.M. Clostridium perfringens α and β Recombinant Toxoids in Equine Immunization. Pesqui. Veterinária Bras. 2020, 40, 776–780. [Google Scholar] [CrossRef]
  85. Bai, J.; Qiao, X.; Ma, Y.; Han, M.; Jia, S.; Huang, X.; Han, B.; Wang, L.; Li, Y.; Xu, Y. Protection Efficacy of Oral Bait Probiotic Vaccine Constitutively Expressing Tetravalent Toxoids against Clostridium perfringens Exotoxins in Livestock (Rabbits). Vaccines 2020, 8, 17. [Google Scholar] [CrossRef] [PubMed]
  86. Hoelzer, K.; Bielke, L.; Blake, D.P.; Cox, E.; Cutting, S.M.; Devriendt, B.; Erlacher-Vindel, E.; Goossens, E.; Karaca, K.; Lemiere, S.; et al. Vaccines as Alternatives to Antibiotics for Food Producing Animals. Part 2: New Approaches and Potential Solutions. Vet. Res. 2018, 49, 70. [Google Scholar] [CrossRef] [PubMed]
  87. Soto, L.F.; Romaní, A.C.; Jiménez-Avalos, G.; Silva, Y.; Ordinola-Ramirez, C.M.; Lopez Lapa, R.M.; Requena, D. Immunoinformatic Analysis of the Whole Proteome for Vaccine Design: An Application to Clostridium perfringens. Front. Immunol. 2022, 13, 942907. [Google Scholar] [CrossRef] [PubMed]
  88. Aldakheel, F.M.; Abrar, A.; Munir, S.; Aslam, S.; Allemailem, K.S.; Khurshid, M.; Ashfaq, U.A. Proteome-Wide Mapping and Reverse Vaccinology Approaches to Design a Multi-Epitope Vaccine against Clostridium perfringens. Vaccines 2021, 9, 1079. [Google Scholar] [CrossRef]
  89. Mak, P.H.W.; Rehman, M.A.; Kiarie, E.G.; Topp, E.; Diarra, M.S. Production Systems and Important Antimicrobial Resistant-Pathogenic Bacteria in Poultry: A Review. J. Anim. Sci. Biotechnol. 2022, 13, 148. [Google Scholar] [CrossRef]
  90. Gambino, D.; Vicari, D.; Vitale, M.; Schirò, G.; Mira, F.; Giglia, M.L.; Riccardi, A.; Gentile, A.; Giardina, S.; Carrozzo, A.; et al. Study on Bacteria Isolates and Antimicrobial Resistance in Wildlife in Sicily, Southern Italy. Microorganisms 2021, 9, 203. [Google Scholar] [CrossRef]
  91. García-Vela, S.; Martínez-Sancho, A.; Said, L.B.; Torres, C.; Fliss, I. Pathogenicity and Antibiotic Resistance Diversity in Clostridium perfringens Isolates from Poultry Affected by Necrotic Enteritis in Canada. Pathogens 2023, 12, 905. [Google Scholar] [CrossRef] [PubMed]
  92. Duc, H.M.; Hoa, T.T.K.; Ha, C.T.T.; Van Hung, L.; Van Thang, N.; Minh Son, H.; Flory, G.A. Prevalence and Antibiotic Resistance Profile of Clostridium perfringens Isolated from Pork and Chicken Meat in Vietnam. Pathogens 2024, 13, 400. [Google Scholar] [CrossRef]
  93. Vieco-Saiz, N.; Belguesmia, Y.; Raspoet, R.; Auclair, E.; Padgett, C.; Bailey, C.; Gancel, F.; Drider, D. Protective Effects of Novel Lactobacillaceae Strains Isolated from Chicken Caeca against Necrotic Enteritis Infection: In Vitro and In Vivo Evidences. Microorganisms 2022, 10, 152. [Google Scholar] [CrossRef] [PubMed]
  94. Gong, L.; Wang, B.; Zhou, Y.; Tang, L.; Zeng, Z.; Zhang, H.; Li, W. Protective Effects of Lactobacillus Plantarum 16 and Paenibacillus Polymyxa 10 Against Clostridium perfringens Infection in Broilers. Front. Immunol. 2021, 11, 628374. [Google Scholar] [CrossRef] [PubMed]
  95. Hyun, W.B.; Kang, H.S.; Lee, J.W.; Abraha, H.B.; Kim, K.P. A Newly-Isolated Bacillus Subtilis BSC35 Produces Bacteriocin-like Inhibitory Substance with High Potential to Control Clostridium perfringens in Food. LWT 2021, 138, 110625. [Google Scholar] [CrossRef]
  96. Kadekar, D.; Udrea, A.C.; Bak, S.Y.; Christensen, N.; Gibbs, K.; Shen, C.; Bernardeau, M. Cell-Free Culture Supernatant of Lactobacillus Acidophilus AG01 and Bifidobacterium Animalis Subsp. Lactis AG02 Reduces the Pathogenicity of NetB-Positive Clostridium perfringens in a Chicken Intestinal Epithelial Cell Line. Microorganisms 2024, 12, 839. [Google Scholar] [CrossRef]
  97. Zhu La, A.L.T.; Wen, Q.; Xiao, Y.; Hu, D.; Liu, D.; Guo, Y.; Hu, Y. A New Bacillus Velezensis Strain CML532 Improves Chicken Growth Performance and Reduces Intestinal Clostridium perfringens Colonization. Microorganisms 2024, 12, 771. [Google Scholar] [CrossRef] [PubMed]
  98. Monteiro, C.R.A.V.; do Carmo, M.S.; Melo, B.O.; Alves, M.S.; dos Santos, C.I.; Monteiro, S.G.; Bomfim, M.R.Q.; Fernandes, E.S.; Monteiro-Neto, V. In Vitro Antimicrobial Activity and Probiotic Potential of Bifidobacterium and Lactobacillus against Species of Clostridium. Nutrients 2019, 11, 448. [Google Scholar] [CrossRef]
  99. Jiang, Z.; Li, W.; Su, W.; Wen, C.; Gong, T.; Zhang, Y.; Wang, Y.; Jin, M.; Lu, Z. Protective Effects of Bacillus Amyloliquefaciens 40 Against Clostridium perfringens Infection in Mice. Front. Nutr. 2021, 8, 733591. [Google Scholar] [CrossRef]
  100. Hernández, S.; Vives, M.J. Phages in Anaerobic Systems. Viruses 2020, 12, 1091. [Google Scholar] [CrossRef]
  101. Bloot, A.P.M.; Kalschne, D.L.; Nogues, D.R.N.; Amaral, J.S.; Flores, E.L.M.; Colla, E.; Habu, S.; Baraldi, I.J.; Canan, C. Phytic Acid against Clostridium perfringens Type A: A Food Matrix Study. Foods 2022, 11, 406. [Google Scholar] [CrossRef] [PubMed]
  102. Wang, X.; Zhang, T.; Li, W.; Wang, H.; Yan, L.; Zhang, X.; Zhao, L.; Wang, N.; Zhang, B. Arginine Alleviates Clostridium perfringens α Toxin-Induced Intestinal Injury in Vivo and in Vitro via the SLC38A9/mTORC1 Pathway. Front. Immunol. 2024, 15, 1357072. [Google Scholar] [CrossRef] [PubMed]
  103. Kumar, A.; Toghyani, M.; Kheravii, S.K.; Pineda, L.; Han, Y.; Swick, R.A.; Wu, S.-B. Organic Acid Blends Improve Intestinal Integrity, Modulate Short-Chain Fatty Acids Profiles and Alter Microbiota of Broilers under Necrotic Enteritis Challenge. Anim. Nutr. 2022, 8, 82–90. [Google Scholar] [CrossRef] [PubMed]
  104. Lu, R.; Liu, B.; Wu, L.; Bao, H.; García, P.; Wang, Y.; Zhou, Y.; Zhang, H. A Broad-Spectrum Phage Endolysin (LysCP28) Able to Remove Biofilms and Inactivate Clostridium perfringens Strains. Foods 2023, 12, 411. [Google Scholar] [CrossRef]
  105. Johnson, S.; Gerding, D.N. Fecal Fixation: Fecal Microbiota Transplantation for Clostridium difficile Infection. Clin. Infect. Dis. 2017, 64, 272–274. [Google Scholar] [CrossRef]
Figure 1. Roadmap of C. perfringens.
Figure 1. Roadmap of C. perfringens.
Microorganisms 12 01610 g001
Figure 2. (a) Diseases caused by C. perfringens. (b) The direction of research on C. perfringens (Top 10): (I) Global (II) the United States (III) the United Kingdom (IV) China.
Figure 2. (a) Diseases caused by C. perfringens. (b) The direction of research on C. perfringens (Top 10): (I) Global (II) the United States (III) the United Kingdom (IV) China.
Microorganisms 12 01610 g002
Figure 3. Literature published on C. perfringens in the past 7 years.
Figure 3. Literature published on C. perfringens in the past 7 years.
Microorganisms 12 01610 g003
Figure 4. (A) Vascular pathogenesis of C. perfringens CPA: C. perfringens alpha toxin, CPB: C. perfringens beta toxin, ETX: Epsilon toxin, G-CSFR: granulocyte colony-stimulating factor receptor, MEKK: mitogen-activated protein kinase kinase kinase, MKK4: mitogen-activated protein kinase kinase 4, MKK7: mitogen-activated protein kinase kinase 7, JNK: c-Jun N-terminal kinase (B) Intestinal pathogenesis of C. perfringens. Cp: C. perfringens, CPB: C. perfringens beta toxin, CPE: C. perfringens enterotoxin, CPI: Iota toxin, NetB: Necrotizing Enterocolitis B-like toxin, LSR: lipolysis-stimulated lipoprotein receptor. + denotes promotion and − denotes inhibition.
Figure 4. (A) Vascular pathogenesis of C. perfringens CPA: C. perfringens alpha toxin, CPB: C. perfringens beta toxin, ETX: Epsilon toxin, G-CSFR: granulocyte colony-stimulating factor receptor, MEKK: mitogen-activated protein kinase kinase kinase, MKK4: mitogen-activated protein kinase kinase 4, MKK7: mitogen-activated protein kinase kinase 7, JNK: c-Jun N-terminal kinase (B) Intestinal pathogenesis of C. perfringens. Cp: C. perfringens, CPB: C. perfringens beta toxin, CPE: C. perfringens enterotoxin, CPI: Iota toxin, NetB: Necrotizing Enterocolitis B-like toxin, LSR: lipolysis-stimulated lipoprotein receptor. + denotes promotion and − denotes inhibition.
Microorganisms 12 01610 g004
Table 1. Prevalence Rate and Number of Infections of C. perfringens in Humans.
Table 1. Prevalence Rate and Number of Infections of C. perfringens in Humans.
SiteDiseasePrevalence Rate (%) or Number of InfectionsReference
USAFoodborne illness107[19]
CanadaFoodborne infection1.6 × 105[20]
EnglandFoodborne illness9 × 106[21]
IranAntibiotic-associated diarrhea15%[30]
Antibiotic-associated diarrhea5%–20%[16]
Table 2. Prevalence Rate and Number of Infections of C. perfringens in Animals.
Table 2. Prevalence Rate and Number of Infections of C. perfringens in Animals.
YearSiteDiseaseSpeciesPrevalence Rate (%) or Number of InfectionsReference
2013–2016SwitzerlandHorse8%[35]
2014–2015Samsun (Turkey)SepticemiaSheep45.2%[36]
2021–2022Kalubiya and Menofia (Egypt)Lamb17.5%[37]
2000–2021IndiaPig60%[38]
Sheep56.3%[38]
Goat38.7%[38]
Cattle35%[38]
Weifang, Guangdong, Taian, Pingying (China)Chicken38.42%[36]
2019–2020Gansu (China)Sheep14.7%[39]
2020Anhui, Guangdong, Guangxi, Fujian (China)Ruminant44.173%[40]
Table 3. C. perfringens vaccines.
Table 3. C. perfringens vaccines.
YearNationType of VaccineSpeciesAdvantagesPatent Number
2018ChinaSubunit vaccineSimple preparation process, low immunizing dose, higher vaccine efficacyCN107753940A
2017ChinaToxin vaccineCattle and SheepStrong immunity, and no toxic or side effectsCN107875377A
2013South KoreaGenetically engineered vaccinePoultrySafe and efficientKP101293667B1
2008CanadaPeptide vaccinePoultrySafe and cost-effectiveCA2685533A1
2008ChinaAttenuated live vaccineSafe and strong immunityEP2007420A2
1995UKGenetically engineered vaccineSafe and easy to produceEP0642581A1
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Ba, X.; Jin, Y.; Ning, X.; Gao, Y.; Li, W.; Li, Y.; Wang, Y.; Zhou, J. Clostridium perfringens in the Intestine: Innocent Bystander or Serious Threat? Microorganisms 2024, 12, 1610. https://doi.org/10.3390/microorganisms12081610

AMA Style

Ba X, Jin Y, Ning X, Gao Y, Li W, Li Y, Wang Y, Zhou J. Clostridium perfringens in the Intestine: Innocent Bystander or Serious Threat? Microorganisms. 2024; 12(8):1610. https://doi.org/10.3390/microorganisms12081610

Chicago/Turabian Style

Ba, Xuli, Youshun Jin, Xuan Ning, Yidan Gao, Wei Li, Yunhui Li, Yihan Wang, and Jizhang Zhou. 2024. "Clostridium perfringens in the Intestine: Innocent Bystander or Serious Threat?" Microorganisms 12, no. 8: 1610. https://doi.org/10.3390/microorganisms12081610

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop