1
|
Li J, Sayeed S, McClane BA. The presence of differentiated C2C12 muscle cells enhances toxin production and growth by Clostridium perfringens type A strain ATCC3624. Virulence 2024; 15:2388219. [PMID: 39192628 PMCID: PMC11364075 DOI: 10.1080/21505594.2024.2388219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 07/03/2024] [Accepted: 07/28/2024] [Indexed: 08/29/2024] Open
Abstract
Clostridium perfringens type A causes gas gangrene, which involves muscle infection. Both alpha toxin (PLC), encoded by the plc gene, and perfringolysin O (PFO), encoded by the pfoA gene, are important when type A strains cause gas gangrene in a mouse model. This study used the differentiated C2C12 muscle cell line to test the hypothesis that one or both of those toxins contributes to gas gangrene pathogenesis by releasing growth nutrients from muscle cells. RT-qPCR analyses showed that the presence of differentiated C2C12 cells induces C. perfringens type A strain ATCC3624 to upregulate plc and pfoA expression, as well as increase expression of several regulatory genes, including virS/R, agrB/D, and eutV/W. The VirS/R two component regulatory system (TCRS) and its coupled Agr-like quorum sensing system, along with the EutV/W TCRS (which regulates expression of genes involved in ethanolamine [EA] utilization), were shown to mediate the C2C12 cell-induced increase in plc and pfoA expression. EA was demonstrated to increase toxin gene expression. ATCC3624 growth increased in the presence of differentiated C2C12 muscle cells and this effect was shown to involve both PFO and PLC. Those membrane-active toxins were each cytotoxic for differentiated C2C12 cells, suggesting they support ATCC3624 growth by releasing nutrients from differentiated C2C12 cells. These findings support a model where, during gas gangrene, increased production of PFO and PLC in the presence of muscle cells causes more damage to those host cells, which release nutrients like EA that are then used to support C. perfringens growth in muscle.
Collapse
Affiliation(s)
- Jihong Li
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Sameera Sayeed
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Bruce A. McClane
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| |
Collapse
|
2
|
Salvarani FM, Vieira EV. Clostridial Infections in Cattle: A Comprehensive Review with Emphasis on Current Data Gaps in Brazil. Animals (Basel) 2024; 14:2919. [PMID: 39457848 PMCID: PMC11506116 DOI: 10.3390/ani14202919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Revised: 10/02/2024] [Accepted: 10/03/2024] [Indexed: 10/28/2024] Open
Abstract
Clostridial infections in cattle are a significant concern for Brazilian livestock. These diseases are caused by various species of Clostridium, which are known for their ability to produce potent toxins. Botulism in cattle is a serious and often fatal condition caused by the ingestion of neurotoxins produced by C. botulinum. This bacterium thrives in decomposing organic matter, such as spoiled feed, carcasses, and contaminated water. Tetanus is less common, but it is a serious disease that follows the contamination of wounds with Clostridium tetani spores. It results in muscle stiffness, spasms, and often death due to respiratory failure. Blackleg (C. chauvoei) is a disease that primarily affects young cattle, leading to acute lameness, swelling, and high fever. Malignant edema (C. septicum and others) is characterized by rapid onset of swelling at wound sites, and it can occur after injuries or surgical procedures. Enterotoxemia is triggered by the rapid growth of C. perfringens in the gut following excessive carbohydrate intake. This leads to toxin production that causes sudden death. In conclusion, clostridial bovine infections remain a persistent challenge for Brazilian cattle farmers. With continued focus on vaccination, good management practices, and research, the impact of these diseases can be minimized, safeguarding the livestock industry's economic viability.
Collapse
Affiliation(s)
- Felipe Masiero Salvarani
- Instituto de Medicina Veterinária, Universidade Federal do Pará, Castanhal 68740-970, PA, Brazil
| | | |
Collapse
|
3
|
Motta JF, Ferreira MRA, Waller SB, Rodrigues RR, Donassolo RA, Moreira Júnior C, Alves MLF, Feijó FD, Conceição FR. Immunogenicity of a pentavalent recombinant Escherichiacoli bacterin against enterotoxemia and botulism in sheep. Anaerobe 2024; 89:102895. [PMID: 39122140 DOI: 10.1016/j.anaerobe.2024.102895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 05/24/2024] [Accepted: 08/06/2024] [Indexed: 08/12/2024]
Abstract
INTRODUCTION Producing commercial bacterins/toxoids against Clostridium spp. is laborious and hazardous. Conversely, developing prototype vaccines using purified recombinant toxoids, though safe and effective, is both laborious and costly for application in production animals. OBJECTIVE Considering that inactivated recombinant Escherichiacoli (bacterin) is a simple, cost-effective, and to be safe solution, we evaluated, for the first time, a pentavalent formulation of recombinant bacterins containing the alpha, beta, and epsilon toxins of Clostridiumperfringens and C and D neurotoxins of Clostridiumbotulinum in sheep. METHODS Subcutaneously, 18 Texel sheep received two doses (200 μg of each antigen) of recombinant bacterin (n = 7) or purified recombinant antigens (n = 6) on days 0 and 28, while the control group (n = 5) did not receive an immunization. Sera samples from days 0 (before the 1st dose), 28 (before the 2nd dose), and 56, 84, and 112 were used for measuring IgG (indirect ELISA) and neutralizing antibodies (mouse serum neutralization). RESULTS Both formulations induced significant levels of IgG against all five toxins (p < 0.05) up to day 112, with peaks at days 28 and 56 post-immunization. The expected booster effect occurred only for the botulinum toxins. The neutralizing antibody titers were satisfactory against ETX (≥2 IU/ml for both formulations) and BoNT-D [5 IU/ml (bacterin) and 10 IU/ml (purified)]. CONCLUSION While adjustments are required, the recombinant bacterin platform holds great potential for polyvalent vaccines due to its straightforward, safe, and cost-effective production, establishing it as a user-friendly technology for the veterinary immunobiological industry.
Collapse
Affiliation(s)
- Jaqueline Freitas Motta
- Programa de Pós-Graduação em Veterinária, Faculdade de Veterinária, Universidade Federal de Pelotas (UFPEL), Pelotas, RS, Brazil
| | - Marcos Roberto A Ferreira
- Centro de Desenvolvimento Tecnológico, Biotecnologia, Universidade Federal de Pelotas (UFPEL), Pelotas, RS, Brazil.
| | - Stefanie Bressan Waller
- Programa de Pós-Graduação em Veterinária, Faculdade de Veterinária, Universidade Federal de Pelotas (UFPEL), Pelotas, RS, Brazil; Centro de Desenvolvimento Tecnológico, Biotecnologia, Universidade Federal de Pelotas (UFPEL), Pelotas, RS, Brazil
| | - Rafael Rodrigues Rodrigues
- Centro de Desenvolvimento Tecnológico, Biotecnologia, Universidade Federal de Pelotas (UFPEL), Pelotas, RS, Brazil
| | - Rafael Amaral Donassolo
- Centro de Desenvolvimento Tecnológico, Biotecnologia, Universidade Federal de Pelotas (UFPEL), Pelotas, RS, Brazil
| | - Clóvis Moreira Júnior
- Centro de Desenvolvimento Tecnológico, Biotecnologia, Universidade Federal de Pelotas (UFPEL), Pelotas, RS, Brazil
| | - Mariliana Luiza Ferreira Alves
- Centro de Desenvolvimento Tecnológico, Biotecnologia, Universidade Federal de Pelotas (UFPEL), Pelotas, RS, Brazil; Instituto Federal Sul-rio-grandense (IFSul), Campus Pelotas, Pelotas, RS, Brazil
| | - Fernanda Dornelles Feijó
- Departamento de Zootecnia, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - Fabricio Rochedo Conceição
- Programa de Pós-Graduação em Veterinária, Faculdade de Veterinária, Universidade Federal de Pelotas (UFPEL), Pelotas, RS, Brazil; Centro de Desenvolvimento Tecnológico, Biotecnologia, Universidade Federal de Pelotas (UFPEL), Pelotas, RS, Brazil
| |
Collapse
|
4
|
Teige ES, Hillestad EMR, Steinsvik EK, Brønstad I, Lundervold A, Lundervold AJ, Valeur J, Hausken T, Berentsen B, Lied GA. Fecal bacteria and short-chain fatty acids in irritable bowel syndrome: Relations to subtype. Neurogastroenterol Motil 2024; 36:e14854. [PMID: 38946176 DOI: 10.1111/nmo.14854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 05/16/2024] [Accepted: 06/15/2024] [Indexed: 07/02/2024]
Abstract
BACKGROUND The relationship between gut microbiota and irritable bowel syndrome (IBS) subtype is unclear. We aimed to explore whether differences in fecal bacteria composition and short-chain fatty acid (SCFA) levels were associated with subtypes and symptoms of IBS. METHODS All participants delivered fecal samples and self-reports on IBS Symptom Severity Score (IBS-SSS), Bristol Stool Scale (BSS), and Gastrointestinal Symptom Rating Scale (GSRS). Fecal bacteria composition was assessed by the GA-map® Dysbiosis Test based on 16S rRNA sequences of bacterial species/groups. SCFAs were analyzed by vacuum distillation followed by gas chromatography. KEY RESULTS Sixty patients with IBS were included (mean age 38 years, 46 [77%] females): Twenty-one patients were classified as IBS-D (diarrhea), 31 IBS-M (mixed diarrhea and constipation), and eight IBS-C (constipation). Forty-two healthy controls (HCs) (mean age 35 years, 27 [64%] females) were included. Patients had a significantly higher relative frequency of dysbiosis, lower levels of Actinobacteria, and higher levels of Bacilli than HCs. Eight bacterial markers were significantly different across IBS subgroups and HCs, and 13 bacterial markers were weakly correlated with IBS symptoms. Clostridia and Veillonella spp. had a weak negative correlation with constipation scores (GSRS) and a weak positive correlation with loose stools (BSS). Diarrhea scores (GSRS) and looser stool (BSS) were weakly correlated with levels of total SCFAs, acetic and butyric acid. Levels of total SCFAs and acetic acid were weakly correlated with symptom severity (IBS-SSS). CONCLUSIONS & INFERENCES Patients with IBS had a different fecal bacteria composition compared to HCs, and alterations of SCFAs may contribute to the subtype.
Collapse
Affiliation(s)
- Erica Sande Teige
- Centre for Nutrition, Department of Clinical Medicine, University of Bergen, Bergen, Norway
- National Centre for Functional Gastrointestinal Disorders, Department of Medicine, Haukeland University Hospital, Bergen, Norway
| | - Eline Margrete Randulff Hillestad
- Centre for Nutrition, Department of Clinical Medicine, University of Bergen, Bergen, Norway
- National Centre for Functional Gastrointestinal Disorders, Department of Medicine, Haukeland University Hospital, Bergen, Norway
| | - Elisabeth Kjelsvik Steinsvik
- National Centre for Functional Gastrointestinal Disorders, Department of Medicine, Haukeland University Hospital, Bergen, Norway
| | - Ingeborg Brønstad
- National Centre for Functional Gastrointestinal Disorders, Department of Medicine, Haukeland University Hospital, Bergen, Norway
| | - Arvid Lundervold
- Mohn Medical Imaging and Visualization Centre, Haukeland University Hospital, Bergen, Norway
- Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Astri J Lundervold
- Department of Biological and Medical Psychology, University of Bergen, Bergen, Norway
| | - Jørgen Valeur
- Unger-Vetlesen Institute, Lovisenberg Diaconal Hospital Oslo, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Trygve Hausken
- Centre for Nutrition, Department of Clinical Medicine, University of Bergen, Bergen, Norway
- National Centre for Functional Gastrointestinal Disorders, Department of Medicine, Haukeland University Hospital, Bergen, Norway
| | - Birgitte Berentsen
- National Centre for Functional Gastrointestinal Disorders, Department of Medicine, Haukeland University Hospital, Bergen, Norway
| | - Gülen Arslan Lied
- Centre for Nutrition, Department of Clinical Medicine, University of Bergen, Bergen, Norway
- National Centre for Functional Gastrointestinal Disorders, Department of Medicine, Haukeland University Hospital, Bergen, Norway
| |
Collapse
|
5
|
Mubarak AG, Khalifa FA, Elsobky Y, Abdel-Rady A, Felefel W, Saad AH, Abdelhiee EY, Alhassan AM, Awny H, Elghazaly EM, Abu-Seida AM, Abdulkarim A, Youseef AG. Sudden death due to enterotoxemia among Arabian camels ( Camelus dromedaries) and associated risk factors. Open Vet J 2024; 14:1942-1951. [PMID: 39308733 PMCID: PMC11415913 DOI: 10.5455/ovj.2024.v14.i8.23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Accepted: 07/08/2024] [Indexed: 09/25/2024] Open
Abstract
Background Sudden death is defined as an unexpected death occurring with no observed antecedent clinical signs. Aim The current study was performed to notice the tangible causes of sudden death among 51 out of 340 she-camels on a private farm in the eastern region of El Khafgi, Saudi Arabia. Methods A retrospective cohort study design was conducted to investigate the sudden death of camels through microscopic examination of fecal matter to identify the gastrointestinal parasites, analysis of whole blood thin films to diagnose blood parasites, blood culturing to recognize bacterial infection as Pasteurella multicida, and macroscopic postmortem examination to identify the gastrointestinal adult worm. The quantity and composition of feed were also analyzed. Afterward, a commercial multiscreen Ag-ELISA kit technique determined the toxins of Clostridium perfringens (C. perfringens). Results The results revealed that the incidence rate of sudden death was 15%. The sudden death occurred due to C. perfringens enterotoxins detected in the rumen, intestinal content, and intestinal wall. The enterotoxins and Alpha toxins were noticed, but the other toxin types, including Beta and Epsilon, could not be detected. All C. perfringens toxins were discovered to be negative in fecal matter. A significant association was reported between sudden death, she-camels age, and feeding habits as risk factors (p = 0.020 and 0.028, respectively). Risk factor assessment by relative risk (RR) revealed that the odds of RR of sudden death occurring among she-camels aged over two years were higher than those less than two years (2.24 CI 95%, 1.093-4.591). Furthermore, the odds RR of sudden death occurring due to exposure of she-camels to a concentrated ration of 18% were higher twice than those not exposed (2.346 CI 95%, 1.039-5.296). Conclusion Clostridium perfringens enterotoxaemia should be listed as a cause of sudden death in camels and the alteration in diet with 18% concentration feed changes the intestinal environment, which leads to C. perfringens proliferating and yielding potent toxins. More observations and interferences like regular immunization are recommended to reduce the disease and increase the awareness of the farmers of the importance of risk factors.
Collapse
Affiliation(s)
- Asmaa G. Mubarak
- Department of Zoonoses, Faculty of Veterinary Medicine, South Valley University, Qena, Egypt
| | - Fatma A. Khalifa
- Department of Infectious Diseases, Faculty of Veterinary Medicine, South Valley University, Qena, Egypt
| | - Yumna Elsobky
- Department of Hygiene and Zoonosis, Faculty of Veterinary Medicine, University of Sadat City, Menofia, Sadat City, Egypt
| | - Ahmed Abdel-Rady
- Department of Animal Medicine (Infectious Diseases), Faculty of Veterinary Medicine, Assiut University, Assiut, Egypt
- Consultant of Infectious Diseases in Animal Health Laboratory, WEQAA-Center, Dammam, Kingdom of Saudi Arabia
| | - Wael Felefel
- Department of Parasitology, Faculty of Veterinary Medicine, Matrouh University, Matrouh, Egypt
| | - Adel Hassan Saad
- Nutrition and Clinical Nutrition Department, Faculty of Veterinary Medicine, Matrouh University, Matrouh, Egypt
| | - Ehab Y. Abdelhiee
- Department of Forensic Medicine and Toxicology, Faculty of Veterinary Medicine, Matrouh University, Matrouh, Egypt
| | - Abdullah M. Alhassan
- Director of Laboratories in Eastern Region, WEQAA-Center, National Center for the Prevention and Control of Plants Pests and Animal Diseases, Dammam, Kingdom of Saudi Arabia
| | - Hisham Awny
- High Institute of Public Health, Alexandria University, Alexandria, Egypt
| | - Eman M. Elghazaly
- Department of Microbiology, Faculty of Veterinary Medicine, Matrouh University, Matrouh, Egypt
| | - Ashraf M. Abu-Seida
- Department of Surgery, Anesthesiology and Radiology, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
| | | | - Asmaa G. Youseef
- Department of Zoonoses, Faculty of Veterinary Medicine, South Valley University, Qena, Egypt
| |
Collapse
|
6
|
Arthur JD, Mullen JL, Uzal FA, Nagamine CM, Casey KM. Epizootic of enterocolitis and clostridial overgrowth in NSG and NSG-related mouse strains. Vet Pathol 2024; 61:653-663. [PMID: 38140953 DOI: 10.1177/03009858231217197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2023]
Abstract
While the immunodeficient status of NOD.Cg-Prkdcscid Il2rgtm1Wjl/SzJ (NSG) and NSG-related mice provides utility for numerous research models, it also results in increased susceptibility to opportunistic pathogens. Over a 9-week period, a high rate of mortality was reported in a housing room of NSG and NSG-related mice. Diagnostics were performed to determine the underlying etiopathogenesis. Mice submitted for evaluation included those found deceased (n = 2), cage mates of deceased mice with or without diarrhea (n = 17), and moribund mice (n = 8). Grossly, mice exhibited small intestinal and cecal dilation with abundant gas and/or digesta (n = 18), serosal hemorrhage and congestion (n = 6), or were grossly normal (n = 3). Histologically, there was erosive to ulcerative enterocolitis (n = 7) of the distal small and large intestine or widespread individual epithelial cell death with luminal sloughing (n = 13) and varying degrees of submucosal edema and mucosal hyperplasia. Cecal dysbiosis, a reduction in typical filamentous bacteria coupled with overgrowth of bacterial rods, was identified in 18 of 24 (75%) mice. Clostridium spp. and Paeniclostridium sordellii were identified in 13 of 23 (57%) and 7 of 23 (30%) mice, respectively. Clostridium perfringens (7 of 23, 30%) was isolated most frequently. Toxinotyping of C. perfringens positive mice (n = 2) identified C. perfringens type A. Luminal immunoreactivity to several clostridial species was identified within lesioned small intestine by immunohistochemistry. Clinicopathologic findings were thus associated with overgrowth of various clostridial species, though direct causality could not be ascribed. A diet shift preceding the mortality event may have contributed to loss of intestinal homeostasis.
Collapse
|
7
|
Xu Z, Feng X, Song Z, Li X, Li K, Li M, Wang X, Liu B, Sun C. Cell-Free Supernatant of Bacillus subtilis G2B9-Q Improves Intestinal Health and Modulates Immune Response to Promote Mouse Recovery in Clostridium perfringens Infection. Curr Microbiol 2024; 81:243. [PMID: 38935166 DOI: 10.1007/s00284-024-03669-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Accepted: 03/17/2024] [Indexed: 06/28/2024]
Abstract
Clostridium perfringens is one of the critical causative agents causing diarrhea in piglets, with significant economic losses to the pig industry. Under normal gut microbiota homeostasis and well-managed barns, diarrhea caused by C. perfringens could be controlled. Some reports show that probiotics, such as Bacillus subtilis, are beneficial in preventing necrotic enteritis (NE) in chickens, but few reports on piglets. Clostridium perfringens was found in the piglets' diarrhea with intestinal microbiota dysbiosis in our survey. Bacillus subtilis G2B9-Q, which was isolated from the feces of healthy pigs, was found to have anti-Clostridium activity after screening. Clostridium perfringens was used to challenge mice by intraperitoneal injection for modeling to evaluate the anti-infective activity of cell-free supernatant (CFS) of B. subtilis G2B9-Q and different concentrations of B. subtilis G2B9-Q by oral administration. The results showed that G2B9-Q can mitigate intestinal lesions caused by C. perfringens infection, reduce inflammatory reactions, and modulate intestinal microbiota. The CFS of G2B9-Q can alleviate the pathological damage of intestinal tissues caused by C. perfringens infection, reduce the concentration of TNF-α and IL-10 in the sera of mice, as well as the relative expression levels of alpha toxin (CPA), perfringolysin O (PFO) toxin, IL-10, IL-22, and TNF-α in the jejunum and colon tissues, and alleviate the changes in gut microbiota structure caused by C. perfringens infection, which showed better therapeutic effects and indicated that the metabolites of G2B9-Q are essential mediators for their beneficial effects. Therefore, the CFS of G2B9-Q could potentially replace antibiotics in treating C. perfringens infection.
Collapse
Affiliation(s)
- Zhiqiang Xu
- College of Veterinary Medicine, Jilin University, Xi'an Street 5333#, Changchun, 130062, Jilin, China
| | - Xin Feng
- College of Veterinary Medicine, Jilin University, Xi'an Street 5333#, Changchun, 130062, Jilin, China
| | - Zhanyun Song
- Changchun Customs District, Changchun, Jilin, China
| | - Xiang Li
- Changchun Customs District, Changchun, Jilin, China
| | - Ke Li
- College of Veterinary Medicine, Jilin University, Xi'an Street 5333#, Changchun, 130062, Jilin, China
| | - Mengjiao Li
- Changchun Customs District, Changchun, Jilin, China
| | | | - Bo Liu
- Changchun Customs District, Changchun, Jilin, China
| | - Changjiang Sun
- College of Veterinary Medicine, Jilin University, Xi'an Street 5333#, Changchun, 130062, Jilin, China.
| |
Collapse
|
8
|
Zheng Y, Yang Q, Luo J, Zhang Y, Li X, He L, Ma C, Tao L. Identification of a hemorrhagic determinant in Clostridioides difficile TcdA and Paeniclostridium sordellii TcsH. Microbiol Spectr 2024; 12:e0035424. [PMID: 38709085 PMCID: PMC11237598 DOI: 10.1128/spectrum.00354-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 04/18/2024] [Indexed: 05/07/2024] Open
Abstract
Paeniclostridium sordellii hemorrhagic toxin (TcsH) and Clostridioides difficile toxin A (TcdA) are two major members of the large clostridial toxin (LCT) family. These two toxins share ~87% similarity and are known to cause severe hemorrhagic pathology in animals. Yet, the pathogenesis of their hemorrhagic toxicity has been mysterious for decades. Here, we examined the liver injury after systemic exposure to different LCTs and found that only TcsH and TcdA induce overt hepatic hemorrhage. By investigating the chimeric and truncated toxins, we demonstrated that the enzymatic domain of TcsH alone is not sufficient to determine its potent hepatic hemorrhagic toxicity in mice. Likewise, the combined repetitive oligopeptide (CROP) domain of TcsH/TcdA alone also failed to explain their strong hemorrhagic activity in mice. Lastly, we showed that disrupting the first two short repeats of CROPs in TcsH and TcdA impaired hemorrhagic toxicity without causing overt changes in cytotoxicity and lethality. These findings lead to a deeper understanding of toxin-induced hemorrhage and the pathogenesis of LCTs and could be insightful in developing therapeutic avenues against clostridial infections. IMPORTANCE Paeniclostridium sordellii and Clostridioides difficile infections often cause hemorrhage in the affected tissues and organs, which is mainly attributed to their hemorrhagic toxins, TcsH and TcdA. In this study, we demonstrate that TcsH and TcdA, but not other related toxins. including Clostridioides difficile toxin B and TcsL, induce severe hepatic hemorrhage in mice. We further determine that a small region in TcsH and TcdA is critical for the hemorrhagic toxicity but not cytotoxicity or lethality of these toxins. Based on these results, we propose that the hemorrhagic toxicity of TcsH and TcdA is due to an uncharacterized mechanism, such as the presence of an unknown receptor, and future studies to identify the interactive host factors are warranted.
Collapse
Affiliation(s)
- Yangling Zheng
- College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, China
- Center for Infectious Disease Research, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
- Research Center for Industries of the Future and Key Laboratory of Multi-omics in Infection and Immunity of Zhejiang Province, School of Medicine, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
- Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China
| | - Qi Yang
- College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, China
- Center for Infectious Disease Research, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
- Research Center for Industries of the Future and Key Laboratory of Multi-omics in Infection and Immunity of Zhejiang Province, School of Medicine, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
- Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China
| | - Jianhua Luo
- Center for Infectious Disease Research, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
- Research Center for Industries of the Future and Key Laboratory of Multi-omics in Infection and Immunity of Zhejiang Province, School of Medicine, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
- Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China
| | - Yuanyuan Zhang
- Center for Infectious Disease Research, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
- Research Center for Industries of the Future and Key Laboratory of Multi-omics in Infection and Immunity of Zhejiang Province, School of Medicine, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
- Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China
| | - Xingxing Li
- Center for Infectious Disease Research, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
- Research Center for Industries of the Future and Key Laboratory of Multi-omics in Infection and Immunity of Zhejiang Province, School of Medicine, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
- Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China
| | - Liuqing He
- Center for Infectious Disease Research, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
- Research Center for Industries of the Future and Key Laboratory of Multi-omics in Infection and Immunity of Zhejiang Province, School of Medicine, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
- Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China
| | - Chao Ma
- Center for Infectious Disease Research, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
- Research Center for Industries of the Future and Key Laboratory of Multi-omics in Infection and Immunity of Zhejiang Province, School of Medicine, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
- Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China
| | - Liang Tao
- College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, China
- Center for Infectious Disease Research, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
- Research Center for Industries of the Future and Key Laboratory of Multi-omics in Infection and Immunity of Zhejiang Province, School of Medicine, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
- Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China
| |
Collapse
|
9
|
Lakes JE, Ferrell JL, Berhow MA, Flythe MD. Antimicrobial effects of cannabidiol on select agriculturally important Clostridia. Anaerobe 2024; 87:102843. [PMID: 38537865 DOI: 10.1016/j.anaerobe.2024.102843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 03/22/2024] [Accepted: 03/24/2024] [Indexed: 06/18/2024]
Abstract
Amino acid-fermenting Clostridia have undesirable effects in agricultural systems, which can be mitigated by antibiotics, but resistance necessitates alternatives. Here, we demonstrate the efficacy of cannabidiol on growth and ammonia inhibition of five agriculturally relevant Clostridia: Clostridium sporogenes, Peptostreptococcus spp., Clostridioides difficile, Acetoanaerobium sticklandii, and Clostridium aminophilum.
Collapse
Affiliation(s)
- Jourdan E Lakes
- USDA Agricultural Research Service, Forage-Animal Production Research Unit, Lexington, KY, USA
| | - Jessica L Ferrell
- USDA Agricultural Research Service, Forage-Animal Production Research Unit, Lexington, KY, USA
| | - Mark A Berhow
- USDA Agricultural Research Service, National Center for Agricultural Utilization Research, Peoria, IL, USA
| | - Michael D Flythe
- USDA Agricultural Research Service, Forage-Animal Production Research Unit, Lexington, KY, USA; Department of Animal and Food Sciences, College of Agriculture, Food and Environment, University of Kentucky, Lexington, KY, USA.
| |
Collapse
|
10
|
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. [PMID: 38431330 DOI: 10.1016/j.fm.2024.104485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Revised: 01/03/2024] [Accepted: 02/03/2024] [Indexed: 03/05/2024]
Abstract
This study aimed to elucidate the distribution, transmission, and cross-contamination of Clostridium perfringens during the breeding and milking process from dairy farms. The prevalence of 22.3% (301/1351) yielded 494 C. perfringens isolates; all isolates were type A, except for one type D, and 69.8% (345/494) of the isolates carried atyp. cpb2 and only 0.6% (3/494) of the isolates carried cons. cpb2. C. perfringens detected throughout the whole process but without type F. 150 isolates were classified into 94 pulsed-field gel electrophoresis (PFGE) genotypes; among them, six clusters contained 34 PFGE genotypes with 58.0% isolates which revealed epidemic correlation and genetic diversity; four PFGE genotypes (PT57, PT9, PT61, and PT8) were the predominant genotypes. The isolates from different farms demonstrated high homology. Our study confirmed that C. perfringens demonstrated broad cross-contamination from nipples and hides of dairy cattle, followed by personnel and tools and air-introduced raw milk during the milking process. In conclusion, raw milk could serve as a medium for the transmission of C. perfringens, which could result in human food poisoning. Monitoring and controlling several points of cross-contamination during the milking process are essential as is implementing stringent hygiene measures to prevent further spread and reduce the risk of C. perfringens infection.
Collapse
Affiliation(s)
- Yanfen Jiang
- College of Veterinary Medicine, Northwest A&F University, 712100, Yangling, Shaanxi, China.
| | - Yifan Pan
- College of Food Science and Engineering, Northwest A&F University, 712100, Yangling, Shaanxi, China
| | - Jingyi Yin
- College of Veterinary Medicine, Northwest A&F University, 712100, Yangling, Shaanxi, China
| |
Collapse
|
11
|
Xue P, Xue M, Luo Y, Tang Q, Wang F, Sun R, Song Y, Chao Z, Fang M. Colonic Microbiota Improves Fiber Digestion Ability and Enhances Absorption of Short-Chain Fatty Acids in Local Pigs of Hainan. Microorganisms 2024; 12:1033. [PMID: 38930415 PMCID: PMC11205767 DOI: 10.3390/microorganisms12061033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Revised: 05/05/2024] [Accepted: 05/16/2024] [Indexed: 06/28/2024] Open
Abstract
Compared to commercial breeds, Chinese local pig breeds have a greater ability to digest dietary fiber, which may be due to differences in intestinal microbiota. In this study, we fed Ding'an and DLY pigs high and low levels of dietary fiber, respectively, to investigate factors contributing to high dietary fiber adaption in Ding'an pigs. Twelve Ding'an pigs and DLY pigs were randomly divided into a 2 (diet) × 2 (breed) factorial experiment (n = 3). Compared with commercial pigs, Ding'an pigs have a stronger ability to digest dietary fiber. Prevotella was more prevalent in Ding'an pigs than in DLY pigs, which may be an important reason for the stronger ability of fiber degradation in Ding'an pigs. When the effects of feed and breed factors are considered, differences in abundance of 31 species and 14 species, respectively, may result in a greater ability of fiber degradation in Ding'an pigs. Among them, Prevotella. sp. CAG:520 may be a newly discovered bacterium related to fiber degradation, which positively correlated with many fiber-degrading bacteria (r > 0.7). We also found that the concentration of plant metabolites with anti-inflammatory and antioxidant effects was higher in the colonic chyme of Ding'an pigs after increasing the fiber content, which resulted in the downregulated expression of inflammatory factors in colonic mucosa. Spearman's correlation coefficient revealed a strong correlation between microbiota and the apparent digestibility of dietary fiber (r > 0.7). The mRNA expressions of SLC16A1, PYY, and GCG were significantly increased in the colonic mucosa of Ding'an pigs fed on high-fiber diets, which indicates that Ding'an pigs have an enhanced absorption of SCFAs. Our results suggested that an appropriate increase in dietary fiber content can reduce the inflammatory response and improve feed efficiency in Ding'an pigs, and differences in the intestinal microbial composition may be an important reason for the difference in the fiber degradation capacity between the two breeds of pigs.
Collapse
Affiliation(s)
- Pengxiang Xue
- National Engineering Laboratory for Animal Breeding, MOA Laboratory of Animal Genetics and Breeding, Department of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; (P.X.); (M.X.); (Y.L.); (Q.T.)
| | - Mingming Xue
- National Engineering Laboratory for Animal Breeding, MOA Laboratory of Animal Genetics and Breeding, Department of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; (P.X.); (M.X.); (Y.L.); (Q.T.)
| | - Yabiao Luo
- National Engineering Laboratory for Animal Breeding, MOA Laboratory of Animal Genetics and Breeding, Department of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; (P.X.); (M.X.); (Y.L.); (Q.T.)
| | - Qiguo Tang
- National Engineering Laboratory for Animal Breeding, MOA Laboratory of Animal Genetics and Breeding, Department of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; (P.X.); (M.X.); (Y.L.); (Q.T.)
| | - Feng Wang
- Institute of Animal Science and Veterinary Medicine, Hainan Academy of Agricultural Science, Haikou 571101, China; (F.W.); (R.S.); (Z.C.)
| | - Ruiping Sun
- Institute of Animal Science and Veterinary Medicine, Hainan Academy of Agricultural Science, Haikou 571101, China; (F.W.); (R.S.); (Z.C.)
| | - Yanxia Song
- Sanya Institute, China Agricultural University, Sanya 572024, China;
| | - Zhe Chao
- Institute of Animal Science and Veterinary Medicine, Hainan Academy of Agricultural Science, Haikou 571101, China; (F.W.); (R.S.); (Z.C.)
| | - Meiying Fang
- National Engineering Laboratory for Animal Breeding, MOA Laboratory of Animal Genetics and Breeding, Department of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; (P.X.); (M.X.); (Y.L.); (Q.T.)
- Sanya Institute, China Agricultural University, Sanya 572024, China;
| |
Collapse
|
12
|
Oliveira HGDS, Sousa AIDJ, Zanon IP, Lopes CTDA, Silva ROS, Domingues SFS, Salvarani FM. Occurrence of Clostridium perfringens in Wild Mammals in the Amazon Biome. Animals (Basel) 2024; 14:1333. [PMID: 38731335 PMCID: PMC11083269 DOI: 10.3390/ani14091333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 04/18/2024] [Accepted: 04/18/2024] [Indexed: 05/13/2024] Open
Abstract
The objective of this study was to evaluate the occurrence of Clostridium perfringens in stool samples and swabs collected from wild mammals in the Amazon biome. Sixty-five faecal and swab samples were collected in situ and ex situ from 16 species and three genera of wild mammals, some of which were in good health and some of which had diarrhoea. After pre-enrichment, the samples were plated on selective agar for C. perfringens. Characteristic colonies were subjected to multiplex PCR for the detection of genes encoding the main C. perfringens toxins (alpha, beta, epsilon, and iota toxin and enterotoxin). Among the 65 samples, 40 (61.5%) were positive for the gene encoding the alpha toxin and were classified as type A, 36 of which were asymptomatic animals and four were diarrheal. No other toxinotypes were found. The findings of this study suggest that C. perfringens type A is commonly found in mammal species of the Amazon biome. This seems to be the first study to identify C. perfringens type A in species such as B. variegatus (common ground sloth), C. didactylus (two-toed sloth), P. flavus (Jupará), T. tetradactyla (anteater), S. collinsi (squirrel monkey), S. niger (black marmoset), and S. apella (Guyana capuchin) and in the genus Didelphis sp. (opossum).
Collapse
Affiliation(s)
- Hanna Gabriela da Silva Oliveira
- Instituto de Medicina Veterinária, Universidade Federal do Pará, Castanhal 68740-970, PA, Brazil; (H.G.d.S.O.); (A.I.d.J.S.); (C.T.d.A.L.); (S.F.S.D.)
| | - Ananda Iara de Jesus Sousa
- Instituto de Medicina Veterinária, Universidade Federal do Pará, Castanhal 68740-970, PA, Brazil; (H.G.d.S.O.); (A.I.d.J.S.); (C.T.d.A.L.); (S.F.S.D.)
| | - Isabela Paduá Zanon
- Laboratório de Bacterioses e Pesquisa da Escola de Veterinária, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, MG, Brazil; (I.P.Z.); (R.O.S.S.)
| | - Cinthia Távora de Albuquerque Lopes
- Instituto de Medicina Veterinária, Universidade Federal do Pará, Castanhal 68740-970, PA, Brazil; (H.G.d.S.O.); (A.I.d.J.S.); (C.T.d.A.L.); (S.F.S.D.)
| | - Rodrigo Otavio Silveira Silva
- Laboratório de Bacterioses e Pesquisa da Escola de Veterinária, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, MG, Brazil; (I.P.Z.); (R.O.S.S.)
| | - Sheyla Farhayldes Souza Domingues
- Instituto de Medicina Veterinária, Universidade Federal do Pará, Castanhal 68740-970, PA, Brazil; (H.G.d.S.O.); (A.I.d.J.S.); (C.T.d.A.L.); (S.F.S.D.)
| | - Felipe Masiero Salvarani
- Instituto de Medicina Veterinária, Universidade Federal do Pará, Castanhal 68740-970, PA, Brazil; (H.G.d.S.O.); (A.I.d.J.S.); (C.T.d.A.L.); (S.F.S.D.)
| |
Collapse
|
13
|
Popoff MR. Overview of Bacterial Protein Toxins from Pathogenic Bacteria: Mode of Action and Insights into Evolution. Toxins (Basel) 2024; 16:182. [PMID: 38668607 PMCID: PMC11054074 DOI: 10.3390/toxins16040182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 03/29/2024] [Accepted: 03/30/2024] [Indexed: 04/29/2024] Open
Abstract
Bacterial protein toxins are secreted by certain bacteria and are responsible for mild to severe diseases in humans and animals. They are among the most potent molecules known, which are active at very low concentrations. Bacterial protein toxins exhibit a wide diversity based on size, structure, and mode of action. Upon recognition of a cell surface receptor (protein, glycoprotein, and glycolipid), they are active either at the cell surface (signal transduction, membrane damage by pore formation, or hydrolysis of membrane compound(s)) or intracellularly. Various bacterial protein toxins have the ability to enter cells, most often using an endocytosis mechanism, and to deliver the effector domain into the cytosol, where it interacts with an intracellular target(s). According to the nature of the intracellular target(s) and type of modification, various cellular effects are induced (cell death, homeostasis modification, cytoskeleton alteration, blockade of exocytosis, etc.). The various modes of action of bacterial protein toxins are illustrated with representative examples. Insights in toxin evolution are discussed.
Collapse
Affiliation(s)
- Michel R Popoff
- Unité des Toxines Bactériennes, Institut Pasteur, Université Paris Cité, CNRS UMR 2001 INSERM U1306, F-75015 Paris, France
| |
Collapse
|
14
|
Kandpal M, Varshney N, Rawal KS, Jha HC. Gut dysbiosis and neurological modalities: An engineering approach via proteomic analysis of gut-brain axis. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2024; 140:199-248. [PMID: 38762270 DOI: 10.1016/bs.apcsb.2024.02.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2024]
Abstract
The human gut microbiota is a complex and dynamic community of microorganisms, that influence metabolic, neurodevelopmental, and immune pathways. Microbial dysbiosis, characterized by changes in microbial diversity and relative abundances, is implicated in the development of various chronic neurological and neurodegenerative disorders. These disorders are marked by the accumulation of pathological protein aggregates, leading to the progressive loss of neurons and behavioural functions. Dysregulations in protein-protein interaction networks and signalling complexes, critical for normal brain function, are common in neurological disorders but challenging to unravel, particularly at the neuron and synapse-specific levels. To advance therapeutic strategies, a deeper understanding of neuropathogenesis, especially during the progressive disease phase, is needed. Biomarkers play a crucial role in identifying disease pathophysiology and monitoring disease progression. Proteomics, a powerful technology, shows promise in accelerating biomarker discovery and aiding in the development of novel treatments. In this chapter, we provide an in-depth overview of how proteomic techniques, utilizing various biofluid samples from patients with neurological conditions and diverse animal models, have contributed valuable insights into the pathogenesis of numerous neurological disorders. We also discuss the current state of research, potential challenges, and future directions in proteomic approaches to unravel neuro-pathological conditions.
Collapse
Affiliation(s)
- Meenakshi Kandpal
- Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Indore, India
| | - Nidhi Varshney
- Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Indore, India
| | - Kunal Sameer Rawal
- School of Biosciences and Technology, Vellore Institute of Technology, Vellore, India
| | - Hem Chandra Jha
- Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Indore, India; Centre for Rural Development & Technology, IIT Indore, Indore, India.
| |
Collapse
|
15
|
Chang B, Zhang W, Wang Y, Zhang Y, Zhong S, Gao P, Wang L, Zhao Z. Uncovering the complexity of childhood undernutrition through strain-level analysis of the gut microbiome. BMC Microbiol 2024; 24:73. [PMID: 38443783 PMCID: PMC10916198 DOI: 10.1186/s12866-024-03211-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 01/31/2024] [Indexed: 03/07/2024] Open
Abstract
BACKGROUND Undernutrition (UN) is a critical public health issue that threatens the lives of children under five in developing countries. While evidence indicates the crucial role of the gut microbiome (GM) in UN pathogenesis, the strain-level inspection and bacterial co-occurrence network investigation in the GM of UN children are lacking. RESULTS This study examines the strain compositions of the GM in 61 undernutrition patients (UN group) and 36 healthy children (HC group) and explores the topological features of GM co-occurrence networks using a complex network strategy. The strain-level annotation reveals that the differentially enriched species between the UN and HC groups are due to discriminated strain compositions. For example, Prevotella copri is mainly composed of P. copri ASM1680343v1 and P. copri ASM345920v1 in the HC group, but it is composed of P. copri ASM346549v1 and P. copri ASM347465v1 in the UN group. In addition, the UN-risk model constructed at the strain level demonstrates higher accuracy (AUC = 0.810) than that at the species level (AUC = 0.743). With complex network analysis, we further discovered that the UN group had a more complex GM co-occurrence network, with more hub bacteria and a higher clustering coefficient but lower information transfer efficiencies. Moreover, the results at the strain level suggested the inaccurate and even false conclusions obtained from species level analysis. CONCLUSIONS Overall, this study highlights the importance of examining the GM at the strain level and investigating bacterial co-occurrence networks to advance our knowledge of UN pathogenesis.
Collapse
Affiliation(s)
- Bingmei Chang
- Department of Biochemistry and Molecular Biology, Shanxi Medical University, Taiyuan, People's Republic of China
| | - Wenjie Zhang
- Department of Anesthesiology, First Hospital of Shanxi Medical University, Taiyuan, People's Republic of China
| | - Yinan Wang
- Peking University Shenzhen Hospital, Shenzhen, People's Republic of China
| | - Yuanzheng Zhang
- Shenzhen Byoryn Technology Co., Ltd, Shenzhen, People's Republic of China
| | - Shilin Zhong
- Peking University Shenzhen Hospital, Shenzhen, People's Republic of China
| | - Peng Gao
- BGI-Shenzhen, Beishan Industrial Zone, Shenzhen, People's Republic of China.
| | - Lili Wang
- Department of Anesthesiology, First Hospital of Shanxi Medical University, Taiyuan, People's Republic of China.
| | - Zicheng Zhao
- Shenzhen Byoryn Technology Co., Ltd, Shenzhen, People's Republic of China.
| |
Collapse
|
16
|
Wu X, Li Y, Shang Y, Wang W, Wu L, Han L, Wang Q, Wang Z, Xu H, Liu W. Application of two-dimensional polymerase chain reaction to detect four types of microorganisms in feces for assisted diagnosis of IBD. Clin Chim Acta 2024; 555:117802. [PMID: 38281660 DOI: 10.1016/j.cca.2024.117802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 01/20/2024] [Accepted: 01/22/2024] [Indexed: 01/30/2024]
Abstract
BACKGROUND The incidence of inflammatory bowel disease (IBD) continues to increase annually, accounting for about 6.8 million cases in 2017 worldwide. However, there is currently no gold standard for the diagnosis of IBD. METHODS A method for the detection of four microorganisms in feces by two-dimensional polymerase chain reaction (2D-PCR) has been developed. Plasmids were used to validate the sensitivity and specificity of the method. Clinical samples were tested using a 2D-PCR method. Optimal diagnostic thresholds for IBD were determined based on ROC results. RESULTS Of the 112 samples, 78 were from IBD patients and 34 from patients with other gastrointestinal (GI) diseases. Thomasclavelia ramosum and univ907-1062 positivity are necessary, and two or more positives of the three bacteria (Thomasclavelia spiroforme, Thomasclavelia saccharogumia or Clostridium cluster XVIII) are the optimal diagnostic thresholds for IBD. The area under the curve was 0.826 with a 95% confidence interval of 0.735-0.981 and a p-value of 0.000, corresponding to a sensitivity of 0.769 and a specificity of 0.853. CONCLUSIONS Based on the detection results of microorganisms, IBD and GI can be effectively distinguished. The detection of four microorganisms in feces can assist clinicians in the differential diagnosis of IBD. Our experiment aims to provide a better program for early clinical diagnosis and regular dynamic monitoring of IBD.
Collapse
Affiliation(s)
- Xuan Wu
- Department of Laboratory Medicine, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China; Central Laboratory and Department of Laboratory Medicine, Shanghai Tenth People's Hospital, Tongji University, Shanghai 200070, China
| | - Yueying Li
- Department of Laboratory Medicine, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China; Central Laboratory and Department of Laboratory Medicine, Shanghai Tenth People's Hospital, Tongji University, Shanghai 200070, China
| | - Yuanjiang Shang
- Central Laboratory and Department of Laboratory Medicine, Shanghai Tenth People's Hospital, Tongji University, Shanghai 200070, China
| | - Weifeng Wang
- Department of Laboratory Medicine, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Lixia Wu
- Central Laboratory and Department of Laboratory Medicine, Shanghai Tenth People's Hospital, Tongji University, Shanghai 200070, China
| | - Lin Han
- Central Laboratory and Department of Laboratory Medicine, Shanghai Tenth People's Hospital, Tongji University, Shanghai 200070, China
| | - Qiong Wang
- Jiangsu Province Hospital of Chinese Medicine, Nanjing, Jiangsu 210029, China; Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210022, China.
| | - Zhujian Wang
- Clinical Laboratory, Eye & ENT Hospital, Shanghai Medical College, Fudan University, Shanghai 200031, China.
| | - Hanchen Xu
- Institute of Digestive Diseases, Longhua Hospital, China-Canada Center of Research for Digestive Diseases (ccCRDD), Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China.
| | - Weiwei Liu
- Department of Laboratory Medicine, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China.
| |
Collapse
|
17
|
Ibrahim GA, Altammar KA. Moringa oleifera as a potential antimicrobial against pathogenic Clostridium perfringens isolates in farm animals. Open Vet J 2024; 14:242-255. [PMID: 38633192 PMCID: PMC11018413 DOI: 10.5455/ovj.2024.v14.i1.21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2023] [Accepted: 12/15/2023] [Indexed: 04/19/2024] Open
Abstract
Background Clostridium perfringens (CP) is an emerging anaerobic pathogen that can aggravate severe fatal infections in different hosts and livestock. Aim This paper was designed to monitor the antibacterial efficacy of Moringa oleifera (M. oleifera) plant against different CP isolates of variant toxin genotypes comparing that with commercial antibiotics in the veterinary field. Methods A total of 200 examined fecal, intestinal, and liver samples from cattle, sheep, and goats were investigated bacteriologically and biochemically for CP. Then, the isolates were examined by polymerase chain reaction (PCR) for toxin gene typing. Thereafter, the antimicrobial susceptibility testing as well as the antibacterial efficacy of M. oleifera were evaluated and statistically analyzed against recovered isolates. Results The prevalence rate of CP was 51% (102/200); of which 54.5% was from cattle, 50% from sheep, and 40% from goat. Moreover, all CP isolates were highly resistant to tetracycline and lincomycin drugs; meanwhile, they were of the least resistance against ciprofloxacin (8.3%-16.7%), cefotaxime (16.7%-25%), and gentamycin (26.7%-33.3%). For M. oleifera, high antibacterial efficacy with greater inhibition zones of the plant was recorded with its oil (20-24 mm) and ethanolic extracts (16-20 mm) against CP than the aqueous extract (≤ 10 mm). A good correlation was stated between M. oleifera oil and toxin type of CP isolates particularly type A followed by D and B types. Interestingly, the oil and ethanolic extracts of M. oleifera gave higher antibacterial efficacy than most commercial antibiotics against the recovered isolates. Conclusion This study highlighted the potent antibacterial properties of M. oleifera for suppressing CP isolated from farm animals; hence, more investigations on M. oleifera are suggested to support its use as a medical herbal plant substituting antibiotics hazards and resistance problems worldwide.
Collapse
Affiliation(s)
- Ghada A. Ibrahim
- Agriculture Research Center (ARC), Animal Health Research Institute (AHRI), Bacteriology Department, Ismailia Branch, Egypt
| | - Khadijah A. Altammar
- Department of Biology, College of Science, University of Hafr Al Batin, Hafr Al Batin, Saudi Arabia
| |
Collapse
|
18
|
Chandrasekaran P, Han Y, Zerbe CS, Heller T, DeRavin SS, Kreuzberg SA, Marciano BE, Siu Y, Jones DR, Abraham RS, Stephens MC, Tsou AM, Snapper S, Conlan S, Subramanian P, Quinones M, Grou C, Calderon V, Deming C, Leiding JW, Arnold DE, Logan BR, Griffith LM, Petrovic A, Mousallem TI, Kapoor N, Heimall JR, Barnum JL, Kapadia M, Wright N, Rayes A, Chandra S, Broglie LA, Chellapandian D, Deal CL, Grunebaum E, Lim SS, Mallhi K, Marsh RA, Murguia-Favela L, Parikh S, Touzot F, Cowan MJ, Dvorak CC, Haddad E, Kohn DB, Notarangelo LD, Pai SY, Puck JM, Pulsipher MA, Torgerson TR, Kang EM, Malech HL, Segre JA, Bryant CE, Holland SM, Falcone EL. Intestinal microbiome and metabolome signatures in patients with chronic granulomatous disease. J Allergy Clin Immunol 2023; 152:1619-1633.e11. [PMID: 37659505 PMCID: PMC11279821 DOI: 10.1016/j.jaci.2023.07.022] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Revised: 07/18/2023] [Accepted: 07/27/2023] [Indexed: 09/04/2023]
Abstract
BACKGROUND Chronic granulomatous disease (CGD) is caused by defects in any 1 of the 6 subunits forming the nicotinamide adenine dinucleotide phosphate oxidase complex 2 (NOX2), leading to severely reduced or absent phagocyte-derived reactive oxygen species production. Almost 50% of patients with CGD have inflammatory bowel disease (CGD-IBD). While conventional IBD therapies can treat CGD-IBD, their benefits must be weighed against the risk of infection. Understanding the impact of NOX2 defects on the intestinal microbiota may lead to the identification of novel CGD-IBD treatments. OBJECTIVE We sought to identify microbiome and metabolome signatures that can distinguish individuals with CGD and CGD-IBD. METHODS We conducted a cross-sectional observational study of 79 patients with CGD, 8 pathogenic variant carriers, and 19 healthy controls followed at the National Institutes of Health Clinical Center. We profiled the intestinal microbiome (amplicon sequencing) and stool metabolome, and validated our findings in a second cohort of 36 patients with CGD recruited through the Primary Immune Deficiency Treatment Consortium. RESULTS We identified distinct intestinal microbiome and metabolome profiles in patients with CGD compared to healthy individuals. We observed enrichment for Erysipelatoclostridium spp, Sellimonas spp, and Lachnoclostridium spp in CGD stool samples. Despite differences in bacterial alpha and beta diversity between the 2 cohorts, several taxa correlated significantly between both cohorts. We further demonstrated that patients with CGD-IBD have a distinct microbiome and metabolome profile compared to patients without CGD-IBD. CONCLUSION Intestinal microbiome and metabolome signatures distinguished patients with CGD and CGD-IBD, and identified potential biomarkers and therapeutic targets.
Collapse
Affiliation(s)
| | - Yu Han
- Division of Molecular Genetics and Pathology, Center for Devices and Radiological Health, Food and Drug Administration, Silver Spring, Md; Laboratory of Clinical Immunology and Microbiology (LCIM), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, Md
| | - Christa S Zerbe
- Laboratory of Clinical Immunology and Microbiology (LCIM), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, Md
| | - Theo Heller
- Translational Hepatology Section, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), NIH, Bethesda, Md
| | - Suk See DeRavin
- Laboratory of Clinical Immunology and Microbiology (LCIM), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, Md
| | - Samantha A Kreuzberg
- Laboratory of Clinical Immunology and Microbiology (LCIM), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, Md
| | - Beatriz E Marciano
- Laboratory of Clinical Immunology and Microbiology (LCIM), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, Md
| | - Yik Siu
- Department of Biochemistry and Molecular Pharmacology, New York University Langone Health, New York, NY
| | - Drew R Jones
- Department of Biochemistry and Molecular Pharmacology, New York University Langone Health, New York, NY
| | - Roshini S Abraham
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minn; Department of Pathology and Laboratory Medicine, Nationwide Children's Hospital, Columbus, Ohio
| | | | - Amy M Tsou
- Division of Gastroenterology, Hepatology and Nutrition, Boston Children's Hospital, and Department of Pediatrics, Harvard Medical School, Boston, Mass; Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medical College, New York, NY
| | - Scott Snapper
- Division of Gastroenterology, Hepatology and Nutrition, Boston Children's Hospital, and Department of Pediatrics, Harvard Medical School, Boston, Mass
| | - Sean Conlan
- National Human Genome Research Institute (NHGRI), NIH, Bethesda, Md
| | - Poorani Subramanian
- Bioinformatics and Computational Biosciences Branch, Office of Cyber Infrastructure and Computational Biology, NIAID, NIH, Bethesda, Md
| | - Mariam Quinones
- Bioinformatics and Computational Biosciences Branch, Office of Cyber Infrastructure and Computational Biology, NIAID, NIH, Bethesda, Md
| | - Caroline Grou
- Bioinformatics Core, Montreal Clinical Research Institute (IRCM), Montreal, Quebec, Canada
| | - Virginie Calderon
- Bioinformatics Core, Montreal Clinical Research Institute (IRCM), Montreal, Quebec, Canada
| | - Clayton Deming
- National Human Genome Research Institute (NHGRI), NIH, Bethesda, Md
| | - Jennifer W Leiding
- Division of Allergy and Immunology, Department of Pediatrics, Johns Hopkins University, Baltimore, Md
| | - Danielle E Arnold
- Immune Deficiency-Cellular Therapy Program, Center for Cancer Research, National Cancer Institute (NCI), NIH, Bethesda, Md
| | - Brent R Logan
- Division of Biostatistics, Institute for Health and Equity, Medical College of Wisconsin, Milwaukee, Wis
| | - Linda M Griffith
- Division of Allergy, Immunology, and Transplantation, NIAID, NIH, Bethesda, Md
| | - Aleksandra Petrovic
- Department of Pediatrics, University of Washington School of Medicine and Seattle Children's Hospital and Research Center, Seattle, Wash
| | - Talal I Mousallem
- Department of Pediatrics, Duke University Medical Center, Durham, NC
| | - Neena Kapoor
- Division of Hematology, Oncology, and Blood and Marrow Transplantation, Children's Hospital Los Angeles, Los Angeles, Calif
| | - Jennifer R Heimall
- Division of Allergy and Immunology, Children's Hospital of Philadelphia and Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pa
| | - Jessie L Barnum
- Division of Blood and Marrow Transplantation and Cellular Therapies, University of Pittsburgh Medical Center (UPMC) and Children's Hospital of Pittsburgh, Pittsburgh, Pa
| | - Malika Kapadia
- Department of Pediatrics, Harvard University Medical School, Boston, Mass
| | - Nicola Wright
- Section of Hematology/Immunology, Alberta Children's Hospital, University of Calgary, Calgary, Alberta, Canada
| | - Ahmad Rayes
- Intermountain Primary Children's Hospital, University of Utah, Salt Lake City, Utah
| | - Sharat Chandra
- Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children's Hospital Medical Center, and Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Larisa A Broglie
- Division of Pediatric Hematology-Oncology, Department of Pediatrics, Medical College of Wisconsin, Milwaukee, Wis
| | - Deepak Chellapandian
- Center for Cell and Gene Therapy for Non-Malignant Conditions, Johns Hopkins All Children's Hospital, St Petersburg, Fla
| | - Christin L Deal
- Division of Allergy and Immunology, UPMC, Children's Hospital of Pittsburgh, Pittsburgh, Pa
| | - Eyal Grunebaum
- Division of Immunology and Allergy, Department of Pediatrics, Hospital for Sick Children, Toronto, Ontario, Canada; Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Stephanie Si Lim
- Department of Pediatrics, John A. Burns School of Medicine, University of Hawai'i at Mānoa, Honolulu, Hawaii; University of Hawai'i Cancer Center, University of Hawai'i at Mānoa, Honolulu, Hawaii
| | | | - Rebecca A Marsh
- Cincinnati Children's Hospital Medical Center, and University of Cincinnati, Cincinnati, Ohio
| | - Luis Murguia-Favela
- Section of Hematology/Immunology, Alberta Children's Hospital, University of Calgary, Calgary, Alberta, Canada
| | - Suhag Parikh
- Department of Pediatrics, Emory University School of Medicine, Atlanta, Ga
| | - Fabien Touzot
- Department of Pediatrics, CHU Sainte-Justine, Université de Montréal, Montreal, Quebec, Canada; Department of Microbiology, Infectious Diseases, and Immunology, Université de Montréal, Montreal, Quebec, Canada
| | - Morton J Cowan
- University of California San Francisco Benioff Children's Hospital, San Francisco, Calif
| | - Christopher C Dvorak
- University of California San Francisco Benioff Children's Hospital, San Francisco, Calif
| | - Elie Haddad
- Department of Pediatrics, CHU Sainte-Justine, Université de Montréal, Montreal, Quebec, Canada; Department of Microbiology, Infectious Diseases, and Immunology, Université de Montréal, Montreal, Quebec, Canada
| | - Donald B Kohn
- Microbiology, Immunology, & Molecular Genetics, University of California, Los Angeles, Calif
| | - Luigi D Notarangelo
- Laboratory of Clinical Immunology and Microbiology (LCIM), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, Md
| | - Sung-Yun Pai
- Immune Deficiency-Cellular Therapy Program, Center for Cancer Research, National Cancer Institute (NCI), NIH, Bethesda, Md
| | - Jennifer M Puck
- University of California San Francisco Benioff Children's Hospital, San Francisco, Calif
| | - Michael A Pulsipher
- Division of Pediatric Hematology and Oncology, Intermountain Primary Children's Hospital, Huntsman Cancer Institute at the University of Utah Spencer Fox Eccles School of Medicine, Salt Lake City, Utah
| | | | - Elizabeth M Kang
- Laboratory of Clinical Immunology and Microbiology (LCIM), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, Md
| | - Harry L Malech
- Laboratory of Clinical Immunology and Microbiology (LCIM), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, Md
| | - Julia A Segre
- National Human Genome Research Institute (NHGRI), NIH, Bethesda, Md
| | - Clare E Bryant
- Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Steven M Holland
- Laboratory of Clinical Immunology and Microbiology (LCIM), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, Md
| | - Emilia Liana Falcone
- Laboratory of Clinical Immunology and Microbiology (LCIM), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, Md; Department of Microbiology, Infectious Diseases, and Immunology, Université de Montréal, Montreal, Quebec, Canada; Center for Immunity, Inflammation and Infectious Diseases, IRCM, Montreal, Quebec, Canada; Department of Medicine, Université de Montréal, Montreal, Quebec, Canada.
| |
Collapse
|
19
|
Du Z, Yang F, Fang J, Yamasaki S, Oya T, Nguluve D, Kumagai H, Cai Y. Silage preparation and sustainable livestock production of natural woody plant. FRONTIERS IN PLANT SCIENCE 2023; 14:1253178. [PMID: 37746011 PMCID: PMC10514673 DOI: 10.3389/fpls.2023.1253178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Accepted: 08/23/2023] [Indexed: 09/26/2023]
Abstract
As the global population increases and the economy grows rapidly, the demand for livestock products such as meat, egg and milk continue to increase. The shortage of feed in livestock production is a worldwide problem restricting the development of the animal industry. Natural woody plants are widely distributed and have a huge biomass yield. The fresh leaves and branches of some woody plants are rich in nutrients such as proteins, amino acids, vitamins and minerals and can be used to produce storage feed such as silage for livestock. Therefore, the development and utilization of natural woody plants for clean fermented feed is important for the sustainable production of livestock product. This paper presents a comprehensive review of the research progress, current status and development prospects of forageable natural woody plant feed resources. The nutritional composition and uses of natural woody plants, the main factors affecting the fermentation of woody plant silage and the interaction mechanism between microbial co-occurrence network and secondary metabolite are reviewed. Various preparation technologies for clean fermentation of woody plant silage were summarized comprehensively, which provided a sustainable production mode for improving the production efficiency of livestock and producing high-quality livestock product. Therefore, woody plants play an increasingly important role as a potential natural feed resource in alleviating feed shortage and promoting sustainable development of livestock product.
Collapse
Affiliation(s)
- Zhumei Du
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China
- Crop, Livestock, and Environment Division, Japan International Research Center for Agricultural Sciences (JIRCAS), Tsukuba, Ibaraki, Japan
| | - Fuyu Yang
- College of Animal Science, Guizhou University, Guiyang, China
| | - Jiachen Fang
- Faculty of Agriculture and Life Science, Hirosaki University, Hirosaki, Japan
| | - Seishi Yamasaki
- Crop, Livestock, and Environment Division, Japan International Research Center for Agricultural Sciences (JIRCAS), Tsukuba, Ibaraki, Japan
| | - Tetsuji Oya
- Crop, Livestock, and Environment Division, Japan International Research Center for Agricultural Sciences (JIRCAS), Tsukuba, Ibaraki, Japan
| | - Damiao Nguluve
- Animal Science Directorate, Agricultural Research Institute of Mozambique, Matola, Mozambique
| | - Hajime Kumagai
- Graduate School of Agriculture, Kyoto University, Kyoto, Japan
| | - Yimin Cai
- Crop, Livestock, and Environment Division, Japan International Research Center for Agricultural Sciences (JIRCAS), Tsukuba, Ibaraki, Japan
| |
Collapse
|
20
|
Zhang P, Chen J, Ming Y, Niu Y. Probiotics treatment ameliorated mycophenolic acid-induced colitis by enhancing intestinal barrier function and improving intestinal microbiota dysbiosis in mice. Front Microbiol 2023; 14:1153188. [PMID: 37533828 PMCID: PMC10390739 DOI: 10.3389/fmicb.2023.1153188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Accepted: 07/04/2023] [Indexed: 08/04/2023] Open
Abstract
Background Mycophenolic acid (MPA)-induced colitis was still a severe side effect and challenge faced by solid transplant recipients. We aimed to explore the function and mechanism of probiotics in the treatment of MPA-induced colitis. Methods In this study, 15 mice (C57BL/6) were randomly assigned into three groups: control (CNTL) group (n = 5), MPA group (n = 5) and the MPA + Probiotic group (n = 5). Bifid Triple Viable capsules, which were drugs for clinical use and consisted of Bifidobacterium longum, Lactobacillus acidophilus, and Enterococcus faecalis, were used in Probiotic group. Body weight change, stool scores, colon histopathology and morphology were used to evaluate the disease severity. The intestinal mucosal barrier function was assessed by measuring the expression level of secretory immunoglobulin A (sIgA), Zonula occludens-1 (ZO-1) and Occludin. Finally, 16S rDNA sequencing and bioinformatics analysis were performed on mice feces to compare the different intestinal microbial composition and diversity among three groups. Results Compared with the CNTL group, the mice in MPA group showed a significantly decreased body weight, increased stool scores, shortened colon length and severe colon inflammation. However, probiotics treated MPA mice reversed the disease severity, indicating that probiotics ameliorated MPA-induced colitis in mice. Mechanistically, probiotics improved the intestinal barrier function by up-regulating the expression of sIgA, ZO-1 and Occludin. Moreover, MPA-induced colitis led to intestinal microbiota dysbiosis, including the change of Firmicutes/Bacteroidetes ratio, α- and β-diversity. But probiotic treated group showed mild change in these microbial features. Additionally, we found that Clostridiales was the most significantly different microbiota flora in MPA group. Conclusion Probiotics treatment ameliorated MPA-induced colitis by enhancing intestinal barrier function and improving intestinal microbiota dysbiosis. Clostridiales might be the dominant functional intestinal microflora and serve as the potential therapy target in MPA-induced colitis.
Collapse
Affiliation(s)
- Pengpeng Zhang
- Organ Transplantation Center, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China
- Engineering and Technology Research Center for Transplantation Medicine of the National Ministry of Health, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Jinwen Chen
- Organ Transplantation Center, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China
- Engineering and Technology Research Center for Transplantation Medicine of the National Ministry of Health, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Yingzi Ming
- Organ Transplantation Center, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China
- Engineering and Technology Research Center for Transplantation Medicine of the National Ministry of Health, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Ying Niu
- Organ Transplantation Center, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China
- Engineering and Technology Research Center for Transplantation Medicine of the National Ministry of Health, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China
| |
Collapse
|
21
|
Almuzaini AM. Phytochemicals: potential alternative strategy to fight Salmonella enterica serovar Typhimurium. Front Vet Sci 2023; 10:1188752. [PMID: 37261108 PMCID: PMC10228746 DOI: 10.3389/fvets.2023.1188752] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Accepted: 04/20/2023] [Indexed: 06/02/2023] Open
Abstract
The rise of multidrug resistant (MDR) microorganisms is a great hazard worldwide and has made it difficult to treat many infectious diseases adequately. One of the most prevalent causes of outbreaks of foodborne illness worldwide is Salmonella. The ability of this and other harmful bacteria to withstand antibiotics has recently proven crucial to their effective control. Since the beginning of time, herbal medicines and phytochemicals have been employed for their potent antibacterial action and there is a growing trend toward the production of plant based natural products for the prevention and treatment of pathogenic infections. Numerous phytochemicals have been proven effective against the molecular determinants responsible for attaining drug resistance in pathogens like efflux pumps, membrane proteins, bacterial cell communications and biofilms. The medicinal plants having antibacterial activity and antibiotics combination with phytochemicals have shown synergetic activity against Salmonella enterica serovar Typhimurium. The inhibitory effects of tannins on rumen proteolytic bacteria can be exploited in ruminant nutrition. Improved control of the rumen ecology and practical use of this feed additive technology in livestock production will be made possible by a better knowledge of the modulatory effects of phytochemicals on the rumen microbial populations in combination with fermentation. This review focuses on the development of antibacterial resistance in Salmonella, the mechanism of action of phytochemicals and the use of phytochemicals against S. enterica serovar Typhimurium. The advances and potential future applications of phytochemicals in the fight against resistant are also discussed.
Collapse
|
22
|
Bassotti G, Fruganti A, Stracci F, Marconi P, Fettucciari K. Cytotoxic synergism of Clostridioides difficile toxin B with proinflammatory cytokines in subjects with inflammatory bowel diseases. World J Gastroenterol 2023; 29:582-596. [PMID: 36742168 PMCID: PMC9896618 DOI: 10.3748/wjg.v29.i4.582] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 12/03/2022] [Accepted: 12/27/2022] [Indexed: 01/20/2023] Open
Abstract
Clostridioides difficile (C. difficile) is progressively colonizing humans and animals living with humans. During this process, hypervirulent strains and mutated toxin A and B of C. difficile (TcdA and TcdB) are originating and developing. While in healthy subjects colonization by C. difficile becomes a risk after the use of antibiotics that alter the microbiome, other categories of people are more susceptible to infection and at risk of relapse, such as those with inflammatory bowel disease (IBD). Recent in vitro studies suggest that this increased susceptibility could be due to the strong cytotoxic synergism between TcdB and proinflammatory cytokines the tumor necrosis factor-alpha and interferon-gamma (CKs). Therefore, in subjects with IBD the presence of an inflammatory state in the colon could be the driver that increases the susceptibility to C. difficile infection and its progression and relapses. TcdB is internalized in the cell via three receptors: chondroitin sulphate proteoglycan 4; poliovirus receptor-like 3; and Wnt receptor frizzled family. Chondroitin sulphate proteoglycan 4 and Wnt receptor frizzled family are involved in cell death by apoptosis or necrosis depending on the concentration of TcdB and cell types, while poliovirus receptor-like 3 induces only necrosis. It is possible that cytokines could also induce a greater expression of receptors for TcdB that are more involved in necrosis than in apoptosis. Therefore, in subjects with IBD there are the conditions: (1) For greater susceptibility to C. difficile infection, such as the inflammatory state, and abnormalities of the microbiome and of the immune system; (2) for the enhancement of the cytotoxic activity of TcdB +Cks; and (3) for a greater expression of TcdB receptors stimulated by cytokines that induce cell death by necrosis rather than apoptosis. The only therapeutic approach currently possible in IBD patients is monitoring of C. difficile colonization for interventions aimed at reducing tumor necrosis factor-alpha and interferon-gamma levels when the infection begins. The future perspective is to generate bacteriophages against C. difficile for targeted therapy.
Collapse
Affiliation(s)
- Gabrio Bassotti
- Department of Medicine and Surgery, Gastroenterology, Hepatology & Digestive Endoscopy Section University of Perugia Medical School, Piazza Lucio Severi, Perugia 06132, Italy, and Santa Maria della Misericordia Hospital, Gastroenterology & Hepatology Unit Perugia 06156, Italy
| | - Alessandro Fruganti
- School of Biosciences and Veterinary Medicine, University of Camerino, Matelica 62024, Italy
| | - Fabrizio Stracci
- Medicine and Surgery, Hygiene and Public Health Section, University of Perugia, Perugia 06123, Italy
| | - Pierfrancesco Marconi
- Medicine and Surgery, Biosciences & Medical Embryology Section, University of Perugia, Perugia 06132, Italy
| | - Katia Fettucciari
- Medicine and Surgery, Biosciences & Medical Embryology Section, University of Perugia, Perugia 06132, Italy
| |
Collapse
|
23
|
Lin CN, Okabayashi T, Tummaruk P, Ooi PT. Editorial: Zoonotic diseases among pigs. Front Vet Sci 2023; 9:1122679. [PMID: 36686156 PMCID: PMC9851072 DOI: 10.3389/fvets.2022.1122679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Accepted: 12/20/2022] [Indexed: 01/07/2023] Open
Affiliation(s)
- Chao-Nan Lin
- Department of Veterinary Medicine, College of Veterinary Medicine, National Pingtung University of Science and Technology, Pingtung, Taiwan,*Correspondence: Chao-Nan Lin ✉
| | - Tamaki Okabayashi
- Center for Animal Disease Control, University of Miyazaki, Miyazaki, Japan,Tamaki Okabayashi ✉
| | - Padet Tummaruk
- Faculty of Veterinary Science, Chulalongkorn University, Bangkok, Thailand,Padet Tummaruk ✉
| | - Peck Toung Ooi
- Faculty of Veterinary Medicine, University Putra Malaysia, Serdang, Malaysia,Peck Toung Ooi ✉
| |
Collapse
|
24
|
Pearce SJ, Turner K, Cogan TA, Foster AP. A survey of enteric disease agents in UK deer populations. Res Vet Sci 2023; 154:113-123. [PMID: 36577269 DOI: 10.1016/j.rvsc.2022.12.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 12/13/2022] [Indexed: 12/16/2022]
Abstract
Wild deer products have been linked to outbreaks of Escherichia coli 0157 in humans and bovine tuberculosis (bTB) in domestic cats, raising concerns around foodborne diseases from venison. This study investigated the prevalence of several enteric disease agents in UK deer populations, including foodborne pathogens and other diseases of import to livestock health. Intestinal samples were collected from slaughtered farmed deer (N = 211, eight farms), shot wild deer (N = 136) and ground faecal samples were collected from two farms (N = 90), six parks (N = 228) and five zoos (N = 67). DNA was extracted and multiplex qPCR assays were run to amplify targets of, C. perfringens toxins, Campylobacter spp., E. coli toxins, Mycobacterium avium subsp. Paratuberculosis (MAP), Salmonella spp. and Yersinia spp. The most commonly amplified targets were E. coli astA (61.2%) and Campylobacter spp. (43.3%), but the prevalence of C. coli and C. jejuni were ≤ 3.0% and Salmonella spp., MAP and Yersinia pseudotuberculosis also had low overall prevalences of 1.6%, 3.3% and 2.6%, respectively. However, notable targets included C. perfringens toxins α (20.4%) and β2 (16.9%); E. coli stx1 (14.6% overall, 28.0% in abattoir samples) and stx2 (17.8% overall, 31.6% in wild samples); and Yersinia enterocolitica (10.8% overall, 25% in wild samples). The low prevalence of some foodborne pathogens is reassuring for food safety; further investigations into the commonly found targets are needed to quantify the risk to public health and the possibility of wild deer acting as reservoirs of disease.
Collapse
Affiliation(s)
- Samuel J Pearce
- Bristol Veterinary School, University of Bristol, Langford House, Langford, North Somerset BS40 5DU, United Kingdom
| | - Katy Turner
- Bristol Veterinary School, University of Bristol, Langford House, Langford, North Somerset BS40 5DU, United Kingdom
| | - Tristan A Cogan
- Bristol Veterinary School, University of Bristol, Langford House, Langford, North Somerset BS40 5DU, United Kingdom
| | - Aiden P Foster
- Bristol Veterinary School, University of Bristol, Langford House, Langford, North Somerset BS40 5DU, United Kingdom.
| |
Collapse
|
25
|
Zhong T, Wang Y, Wang X, Freitas-de-Melo A, Li H, Zhan S, Wang L, Cao J, Dai D, Guo J, Li L, Zhang H, Liu J, Niu L. Diarrhea in suckling lambs is associated with changes in gut microbiota, serum immunological and biochemical parameters in an intensive production system. Front Microbiol 2022; 13:1020657. [PMID: 36466638 PMCID: PMC9712182 DOI: 10.3389/fmicb.2022.1020657] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Accepted: 10/21/2022] [Indexed: 07/30/2023] Open
Abstract
The incidence of diarrhea in lambs is frequent in large-scale sheep farms, which greatly impacts the growth and health of lambs. The aim of this study was to assess the changes in serum biochemical and immunological parameters and gut microbiome composition in suckling lambs suffering from diarrhea or not, reared on an intensive commercial farm. We found a reduced diversity of intestinal bacteria in suckling lambs suffering from diarrhea. Firmicutes and Bacteroidetes were the dominant flora in both groups of lambs, while the Bacteroidetes decreased in diarrheic lambs, no changes were reported in Firmicutes. Compared with healthy lambs, the proportion of aerobic bacteria, facultative anaerobic bacteria, and stress tolerant bacteria increased in lambs suffering from diarrhea, while that of anaerobic bacteria and potentially pathogenic bacteria decreased slightly. In addition, the contents of total cholesterol, immunoglobulins (Ig) G, and IgM in the serum of lambs suffering from diarrhea were lower than those of healthy lambs. This study explored the association between diarrhea occurrence, intestinal microbial community structure, and metabolic and immunological status in Hu lambs.
Collapse
Affiliation(s)
- Tao Zhong
- Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Yaxuan Wang
- Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Xinlu Wang
- Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Aline Freitas-de-Melo
- Departamento de Biociencias Veterinarias, Facultad de Veterinaria, Universidad de la República, Montevideo, Uruguay
| | - Hua Li
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, China
| | - Siyuan Zhan
- Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Linjie Wang
- Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Jiaxue Cao
- Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Dinghui Dai
- Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Jiazhong Guo
- Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Li Li
- Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Hongping Zhang
- Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Jinwang Liu
- Yulin Sannong Breeding Service Co., Ltd, Yulin, China
| | - Lili Niu
- Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| |
Collapse
|
26
|
Widhani A, Djauzi S, Suyatna FD, Dewi BE. Changes in Gut Microbiota and Systemic Inflammation after Synbiotic Supplementation in Patients with Systemic Lupus Erythematosus: A Randomized, Double-Blind, Placebo-Controlled Trial. Cells 2022; 11:3419. [PMID: 36359816 PMCID: PMC9658918 DOI: 10.3390/cells11213419] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 10/22/2022] [Accepted: 10/23/2022] [Indexed: 08/04/2023] Open
Abstract
Gut dysbiosis has a role in the pathogenesis of lupus. Synbiotic supplementation may restore the balance of gut microbiota. This study investigated whether synbiotics could improve gut microbiota and systemic inflammation in lupus patients. This randomized, double-blind, placebo-controlled trial was conducted in adult systemic lupus erythematosus (SLE) patients. Subjects were randomized to receive either synbiotics or a placebo. Fecal microbiota, hs-CRP, IL-6, and IL-17 were measured at baseline and after 60 days. Patients who fulfilled the inclusion criteria were randomized into synbiotic (n = 23) and placebo groups (n = 23). In the synbiotic group, hs-CRP was not significantly increased (1.8 [0.9; 4.85] vs. 2.1 [0.9; 4.25] mg/L; pre vs. post; p = 0.23), whereas in the placebo group hs-CRP was increased significantly (1.75 [0.4; 4.45] vs. 3.75 [0.58; 7.05] mg/L; pre vs. post; p = 0.005). In the synbiotic group, IL-6 decreased significantly (8.76 [6.62; 11.39] vs. 6.59 [4.96; 8.01]; pre vs. post; p = 0.02), while there was no significant change in IL-17 level. In the placebo group, there was no significant change in IL-6 and IL-17. Synbiotic supplementation increased the Firmicutes:Bacteroidetes ratio (0.05 ± 0.60 vs. -0.08 ± 0.63, synbiotic vs. placebo p = 0.48) and butyrate metabolism (p = 0.037) and decreased amino sugar and nucleotide sugar metabolism (p = 0.040). There was improvement in the SLE disease activity index 2K (SLEDAI-2K) score in the synbiotic group (14 [9; 16] vs. 8 [2; 12]; pre vs. post; p < 0.001), while no change in the placebo group (9 [8; 18.25] vs. 9 [5.5; 15]; pre vs. post; p = 0.31). Synbiotic supplementation could reduce systemic inflammation and SLE disease activity and alter the composition and functions of gut microbiota.
Collapse
Affiliation(s)
- Alvina Widhani
- Allergy and Clinical Immunology Division, Department of Internal Medicine, Faculty of Medicine, Universitas Indonesia, Jakarta 10430, Indonesia
- Dr. Cipto Mangunkusumo Hospital, Jakarta 10430, Indonesia
- Doctoral Program in Biomedical Science, Faculty of Medicine, Universitas Indonesia, Jakarta 10430, Indonesia
| | - Samsuridjal Djauzi
- Allergy and Clinical Immunology Division, Department of Internal Medicine, Faculty of Medicine, Universitas Indonesia, Jakarta 10430, Indonesia
- Dr. Cipto Mangunkusumo Hospital, Jakarta 10430, Indonesia
| | | | - Beti Ernawati Dewi
- Department of Microbiology, Faculty of Medicine, Universitas Indonesia, Jakarta 10430, Indonesia
| |
Collapse
|
27
|
Oba S, Yildirim T, Karataş ŞM. Probiotics Safety Aspect of Functional Foods. JOURNAL OF CULINARY SCIENCE & TECHNOLOGY 2022. [DOI: 10.1080/15428052.2022.2135156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Affiliation(s)
- Sirin Oba
- Department of Food Processing, Suluova Vocational School, Amasya University, Amasya, Turkey
| | - Tugce Yildirim
- Department of Biotechnology, Institution of Science, Amasya University, Amasya, Turkey
| | | |
Collapse
|
28
|
Yadav JP, Kaur S, Dhaka P, Vijay D, Bedi JS. Prevalence, molecular characterization, and antimicrobial resistance profile of Clostridium perfringens from India: A scoping review. Anaerobe 2022; 77:102639. [PMID: 36108893 DOI: 10.1016/j.anaerobe.2022.102639] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 04/27/2022] [Accepted: 09/07/2022] [Indexed: 11/01/2022]
Abstract
Clostridium perfringens is one of the most important foodborne pathogens that causes histotoxic diseases and intestinal infections in both humans and animals. The present scoping review has been designed to analyze the literature published during 2000-2021 from India on the prevalence, molecular characterization, and antimicrobial resistance profile of C. perfringens isolates recovered from humans, animals, animal-based foods, and associated environmental samples. The peer-reviewed articles retrieved from four electronic databases (Google Scholar, PubMed, Science Direct, and Web of Science) were assessed using PRISMA-ScR guidelines. A total of 32 studies from India were selected on the basis of their relevance and inclusion criteria. The overall prevalence of C. perfringens among domestic animals having history of clinical symptoms and among healthy animals was found to be 65.8% (508/772) and 42.8% (493/1152), respectively. The pathogen was also detected in clinically affected wild animals (75%), healthy wild animals (35.4%), and captive birds (24.5%). The detection of C. perfringens among poultry having necrotic enteritis and among healthy birds was found to be 66.8% (321/480) and 25.6% (80/312), respectively. The detection of pathogen among animal-based foods (i.e., meat, milk, and fish and their products) and environmental samples depicted a prevalence of 20.8% (325/1562) and 30.2% (23/76), respectively. However, the prevalence of C. perfringens among humans having history of diarrhea and among healthy humans was found to be 25% (70/280) and 23.2% (36/155), respectively. The genotyping of C. perfringens isolates revealed that toxin type A was found to be the most prevalent genotype. Along with the alpha toxin gene (cpa), beta (cpb), epsilon (etx), iota (itx), enterotoxin (cpe), beta-2 toxin (cpb2), and NetB (netB) toxins were also detected in different combinations. Antimicrobial resistance profile of C. perfringens isolates recovered from different sources demonstrated that the highest resistance was detected against sulphonamides (76.8%) and tetracycline (41.3%) by phenotypic and genotypic detection methods, respectively. Comprehensive scientific studies covering different geographical areas at the human-animal-environment interface are crucial to generalize the real magnitude of C. perfringens-associated problem in India and for establishing a reliable database.
Collapse
Affiliation(s)
- Jay Prakash Yadav
- Department of Veterinary Public Health and Epidemiology, College of Veterinary Science, Guru Angad Dev Veterinary and Animal Sciences University, Rampura Phul, Bathinda, 151103, India.
| | - Simranpreet Kaur
- Centre for One Health, College of Veterinary Science, Guru Angad Dev Veterinary and Animal Sciences University, Ludhiana, 141004, India
| | - Pankaj Dhaka
- Centre for One Health, College of Veterinary Science, Guru Angad Dev Veterinary and Animal Sciences University, Ludhiana, 141004, India
| | - Deepthi Vijay
- Department of Veterinary Public Health, College of Veterinary and Animal Sciences, Mannuthy, Thrissur, 680651, India
| | - Jasbir Singh Bedi
- Centre for One Health, College of Veterinary Science, Guru Angad Dev Veterinary and Animal Sciences University, Ludhiana, 141004, India
| |
Collapse
|
29
|
Karagulle B, Celik F, Simsek S, Ahmed H, Shen Y, Cao J. First molecular evidence of Clostridium perfringens in adult Fasciola spp. isolates in cattle hosts. Front Vet Sci 2022; 9:967045. [PMID: 36118337 PMCID: PMC9478789 DOI: 10.3389/fvets.2022.967045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Accepted: 08/12/2022] [Indexed: 11/18/2022] Open
Abstract
Fasciolosis is a parasitic disease caused by Fasciola spp. It is a prevalent helminth infection globally. Clostridial hepatitis is a general name refer to disorders caused by a few clostridial agents that most severely affect the liver. Migration of young parasite forms (mostly Fasciola hepatica) in the parenchymal tissue of the liver causes necrosis and anaerobic environment, stimulating the proliferation of C. novyi type B spores. This study investigated the occurrence of Clostridium spp in adult Fasciola spp isolates. Isolates (n = 100) were collected from the bile ducts of infected cattle after slaughter. Total genomic DNA was extracted from each sample. A multiplex-PCR based on the flagellin C (fliC) gene was used for quick identification of C. chauvoei, C. haemolyticum, C. novyi types A and B, and C. septicum. In addition, a pair of primers Cpa (F) and Cpa (R) were used for detection of the C. perfringens alpha toxin gene. The products were sequenced. No band was obtained after multiplex-PCR of the fliC gene. A 247 bp band was detected in two isolates using the Cpa primers. BLAST analysis of these two isolates characterized both as C. perfringens alpha toxin. This is the first description of the molecular detection of C. perfringens in flukes. Further studies are needed to investigate whether Clostridum species is also carried by other developmental forms (egg and larval stages) of Fasciola spp.
Collapse
Affiliation(s)
- Burcu Karagulle
- Department of Microbiology, Faculty of Veterinary Medicine, University of Firat, Elazig, Turkey
| | - Figen Celik
- Department of Parasitology, Faculty of Veterinary Medicine, University of Firat, Elazig, Turkey
| | - Sami Simsek
- Department of Parasitology, Faculty of Veterinary Medicine, University of Firat, Elazig, Turkey
- *Correspondence: Sami Simsek
| | - Haroon Ahmed
- Department of Biosciences, COMSATS University Islamabad (CUI), Islamabad, Pakistan
| | - Yujuan Shen
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, Shanghai, China
- Chinese Center for Tropical Diseases Research, Shanghai, China
- Key Laboratory of Parasite and Vector Biology, National Health Commission of the People's Republic of China, Shanghai, China
- WHO Collaborating Center for Tropical Diseases, Shanghai, China
- The School of Global Health, Chinese Center for Tropical Diseases Research, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jianping Cao
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, Shanghai, China
- Chinese Center for Tropical Diseases Research, Shanghai, China
- Key Laboratory of Parasite and Vector Biology, National Health Commission of the People's Republic of China, Shanghai, China
- WHO Collaborating Center for Tropical Diseases, Shanghai, China
- The School of Global Health, Chinese Center for Tropical Diseases Research, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Jianping Cao
| |
Collapse
|
30
|
Pathology and Pathogenesis of Brain Lesions Produced by Clostridium perfringens Type D Epsilon Toxin. Int J Mol Sci 2022; 23:ijms23169050. [PMID: 36012315 PMCID: PMC9409160 DOI: 10.3390/ijms23169050] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 08/08/2022] [Accepted: 08/09/2022] [Indexed: 11/23/2022] Open
Abstract
Clostridium perfringens type D epsilon toxin (ETX) produces severe, and frequently fatal, neurologic disease in ruminant livestock. The disorder is of worldwide distribution and, although vaccination has reduced its prevalence, ETX still causes substantial economic loss in livestock enterprises. The toxin is produced in the intestine as a relatively inactive prototoxin, which is subsequently fully enzymatically activated to ETX. When changed conditions in the intestinal milieu, particularly starch overload, favor rapid proliferation of this clostridial bacterium, large amounts of ETX can be elaborated. When sufficient toxin is absorbed from the intestine into the systemic circulation and reaches the brain, two neurologic syndromes can develop from this enterotoxemia. If the brain is exposed to large amounts of ETX, the lesions are fundamentally vasculocentric. The neurotoxin binds to microvascular endothelial receptors and other brain cells, the resulting damage causing increased vascular permeability and extravasation of plasma protein and abundant fluid into the brain parenchyma. While plasma protein, particularly albumin, pools largely perivascularly, the vasogenic edema becomes widely distributed in the brain, leading to a marked rise in intracranial pressure, coma, sometimes cerebellar herniation, and, eventually, often death. When smaller quantities of ETX are absorbed into the bloodstream, or livestock are partially immune, a more protracted clinical course ensues. The resulting brain injury is characterized by bilaterally symmetrical necrotic foci in certain selectively vulnerable neuroanatomic sites, termed focal symmetrical encephalomalacia. ETX has also been internationally listed as a potential bioterrorism agent. Although there are no confirmed human cases of ETX intoxication, the relatively wide species susceptibility to this toxin and its high toxicity mean it is likely that human populations would also be vulnerable to its neurotoxic actions. While the pathogenesis of ETX toxicity in the brain is incompletely understood, the putative mechanisms involved in neural lesion development are discussed.
Collapse
|
31
|
Yuan B, Sun Z, Lu M, Lillehoj H, Lee Y, Liu L, Yan X, Yang DA, Li C. Immunization with Pooled Antigens for Clostridium perfringens Conferred Partial Protection against Experimental Necrotic Enteritis in Broiler Chickens. Vaccines (Basel) 2022; 10:vaccines10060979. [PMID: 35746587 PMCID: PMC9229587 DOI: 10.3390/vaccines10060979] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Revised: 06/09/2022] [Accepted: 06/15/2022] [Indexed: 12/10/2022] Open
Abstract
Necrotic enteritis (NE) is a multifactorial and important enteric infectious disease etiologically caused by pathogenic C. perfringens infection, accounting for the estimated loss of around USD 6 billion in the global poultry industry. The increasing incidence of NE was found to be associated with the voluntary reduction or withdrawal of antibiotic growth promoters from animal feed during recent years. Therefore, the development of effective vaccines specific to NE assumes a priority for the poultry industry. This study aimed to identify the potential C. perfringens proteins as vaccine targets for NE. Three recombinant C. perfringens proteins targeting five antigens were prepared: two chimeric proteins (alpha-toxin and NetB, fructose-1,6-bisphosphate aldolase (FBA) and a zinc metalloprotease (Zm)), and one single collagen adhesion protein (Cna). Their protection efficacies were evaluated with a potent challenge model of Eimeria maxima/C. perfringens dual infections using a netB+tpeL+ C. perfringens strain. Young chicks were immunized twice subcutaneously with adjuvanted C. perfringens proteins on Days 4 and 15. At six days after the second immunization, the chickens immunized with Cna, FBA, and Zm antigens, and alpha-toxin had much higher serum antibody titers than unvaccinated controls prior to the challenge. Following the challenge, the pooled antigen-immunized group demonstrated no mortality and the least lesion scores against virulent challenge. The results indicate that the immunization with multicomponent antigens, including C. perfringens housekeeping protein Cna, may confer partial protection.
Collapse
Affiliation(s)
- Baohong Yuan
- Animal Bioscience and Biotechnology Laboratory, Agricultural Research Service (ARS), United States Department of Agriculture (USDA), Beltsville, MD 20705, USA; (B.Y.); (Z.S.); (M.L.); (H.L.); (Y.L.); (L.L.)
- School of Basic Medicine Sciences, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Zhifeng Sun
- Animal Bioscience and Biotechnology Laboratory, Agricultural Research Service (ARS), United States Department of Agriculture (USDA), Beltsville, MD 20705, USA; (B.Y.); (Z.S.); (M.L.); (H.L.); (Y.L.); (L.L.)
| | - Mingmin Lu
- Animal Bioscience and Biotechnology Laboratory, Agricultural Research Service (ARS), United States Department of Agriculture (USDA), Beltsville, MD 20705, USA; (B.Y.); (Z.S.); (M.L.); (H.L.); (Y.L.); (L.L.)
| | - Hyun Lillehoj
- Animal Bioscience and Biotechnology Laboratory, Agricultural Research Service (ARS), United States Department of Agriculture (USDA), Beltsville, MD 20705, USA; (B.Y.); (Z.S.); (M.L.); (H.L.); (Y.L.); (L.L.)
| | - Youngsub Lee
- Animal Bioscience and Biotechnology Laboratory, Agricultural Research Service (ARS), United States Department of Agriculture (USDA), Beltsville, MD 20705, USA; (B.Y.); (Z.S.); (M.L.); (H.L.); (Y.L.); (L.L.)
| | - Liheng Liu
- Animal Bioscience and Biotechnology Laboratory, Agricultural Research Service (ARS), United States Department of Agriculture (USDA), Beltsville, MD 20705, USA; (B.Y.); (Z.S.); (M.L.); (H.L.); (Y.L.); (L.L.)
| | - Xianghe Yan
- Environmental Microbial and Food Safety Laboratory, Agricultural Research Service (ARS), United States Department of Agriculture (USDA), Beltsville, MD 20705, USA;
| | - Danchen Aaron Yang
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China;
| | - Charles Li
- Animal Bioscience and Biotechnology Laboratory, Agricultural Research Service (ARS), United States Department of Agriculture (USDA), Beltsville, MD 20705, USA; (B.Y.); (Z.S.); (M.L.); (H.L.); (Y.L.); (L.L.)
- Correspondence:
| |
Collapse
|
32
|
Jiang Z, Su W, Wen C, Li W, Zhang Y, Gong T, Du S, Wang X, Lu Z, Jin M, Wang Y. Effect of Porcine Clostridium perfringens on Intestinal Barrier, Immunity, and Quantitative Analysis of Intestinal Bacterial Communities in Mice. Front Vet Sci 2022; 9:881878. [PMID: 35769317 PMCID: PMC9234579 DOI: 10.3389/fvets.2022.881878] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Accepted: 04/26/2022] [Indexed: 11/16/2022] Open
Abstract
Clostridium perfringens (C. perfringens) is one of the main pathogens which can cause a range of histotoxic and enteric diseases in humans or animals (pigs, or broilers). The Centers for Disease Control and Prevention (CDC) estimates these bacteria cause nearly 1 million illnesses in the United States every year. For animal husbandry, necrotizing enteritis caused by C. perfringens can cost the global livestock industry between $2 billion and $6 billion per year. C. perfringens-infected animals can be isolated for its identification and pathology. A suitable animal model is one of the essential conditions for studying the disease pathogenesis. In previous studies, mice have been used as subjects for a variety of Clostridium perfringens toxicity tests. Thus, this study was designed to build a mouse model infected porcine C. perfringens which was isolated from the C.perfringens-infected pigs. A total of 32 6-week-old male C57BL/6 mice were randomly divided into four groups. Control group was orally administrated with PBS (200 μL) on day 0. Low group, Medium group, and High group were gavaged with 200 ul of PBS resuspension containing 8.0 × 107 CFU, 4.0 × 108 CFU, and 2.0 × 109 CFU, respectively. We examined growth performance, immune status, intestinal barrier integrity, apoptosis-related genes expression, and copies of C. perfringens in mice. The results showed that the growth performance declined and intestinal structure was seriously damaged in High group. Meanwhile, pro-inflammatory factors (IL-1β, TNF-α, and IL-6) were significantly increased (P < 0.05) in High group compared to other groups. The tight junctions and pro-apoptosis related genes' expression significantly decreased (P < 0.05) in High group, and high dose caused a disruption of intestinal villi integrity and tissue injury in the jejunum of mice. In addition, the enumerations of C. perfringens, Escherichia coli, and Lactobacillus explained why the gut of High group mice was seriously damaged, because the C. perfringens and Escherichia coli significantly enriched (P < 0.05), and Lactobacillus dramatically decreased (P < 0.05). Overall, our results provide an experimental and theoretical basis for understanding the pathogenesis and exploring the effects of porcine C. perfringens on mice.
Collapse
Affiliation(s)
- Zipeng Jiang
- National Engineering Laboratory of Biological Feed Safety and Pollution Prevention and Control, Zhejiang University, Hangzhou, China
- Key Laboratory of Molecular Nutrition, Ministry of Education, Zhejiang University, Hangzhou, China
- Key Laboratory of Animal Nutrition and Feed, Ministry of Agriculture and Rural Affairs, Zhejiang University, Hangzhou, China
- Key Laboratory of Animal Nutrition and Feed Nutrition of Zhejiang Province, Zhejiang University, Hangzhou, China
- College of Animal Science, Institute of Feed Science, Zhejiang University, Hangzhou, China
| | - Weifa Su
- National Engineering Laboratory of Biological Feed Safety and Pollution Prevention and Control, Zhejiang University, Hangzhou, China
- Key Laboratory of Molecular Nutrition, Ministry of Education, Zhejiang University, Hangzhou, China
- Key Laboratory of Animal Nutrition and Feed, Ministry of Agriculture and Rural Affairs, Zhejiang University, Hangzhou, China
- Key Laboratory of Animal Nutrition and Feed Nutrition of Zhejiang Province, Zhejiang University, Hangzhou, China
- College of Animal Science, Institute of Feed Science, Zhejiang University, Hangzhou, China
| | - Chaoyue Wen
- National Engineering Laboratory of Biological Feed Safety and Pollution Prevention and Control, Zhejiang University, Hangzhou, China
- Key Laboratory of Molecular Nutrition, Ministry of Education, Zhejiang University, Hangzhou, China
- Key Laboratory of Animal Nutrition and Feed, Ministry of Agriculture and Rural Affairs, Zhejiang University, Hangzhou, China
- Key Laboratory of Animal Nutrition and Feed Nutrition of Zhejiang Province, Zhejiang University, Hangzhou, China
- College of Animal Science, Institute of Feed Science, Zhejiang University, Hangzhou, China
| | - Wentao Li
- National Engineering Laboratory of Biological Feed Safety and Pollution Prevention and Control, Zhejiang University, Hangzhou, China
- Key Laboratory of Molecular Nutrition, Ministry of Education, Zhejiang University, Hangzhou, China
- Key Laboratory of Animal Nutrition and Feed, Ministry of Agriculture and Rural Affairs, Zhejiang University, Hangzhou, China
- Key Laboratory of Animal Nutrition and Feed Nutrition of Zhejiang Province, Zhejiang University, Hangzhou, China
- College of Animal Science, Institute of Feed Science, Zhejiang University, Hangzhou, China
| | - Yu Zhang
- National Engineering Laboratory of Biological Feed Safety and Pollution Prevention and Control, Zhejiang University, Hangzhou, China
- Key Laboratory of Molecular Nutrition, Ministry of Education, Zhejiang University, Hangzhou, China
- Key Laboratory of Animal Nutrition and Feed, Ministry of Agriculture and Rural Affairs, Zhejiang University, Hangzhou, China
- Key Laboratory of Animal Nutrition and Feed Nutrition of Zhejiang Province, Zhejiang University, Hangzhou, China
- College of Animal Science, Institute of Feed Science, Zhejiang University, Hangzhou, China
| | - Tao Gong
- National Engineering Laboratory of Biological Feed Safety and Pollution Prevention and Control, Zhejiang University, Hangzhou, China
- Key Laboratory of Molecular Nutrition, Ministry of Education, Zhejiang University, Hangzhou, China
- Key Laboratory of Animal Nutrition and Feed, Ministry of Agriculture and Rural Affairs, Zhejiang University, Hangzhou, China
- Key Laboratory of Animal Nutrition and Feed Nutrition of Zhejiang Province, Zhejiang University, Hangzhou, China
- College of Animal Science, Institute of Feed Science, Zhejiang University, Hangzhou, China
| | - Shuai Du
- National Engineering Laboratory of Biological Feed Safety and Pollution Prevention and Control, Zhejiang University, Hangzhou, China
- Key Laboratory of Molecular Nutrition, Ministry of Education, Zhejiang University, Hangzhou, China
- Key Laboratory of Animal Nutrition and Feed, Ministry of Agriculture and Rural Affairs, Zhejiang University, Hangzhou, China
- Key Laboratory of Animal Nutrition and Feed Nutrition of Zhejiang Province, Zhejiang University, Hangzhou, China
- College of Animal Science, Institute of Feed Science, Zhejiang University, Hangzhou, China
| | - Xinxia Wang
- National Engineering Laboratory of Biological Feed Safety and Pollution Prevention and Control, Zhejiang University, Hangzhou, China
- Key Laboratory of Molecular Nutrition, Ministry of Education, Zhejiang University, Hangzhou, China
- Key Laboratory of Animal Nutrition and Feed, Ministry of Agriculture and Rural Affairs, Zhejiang University, Hangzhou, China
- Key Laboratory of Animal Nutrition and Feed Nutrition of Zhejiang Province, Zhejiang University, Hangzhou, China
- College of Animal Science, Institute of Feed Science, Zhejiang University, Hangzhou, China
| | - Zeqing Lu
- National Engineering Laboratory of Biological Feed Safety and Pollution Prevention and Control, Zhejiang University, Hangzhou, China
- Key Laboratory of Molecular Nutrition, Ministry of Education, Zhejiang University, Hangzhou, China
- Key Laboratory of Animal Nutrition and Feed, Ministry of Agriculture and Rural Affairs, Zhejiang University, Hangzhou, China
- Key Laboratory of Animal Nutrition and Feed Nutrition of Zhejiang Province, Zhejiang University, Hangzhou, China
- College of Animal Science, Institute of Feed Science, Zhejiang University, Hangzhou, China
- *Correspondence: Zeqing Lu
| | - Mingliang Jin
- National Engineering Laboratory of Biological Feed Safety and Pollution Prevention and Control, Zhejiang University, Hangzhou, China
- Key Laboratory of Molecular Nutrition, Ministry of Education, Zhejiang University, Hangzhou, China
- Key Laboratory of Animal Nutrition and Feed, Ministry of Agriculture and Rural Affairs, Zhejiang University, Hangzhou, China
- Key Laboratory of Animal Nutrition and Feed Nutrition of Zhejiang Province, Zhejiang University, Hangzhou, China
- College of Animal Science, Institute of Feed Science, Zhejiang University, Hangzhou, China
| | - Yizhen Wang
- National Engineering Laboratory of Biological Feed Safety and Pollution Prevention and Control, Zhejiang University, Hangzhou, China
- Key Laboratory of Molecular Nutrition, Ministry of Education, Zhejiang University, Hangzhou, China
- Key Laboratory of Animal Nutrition and Feed, Ministry of Agriculture and Rural Affairs, Zhejiang University, Hangzhou, China
- Key Laboratory of Animal Nutrition and Feed Nutrition of Zhejiang Province, Zhejiang University, Hangzhou, China
- College of Animal Science, Institute of Feed Science, Zhejiang University, Hangzhou, China
- Yizhen Wang
| |
Collapse
|
33
|
Ruiz-Rico M, Renwick S, Allen-Vercoe E, Barat JM. In vitro susceptibility of human gut microbes to potential food preservatives based on immobilized phenolic compounds. Food Chem 2022; 378:132136. [PMID: 35042114 DOI: 10.1016/j.foodchem.2022.132136] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 12/31/2021] [Accepted: 01/09/2022] [Indexed: 11/04/2022]
Abstract
The development of novel food preservatives based on natural antimicrobials such as phenolic compounds is increasing, but their safety should be established before use, including evaluating their impact on the gut microbiota. This work explored the influence of antimicrobial phenolics presented in different forms on selected human gut microbiota members through in vitro susceptibility tests. The bacteria tested exhibited a wide range of susceptibilities to phenolics depending on the molecule structure and mode of administration. Agathobacter rectalis and Clostridium spiroforme, members of the phylum Firmicutes, were the most sensitive strains. Susceptibility was strain- and species-specific, suggesting that it may not be possible to easily extrapolate results across the human microbiome in general. Species of other phyla including Bacteroidetes, Actinobacteria, Proteobacteria and Verrucomicrobia were more resistant than Firmicutes, with growth of some strains even enhanced. Our results provide insights into the biocompatibility of free and immobilized phenolics as potential food additives.
Collapse
Affiliation(s)
- María Ruiz-Rico
- Departamento de Tecnología de Alimentos, Universitat Politècnica de València, Valencia, Spain.
| | - Simone Renwick
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, Ontario, Canada
| | - Emma Allen-Vercoe
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, Ontario, Canada
| | - José M Barat
- Departamento de Tecnología de Alimentos, Universitat Politècnica de València, Valencia, Spain
| |
Collapse
|
34
|
Lu W, Sun H, Xu ZM, Du Z, Si L, Yuan S, Jin J, Jin CH. Diagnostic and therapeutic strategy for Clostridium perfringens infection in postpartum dairy cows: a report of 14 cases. JOURNAL OF APPLIED ANIMAL RESEARCH 2022. [DOI: 10.1080/09712119.2022.2078329] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Affiliation(s)
- Wengeng Lu
- Heilongjiang Provincial Key Laboratory of Prevention and Control of Bovine Diseases, College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, People’s Republic of China
| | - Hongliang Sun
- Heilongjiang Provincial Key Laboratory of Prevention and Control of Bovine Diseases, College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, People’s Republic of China
| | - Zheng-Mei Xu
- Heilongjiang Provincial Key Laboratory of Prevention and Control of Bovine Diseases, College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, People’s Republic of China
| | - Zhenzhen Du
- Heilongjiang Provincial Key Laboratory of Prevention and Control of Bovine Diseases, College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, People’s Republic of China
| | - Linqing Si
- Heilongjiang Provincial Key Laboratory of Prevention and Control of Bovine Diseases, College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, People’s Republic of China
| | - Siqi Yuan
- Heilongjiang Provincial Key Laboratory of Prevention and Control of Bovine Diseases, College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, People’s Republic of China
| | - Jidong Jin
- Cofeed Feedmill (Changchun) Co., Ltd., Changchun, People’s Republic of China
| | - Cheng-Hao Jin
- Department of Biochemistry and Molecular Biology, College of Life Science & Technology, Heilongjiang Bayi Agricultural University, Daqing, People’s Republic of China
- Department of Food Science and Engineering, College of Food Science, Heilongjiang Bayi Agricultural University, Daqing, People’s Republic of China
- National Coarse Cereals Engineering Research Center, Daqing, People’s Republic of China
| |
Collapse
|
35
|
Blokhin AA, Toropova NN, Burova OA, Iashin IV, Zakharova OI. Blackleg in Cattle in the Irkutsk Region. Front Vet Sci 2022; 9:872386. [PMID: 35647103 PMCID: PMC9133737 DOI: 10.3389/fvets.2022.872386] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 04/11/2022] [Indexed: 11/13/2022] Open
Abstract
Blackleg is an acute, toxic, infectious, non-contagious disease of domestic and wild ruminants that occurs while the animals are pastured. This article describes an outbreak of blackleg on a farm in Siberia (Russia) in 2019. We provide a detailed description of the cases based on the results of comprehensive diagnostic and epidemiological investigations. For description of case and evaluation, we used the following methods: owner observations, descriptive epidemiology, clinical diagnostics, pathological examination and bacteriology. The distinctive features (in addition to the characteristic features) were as follows: the outbreak of the disease occurred in early spring when there was abundant snow cover and under unfavorable living conditions of animals and traumas; the disease appeared in both vaccinated and unvaccinated cattle; the characteristic clinical signs were low-grade fever, the absence of crepitus, and the presence of haematomas containing erythrocytes with basophilic granularity; thrombs in vessel and vacuolization in tissue of the adrenal gland. This paper aimed to present classical and new clinical and pathology changes in cattle with blackleg in winter conditions of Russian Siberia.
Collapse
|
36
|
Sheh A, Artim SC, Burns MA, Molina-Mora JA, Lee MA, Dzink-Fox J, Muthupalani S, Fox JG. Alterations in common marmoset gut microbiome associated with duodenal strictures. Sci Rep 2022; 12:5277. [PMID: 35347206 PMCID: PMC8960757 DOI: 10.1038/s41598-022-09268-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 03/21/2022] [Indexed: 12/13/2022] Open
Abstract
Chronic gastrointestinal (GI) diseases are the most common diseases in captive common marmosets (Callithrix jacchus). Despite standardized housing, diet and husbandry, a recently described gastrointestinal syndrome characterized by duodenal ulcers and strictures was observed in a subset of marmosets sourced from the New England Primate Research Center. As changes in the gut microbiome have been associated with GI diseases, the gut microbiome of 52 healthy, non-stricture marmosets (153 samples) were compared to the gut microbiome of 21 captive marmosets diagnosed with a duodenal ulcer/stricture (57 samples). No significant changes were observed using alpha diversity metrics, and while the community structure was significantly different when comparing beta diversity between healthy and stricture cases, the results were inconclusive due to differences observed in the dispersion of both datasets. Differences in the abundance of individual taxa using ANCOM, as stricture-associated dysbiosis was characterized by Anaerobiospirillum loss and Clostridium perfringens increases. To identify microbial and serum biomarkers that could help classify stricture cases, we developed models using machine learning algorithms (random forest, classification and regression trees, support vector machines and k-nearest neighbors) to classify microbiome, serum chemistry or complete blood count (CBC) data. Random forest (RF) models were the most accurate models and correctly classified strictures using either 9 ASVs (amplicon sequence variants), 4 serum chemistry tests or 6 CBC tests. Based on the RF model and ANCOM results, C. perfringens was identified as a potential causative agent associated with the development of strictures. Clostridium perfringens was also isolated by microbiological culture in 4 of 9 duodenum samples from marmosets with histologically confirmed strictures. Due to the enrichment of C. perfringens in situ, we analyzed frozen duodenal tissues using both 16S microbiome profiling and RNAseq. Microbiome analysis of the duodenal tissues of 29 marmosets from the MIT colony confirmed an increased abundance of Clostridium in stricture cases. Comparison of the duodenal gene expression from stricture and non-stricture marmosets found enrichment of genes associated with intestinal absorption, and lipid metabolism, localization, and transport in stricture cases. Using machine learning, we identified increased abundance of C. perfringens, as a potential causative agent of GI disease and intestinal strictures in marmosets.
Collapse
Affiliation(s)
- Alexander Sheh
- Division of Comparative Medicine, Massachusetts Institute of Technology, Cambridge, MA, USA.
| | - Stephen C Artim
- Division of Comparative Medicine, Massachusetts Institute of Technology, Cambridge, MA, USA
- Merck Research Laboratories, Merck, South San Francisco, CA, USA
| | - Monika A Burns
- Division of Comparative Medicine, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Jose Arturo Molina-Mora
- Centro de Investigación en Enfermedades Tropicales (CIET), Universidad de Costa Rica, San José, Costa Rica
| | - Mary Anne Lee
- Division of Comparative Medicine, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Biological Sciences, Wellesley College, Wellesley, MA, USA
| | - JoAnn Dzink-Fox
- Division of Comparative Medicine, Massachusetts Institute of Technology, Cambridge, MA, USA
| | | | - James G Fox
- Division of Comparative Medicine, Massachusetts Institute of Technology, Cambridge, MA, USA.
| |
Collapse
|
37
|
Clostridial Diseases of Horses: A Review. Vaccines (Basel) 2022; 10:vaccines10020318. [PMID: 35214776 PMCID: PMC8876495 DOI: 10.3390/vaccines10020318] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 02/10/2022] [Accepted: 02/13/2022] [Indexed: 11/17/2022] Open
Abstract
The clostridial diseases of horses can be divided into three major groups: enteric/enterotoxic, histotoxic, and neurotoxic. The main enteric/enterotoxic diseases include those produced by Clostridium perfringens type C and Clostridioides difficile, both of which are characterized by enterocolitis. The main histotoxic diseases are gas gangrene, Tyzzer disease, and infectious necrotic hepatitis. Gas gangrene is produced by one or more of the following microorganisms: C. perfringens type A, Clostridium septicum, Paeniclostridium sordellii, and Clostridium novyi type A, and it is characterized by necrotizing cellulitis and/or myositis. Tyzzer disease is produced by Clostridium piliforme and is mainly characterized by multifocal necrotizing hepatitis. Infectious necrotic hepatitis is produced by Clostridium novyi type B and is characterized by focal necrotizing hepatitis. The main neurotoxic clostridial diseases are tetanus and botulism, which are produced by Clostridium tetani and Clostridium botulinum, respectively. Tetanus is characterized by spastic paralysis and botulism by flaccid paralysis. Neither disease present with specific gross or microscopic lesions. The pathogenesis of clostridial diseases involves the production of toxins. Confirming a diagnosis of some of the clostridial diseases of horses is sometimes difficult, mainly because some agents can be present in tissues of normal animals. This paper reviews the main clostridial diseases of horses.
Collapse
|
38
|
Minich D, Madden C, Navarro MA, Glowacki L, French-Kim K, Chan W, Evans MV, Soares K, Mrofchak R, Madan R, Ballash GA, LaPerle K, Paul S, Vodovotz Y, Uzal FA, Martinez M, Hausmann J, Junge RE, Hale VL. Gut microbiota and age shape susceptibility to clostridial enteritis in lorikeets under human care. Anim Microbiome 2022; 4:7. [PMID: 35000619 PMCID: PMC8744333 DOI: 10.1186/s42523-021-00148-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Accepted: 12/07/2021] [Indexed: 12/14/2022] Open
Abstract
Background Enteritis is a common cause of morbidity and mortality in lorikeets that can be challenging to diagnose and treat. In this study, we examine gut microbiota in two lorikeet flocks with enteritis (Columbus Zoo and Aquarium—CZA; Denver Zoo—DZ). Since 2012, the CZA flock has experienced repeated outbreaks of enteritis despite extensive diet, husbandry, and clinical modifications. In 2018, both CZA and DZ observed a spike in enteritis. Recent research has revealed that the gut microbiota can influence susceptibility to enteropathogens. We hypothesized that a dysbiosis, or alteration in the gut microbial community, was making some lorikeets more susceptible to enteritis, and our goal was to characterize this dysbiosis and determine the features that predicted susceptibility.
Results We employed 16S rRNA sequencing to characterize the cloacal microbiota in lorikeets (CZA n = 67, DZ n = 24) over time. We compared the microbiota of healthy lorikeets, to lorikeets with enteritis, and lorikeets susceptible to enteritis, with “susceptible” being defined as healthy birds that subsequently developed enteritis. Based on sequencing data, culture, and toxin gene detection in intestinal contents, we identified Clostridium perfringens type A (CZA and DZ) and C. colinum (CZA only) at increased relative abundances in birds with enteritis. Histopathology and immunohistochemistry further identified the presence of gram-positive bacilli and C. perfringens, respectively, in the necrotizing intestinal lesions. Finally, using Random Forests and LASSO models, we identified several features (young age and the presence of Rhodococcus fascians and Pseudomonas umsongensis) associated with susceptibility to clostridial enteritis. Conclusions We identified C. perfringens type A and C. colinum associated with lorikeet necrohemorrhagic enteritis at CZA and DZ. Susceptibility testing of isolates lead to an updated clinical treatment plan which ultimately resolved the outbreaks at both institutions. This work provides a foundation for understanding gut microbiota features that are permissive to clostridial colonization and host factors (e.g. age, prior infection) that shape responses to infection. Supplementary Information The online version contains supplementary material available at 10.1186/s42523-021-00148-7.
Collapse
Affiliation(s)
- David Minich
- Department of Veterinary Preventive Medicine, Ohio State University College of Veterinary Medicine, 1902 Coffey Rd., Columbus, OH, 43210, USA
| | - Christopher Madden
- Department of Veterinary Preventive Medicine, Ohio State University College of Veterinary Medicine, 1902 Coffey Rd., Columbus, OH, 43210, USA
| | - Mauricio A Navarro
- California Animal Health & Food Safety Lab, University of California, Davis, San Bernardino, CA, USA.,Instituto de Patología Animal, Facultad de Ciencias Veterinarias, Universidad Austral de Chile, Valdivia, Chile
| | - Leo Glowacki
- Ohio State University College of Arts and Sciences, Columbus, OH, USA
| | - Kristen French-Kim
- Department of Veterinary Preventive Medicine, Ohio State University College of Veterinary Medicine, 1902 Coffey Rd., Columbus, OH, 43210, USA
| | - Willow Chan
- Ohio State University College of Food, Agricultural, and Environmental Sciences, Columbus, OH, USA
| | - Morgan V Evans
- Department of Veterinary Preventive Medicine, Ohio State University College of Veterinary Medicine, 1902 Coffey Rd., Columbus, OH, 43210, USA.,Ohio State University College of Public Health, Columbus, OH, USA
| | - Kilmer Soares
- Department of Veterinary Preventive Medicine, Ohio State University College of Veterinary Medicine, 1902 Coffey Rd., Columbus, OH, 43210, USA.,Department of Animal Science, College of Agricultural Sciences (CCA), Federal University of Paraiba (UFPB), Areia, PB, Brazil
| | - Ryan Mrofchak
- Department of Veterinary Preventive Medicine, Ohio State University College of Veterinary Medicine, 1902 Coffey Rd., Columbus, OH, 43210, USA
| | - Rushil Madan
- Department of Veterinary Preventive Medicine, Ohio State University College of Veterinary Medicine, 1902 Coffey Rd., Columbus, OH, 43210, USA
| | - Gregory A Ballash
- Department of Veterinary Preventive Medicine, Ohio State University College of Veterinary Medicine, 1902 Coffey Rd., Columbus, OH, 43210, USA
| | - Krista LaPerle
- Department of Veterinary Preventive Medicine, Ohio State University College of Veterinary Medicine, 1902 Coffey Rd., Columbus, OH, 43210, USA.,Comparative Pathology & Digital Imaging Shared Resource, Ohio State University, Columbus, OH, USA
| | - Subhadeep Paul
- Ohio State University College of Arts and Sciences, Columbus, OH, USA
| | - Yael Vodovotz
- Ohio State University College of Food, Agricultural, and Environmental Sciences, Columbus, OH, USA
| | - Francisco A Uzal
- California Animal Health & Food Safety Lab, University of California, Davis, San Bernardino, CA, USA
| | - Margaret Martinez
- Department of Veterinary Preventive Medicine, Ohio State University College of Veterinary Medicine, 1902 Coffey Rd., Columbus, OH, 43210, USA.,The Marine Mammal Center, Sausalito, CA, USA
| | | | | | - Vanessa L Hale
- Department of Veterinary Preventive Medicine, Ohio State University College of Veterinary Medicine, 1902 Coffey Rd., Columbus, OH, 43210, USA.
| |
Collapse
|
39
|
Uzal FA, Arroyo LG, Navarro MA, Gomez DE, Asín J, Henderson E. Bacterial and viral enterocolitis in horses: a review. J Vet Diagn Invest 2021; 34:354-375. [PMID: 34763560 DOI: 10.1177/10406387211057469] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Enteritis, colitis, and enterocolitis are considered some of the most common causes of disease and death in horses. Determining the etiology of these conditions is challenging, among other reasons because different causes produce similar clinical signs and lesions, and also because some agents of colitis can be present in the intestine of normal animals. We review here the main bacterial and viral causes of enterocolitis of horses, including Salmonella spp., Clostridium perfringens type A NetF-positive, C. perfringens type C, Clostridioides difficile, Clostridium piliforme, Paeniclostridium sordellii, other clostridia, Rhodococcus equi, Neorickettsia risticii, Lawsonia intracellularis, equine rotavirus, and equine coronavirus. Diarrhea and colic are the hallmark clinical signs of colitis and enterocolitis, and the majority of these conditions are characterized by necrotizing changes in the mucosa of the small intestine, colon, cecum, or in a combination of these organs. The presumptive diagnosis is based on clinical, gross, and microscopic findings, and confirmed by detection of some of the agents and/or their toxins in the intestinal content or feces.
Collapse
Affiliation(s)
- Francisco A Uzal
- California Animal Health and Food Safety Laboratory, University of California-Davis, San Bernardino Laboratory, USA
| | - Luis G Arroyo
- Department of Clinical Studies, Ontario Veterinary College, University of Guelph, Guelph, Ontario, Canada
| | - Mauricio A Navarro
- California Animal Health and Food Safety Laboratory, University of California-Davis, San Bernardino Laboratory, USA.,Instituto de Patología Animal, Facultad de Ciencias Veterinarias, Universidad Austral de Chile, Valdivia, Chile
| | - Diego E Gomez
- Department of Clinical Studies, Ontario Veterinary College, University of Guelph, Guelph, Ontario, Canada
| | - Javier Asín
- California Animal Health and Food Safety Laboratory, University of California-Davis, San Bernardino Laboratory, USA
| | - Eileen Henderson
- California Animal Health and Food Safety Laboratory, University of California-Davis, San Bernardino Laboratory, USA
| |
Collapse
|
40
|
Wang T, Guo XK, Xu H. Disentangling the Progression of Non-alcoholic Fatty Liver Disease in the Human Gut Microbiota. Front Microbiol 2021; 12:728823. [PMID: 34721326 PMCID: PMC8548776 DOI: 10.3389/fmicb.2021.728823] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 09/16/2021] [Indexed: 12/12/2022] Open
Abstract
Gut microbiome dysbiosis has been known to be associated with all stages of non-alcoholic fatty liver disease (NAFLD), but questions remain about microbial profiles in progression and homogeneity across NAFLD stages. We performed a meta-analysis of three publicly shotgun datasets and built predictive models to determine diagnostic capacity. Here, we found consistently microbiome shifts across NAFLD stages, of which co-occurrence patterns and core sets of new biomarkers significantly correlated with NAFLD progression were identified. Machine learning models that are able to distinguish patients with any NAFLD stage from healthy controls remained predictive when applied to patients with other NAFLD stages, suggesting the homogeneity across stages once again. Focusing on species and metabolic pathways specifically associated with progressive stages, we found that increased toxic metabolites and decreased protection of butyrate and choline contributed to advanced NAFLD. We further built models discriminating one stage from the others with an average of 0.86 of area under the curve. In conclusion, this meta-analysis firmly establishes generalizable microbiome dysbiosis and predictive taxonomic and functional signatures as a basis for future diagnostics across NAFLD stages.
Collapse
Affiliation(s)
- Tianjiao Wang
- School of Medicine, Tsinghua University, Beijing, China
| | - Xue-Kun Guo
- School of Medicine, Tsinghua University, Beijing, China
| | - Huji Xu
- School of Medicine, Tsinghua University, Beijing, China.,Department of Rheumatology and Immunology, Shanghai Changzheng Hospital, The Second Military Medical University, Shanghai, China.,Peking-Tsinghua Center for Life Sciences, Tsinghua University, Beijing, China
| |
Collapse
|
41
|
Oliveira RDC, de Oliveira Júnior CA, Alves GG, Assis RA, Silva ROS, de Sousa Xavier MA, Lobato FCF. Cattle and goats' humoral response to vaccination with Clostridium perfringens type D purified epsilon toxoids. Anaerobe 2021; 72:102465. [PMID: 34662696 DOI: 10.1016/j.anaerobe.2021.102465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 10/14/2021] [Accepted: 10/15/2021] [Indexed: 11/30/2022]
Abstract
Herd vaccination is an important preventive measure against enterotoxemia in ruminants. Vaccination in goats should be performed every four months, and recent studies have shown that immunity in cattle lasts for less than one year. One of the mechanisms for increasing the duration of the immune response is to use purified toxoids as immunogens. The aim of the present study was to evaluate the humoral response in cattle and goats after vaccination with purified and semi-purified Clostridium perfringens type D epsilon toxoid. The following three different vaccines were used: vaccine 1 (V1), a semi-purified toxoid adsorbed to aluminum hydroxide; vaccine 2 (V2), a purified toxoid adsorbed to aluminum hydroxide; and vaccine (V3), a purified toxoid adsorbed on chitosan microparticles. Groups of cattle (n = 6-7) and goats (n = 6-7) were vaccinated on days 0 and 30, and serum samples for antitoxin titration were collected every 30 days for one-year post-vaccination. Goats were revaccinated on day 360, and their serum was evaluated on days 367 and 374. The antibody peaks ranged between 6.90 and 11.47 IU/mL in cattle and from 1.11 to 4.40 IU/mL in goats. In cattle administered with the V1 and V2 vaccines, we observed that the antibody titers were maintained above 0.2 IU/mL until the end of the experiment. In goats, V2 elicited long-lasting antibodies, and all animals maintained the protective titers for 210 days after the first dose. In conclusion, the purified toxoid vaccine with aluminum hydroxide adjuvant was able to induce strong and long-lasting humoral responses in both species and could be an alternative for improving the immunization schedule against enterotoxemia in goats and cattle.
Collapse
Affiliation(s)
- Ricardo de Castro Oliveira
- Veterinary School, Universidade Federal de Minas Gerais (UFMG), Avenida Antônio Carlos, 6627, Belo Horizonte, CEP 31.270-901, MG, Brazil; Merck Sharp & Dohme (MSD), Avenida Comendador Loureiro Ramos, 1500, Montes Claros, CEP 39.404-003, MG, Brazil
| | - Carlos Augusto de Oliveira Júnior
- Veterinary School, Universidade Federal de Minas Gerais (UFMG), Avenida Antônio Carlos, 6627, Belo Horizonte, CEP 31.270-901, MG, Brazil
| | - Guilherme Guerra Alves
- Veterinary School, Universidade Federal de Minas Gerais (UFMG), Avenida Antônio Carlos, 6627, Belo Horizonte, CEP 31.270-901, MG, Brazil
| | - Ronnie Antunes Assis
- Veterinary School, Universidade Federal de Minas Gerais (UFMG), Avenida Antônio Carlos, 6627, Belo Horizonte, CEP 31.270-901, MG, Brazil; Merck Sharp & Dohme (MSD), Avenida Comendador Loureiro Ramos, 1500, Montes Claros, CEP 39.404-003, MG, Brazil
| | - Rodrigo Otávio Silveira Silva
- Veterinary School, Universidade Federal de Minas Gerais (UFMG), Avenida Antônio Carlos, 6627, Belo Horizonte, CEP 31.270-901, MG, Brazil
| | - Mauro Aparecido de Sousa Xavier
- Universidade Estadual de Montes Claros (UNIMONTES), Avenida, Professor Rui Braga, S/N Montes Claros, CEP 39.401-089, MG, Brazil
| | - Francisco Carlos Faria Lobato
- Veterinary School, Universidade Federal de Minas Gerais (UFMG), Avenida Antônio Carlos, 6627, Belo Horizonte, CEP 31.270-901, MG, Brazil.
| |
Collapse
|
42
|
Lorine D, Céline D, Caroline LM, Frédéric B, Lorette H, Julie B, Laure M, Christine Z, Typhaine P, Sandra R, Emmanuelle H, Rabab SZ, Jeanne C, Anne-Marie P. Influence of operating conditions on the persistence of E. coli, enterococci, Clostridium perfringens and Clostridioides difficile in semi-continuous mesophilic anaerobic reactors. WASTE MANAGEMENT (NEW YORK, N.Y.) 2021; 134:32-41. [PMID: 34403994 DOI: 10.1016/j.wasman.2021.08.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 06/30/2021] [Accepted: 08/03/2021] [Indexed: 06/13/2023]
Abstract
This study examined the combined effect of hydraulic retention time (HRT), organic loading rate (OLR) and heat pretreatment of manure (70 °C, 1 h) on the fate of E. coli, enterococci, C. perfringens, C. difficile, and on chemical parameters (volatile fatty acids and ammonia) that may inactivate pathogens. Semi-continuous mesophilic anaerobic reactors were fed with pig manure and horse feed. The operating conditions were 2, 3, 4 COD.L-1.d-1 (OLR), 24, 35, 46 days (HRT) and use or not of a thermal pretreatment. The levels of the chemical parameters did not reach concentrations capable of inactivating the four bacteria. Anaerobic digestion led to a Log10 removal > 3 (E. coli), 0.9-2.1 (enterococci), 0.1-0.6 (C. perfringens) and 0-1 (C. difficile). Increasing HRT only reduced the concentration of E. coli in the digestate. Increasing OLR reduced the Log10 removal of enterococci and C. difficile. The heat pretreatment led to non-detection of E. coli in the digestate, reduced the concentration of C. perfringens by 0.8-1.3 Log10 and increased the concentration of C. difficile by 0.04-0.7 Log10. Enterococci, not detected in the heated manure, were present in the digestate. The distribution of genes encoding virulence factors of C. difficile (tcdA and tcdB) and C. perfringens (cpa, cpb2 and cpb) was not impacted by anaerobic digestion or by the heat pretreatment. Enterococci, C. perfringens, C. difficile were present in the digestate at relatively stable concentrations regardless of the operating conditions, indicating that even with heat pretreatment, the biosafety of digestate cannot be guaranteed in mesophilic conditions.
Collapse
Affiliation(s)
- Derongs Lorine
- INRAE, OPAALE Research Unit, CS 64427, F-35044 Rennes, France
| | - Druilhe Céline
- INRAE, OPAALE Research Unit, CS 64427, F-35044 Rennes, France
| | - Le Maréchal Caroline
- ANSES, Ploufragan-Plouzané Laboratory, Hygiene and Quality of Poultry and Pig Products Unit, BP53, F-22440 Ploufragan, France
| | - Barbut Frédéric
- National Reference Laboratory for Clostridium difficile, Saint-Antoine Hospital, Assistance Publique- Hôpitaux de Paris, 34 rue Crozatier, 75012 Paris, France; UMR INSERM S-1139, Faculté de Pharmacie de Paris, Université de Paris, France
| | | | - Buffet Julie
- INRAE, OPAALE Research Unit, CS 64427, F-35044 Rennes, France
| | - Martin Laure
- ANSES, Ploufragan-Plouzané Laboratory, Hygiene and Quality of Poultry and Pig Products Unit, BP53, F-22440 Ploufragan, France
| | | | - Poezevara Typhaine
- ANSES, Ploufragan-Plouzané Laboratory, Hygiene and Quality of Poultry and Pig Products Unit, BP53, F-22440 Ploufragan, France
| | - Rouxel Sandra
- ANSES, Ploufragan-Plouzané Laboratory, Hygiene and Quality of Poultry and Pig Products Unit, BP53, F-22440 Ploufragan, France
| | - Houard Emmanuelle
- ANSES, Ploufragan-Plouzané Laboratory, Hygiene and Quality of Poultry and Pig Products Unit, BP53, F-22440 Ploufragan, France
| | - Syed Zaidi Rabab
- National Reference Laboratory for Clostridium difficile, Saint-Antoine Hospital, Assistance Publique- Hôpitaux de Paris, 34 rue Crozatier, 75012 Paris, France; UMR INSERM S-1139, Faculté de Pharmacie de Paris, Université de Paris, France
| | - Couturier Jeanne
- National Reference Laboratory for Clostridium difficile, Saint-Antoine Hospital, Assistance Publique- Hôpitaux de Paris, 34 rue Crozatier, 75012 Paris, France; UMR INSERM S-1139, Faculté de Pharmacie de Paris, Université de Paris, France
| | | |
Collapse
|
43
|
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 2021; 101:103887. [PMID: 34579847 DOI: 10.1016/j.fm.2021.103887] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 08/06/2021] [Accepted: 08/18/2021] [Indexed: 11/25/2022]
Abstract
The purpose of this study was to investigate the distribution and specify the transmission and cross-contamination of Clostridium perfringens (C. perfringens) in the beef slaughtering and butchering process. The prevalence of 21.2% (150/708) yielded 208 isolates of C. perfringens, including 80.8% type A and 19.2% type D, 0.4% (3/708) samples carried both type A and D strains, and 72.5% type D isolates carried both cpe and atyp.cpb2 genes. C. perfringens were identified through the whole slaughtering process but no type F (cpe and cpa isolates) was found. 69 isolates were further analyzed and classified into 28 PFGE genotypes and clade I contained 94.2% isolates and 24 PFGE genotypes, which showed the genetic diversity and epidemic correlation. Our study traced C. perfringens contamination along the handling processes and showed a gradually ascending contamination rate during the whole process, revealing widespread cross-contamination from the feces and hides of slaughtered cattle to the carcass in the slaughtering workshop, so as from tools and personnel to meat of the cutting workshops. Strains from different slaughterhouses (regions) have high homology, and type A is the predominant toxinotype. It is necessary to monitor and control several key points of cross-contamination during slaughtering process to reduce a risk of C. perfringens infection.
Collapse
Affiliation(s)
- Yanfen Jiang
- College of Veterinary Medicine, Northwest A&F University, 712100, Yangling, Shaanxi, China
| | - Yinghui Ma
- College of Veterinary Medicine, Northwest A&F University, 712100, Yangling, Shaanxi, China
| | - Qianqian Liu
- College of Veterinary Medicine, Northwest A&F University, 712100, Yangling, Shaanxi, China
| | - Tianmei Li
- College of Veterinary Medicine, Northwest A&F University, 712100, Yangling, Shaanxi, China
| | - Yiming Li
- College of Veterinary Medicine, Northwest A&F University, 712100, Yangling, Shaanxi, China
| | - Kangkang Guo
- College of Veterinary Medicine, Northwest A&F University, 712100, Yangling, Shaanxi, China.
| | - Yanming Zhang
- College of Veterinary Medicine, Northwest A&F University, 712100, Yangling, Shaanxi, China.
| |
Collapse
|
44
|
Identifying the Basis for VirS/VirR Two-Component Regulatory System Control of Clostridium perfringens Beta-Toxin Production. J Bacteriol 2021; 203:e0027921. [PMID: 34228498 DOI: 10.1128/jb.00279-21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Clostridium perfringens toxin production is often regulated by the Agr-like quorum sensing (QS) system signaling the VirS/VirR two-component regulatory system (TCRS), which consists of the VirS membrane sensor histidine kinase and the VirR response regulator. VirS/VirR is known to directly control expression of some genes by binding to a DNA binding motif consisting of two VirR boxes located within 500 bp of the target gene start codon. Alternatively, the VirS/VirR system can indirectly regulate production levels of other proteins by increasing expression of a small regulatory RNA, VR-RNA. Previous studies demonstrated that C. perfringens beta-toxin (CPB) production by C. perfringens type B and C strains is positively regulated by both the Agr-like QS and the VirS/VirR TCRS, but the mechanism has been unclear. The current study first inactivated the vrr gene encoding VR-RNA to show that VirS/VirR regulation of cpb expression does not involve VR-RNA. Subsequently, bioinformatic analyses identified a potential VirR binding motif, along with a predicted strong promoter, ∼1.4 kb upstream of the cpb open reading frame (ORF). Two insertion sequences were present between this VirR binding motif/promoter region and the cpb ORF. PCR screening of a collection of strains carrying cpb showed that the presence and sequence of this VirR binding motif/promoter is highly conserved among CPB-producing strains. Reverse transcription-PCR (RT-PCR) and a GusA reporter assay showed this VirR binding motif is important for regulating CPB production. These findings indicate that VirS/VirR directly regulates cpb expression via VirS binding to a VirR binding motif located unusually distant from the cpb start codon. IMPORTANCE Clostridium perfringens beta-toxin (CPB) is only produced by type B and C strains. Production of CPB is essential for the pathogenesis of type C-associated infections, which include hemorrhagic necrotizing enteritis and enterotoxemia in both humans and animals. In addition, CPB can synergize with other toxins during C. perfringens gastrointestinal diseases. CPB toxin production is cooperatively regulated by the Agr-like quorum sensing (QS) system and the VirS/VirR two-component regulatory system. This study now reports that the VirS/VirR regulatory cascade directly controls expression of the cpb gene via a process involving a VirR box binding motif located unusually far (∼1.4 kb) upstream of the cpb ORF. This study provides a better understanding of the regulatory mechanisms for CPB production by the VirS/VirR regulatory cascade.
Collapse
|
45
|
Fettucciari K, Marconi P, Marchegiani A, Fruganti A, Spaterna A, Bassotti G. Invisible steps for a global endemy: molecular strategies adopted by Clostridioides difficile. Therap Adv Gastroenterol 2021; 14:17562848211032797. [PMID: 34413901 PMCID: PMC8369858 DOI: 10.1177/17562848211032797] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Accepted: 06/26/2021] [Indexed: 02/06/2023] Open
Abstract
Clostridioides difficile infection (CDI) is on the rise worldwide and is associated with an increase in deaths and socio-health burden. C. difficile has become ubiquitous in anthropized environments because of the extreme resistance of its spores. Based on the epidemiological data and knowledge of molecular pathogenesis of C. difficile, it is possible to predict its progressive colonization of the human population for the following reasons: first, its global spread is unstoppable; second, the toxins (Tcds) produced by C. difficile, TcdA and TcdB, mainly cause cell death by apoptosis, but the surviving cells acquire a senescence state that favours persistence of C. difficile in the intestine; third, proinflammatory cytokines, tumour necrosis factor-α and interferon-γ, induced during CDI, enhance the cytotoxicity of Tcds and can increase the survival of senescent cells; fourth, Tcds block mobility and induce apoptosis in immune cells recruited at the infection site; and finally, after remission from primary infection or relapse, C. difficile causes functional abnormalities in the enteric glial cell (EGC) network that can result in irritable bowel syndrome, characterized by a latent inflammatory response that contributes to C. difficile survival and enhances the cytotoxic activity of low doses of TcdB, thus favouring further relapses. Since a 'global endemy' of C. difficile seems inevitable, it is necessary to develop an effective vaccine against Tcds for at-risk individuals, and to perform a prophylaxis/selective therapy with bacteriophages highly specific for C. difficile. We must be aware that CDI will become a global health problem in the forthcoming years, and we must be prepared to face this menace.
Collapse
Affiliation(s)
- Katia Fettucciari
- Biosciences & Medical Embryology Section, Department of Medicine and Surgery, University of Perugia, Medical School -Piazza Lucio Severi 1, Edificio B - IV piano; Sant’Andrea delle Fratte, Perugia, 06132, Italy
| | - Pierfrancesco Marconi
- Biosciences & Medical Embryology Section, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Andrea Marchegiani
- School of Biosciences and Veterinary Medicine, University of Camerino, Macerata, Italy
| | - Alessandro Fruganti
- School of Biosciences and Veterinary Medicine, University of Camerino, Macerata, Italy
| | - Andrea Spaterna
- School of Biosciences and Veterinary Medicine, University of Camerino, Macerata, Italy
| | - Gabrio Bassotti
- Gastroenterology, Hepatology & Digestive Endoscopy Section, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
- Gastroenterology & Hepatology Unit, Santa Maria della Misericordia Hospital, Perugia, Italy
| |
Collapse
|
46
|
Liu L, Yan X, Lillehoj H, Sun Z, Zhao H, Xianyu Z, Lee Y, Melville S, Gu C, Wang Y, Lu M, Li C. Comparison of the Pathogenicity of Five Clostridium perfringens Isolates Using an Eimeria maxima Coinfection Necrotic Enteritis Disease Model in Commercial Broiler Chickens. Avian Dis 2021; 64:386-392. [PMID: 33205165 DOI: 10.1637/aviandiseases-d-19-00098] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Accepted: 04/15/2020] [Indexed: 11/05/2022]
Abstract
Clostridium perfringens (CP) is the etiologic agent of necrotic enteritis (NE) in broiler chickens that is responsible for massive economic losses in the poultry industry in response to voluntary reduction and withdrawal of antibiotic growth promoters. Large variations exist in the CP isolates in inducing intestinal NE lesions. However, limited information is available on CP isolate genetics in inducing NE with other predisposing factors. This study investigated the ability of five CP isolates from different sources to influence NE pathogenesis by using an Eimeria maxima (EM) coinfection NE model: Str.13 (from soil), LLY_N11 (healthy chicken intestine), SM101 (food poisoning), Del1 (netB+tpeL-) and LLY_Tpel17 (netB+tpeL+) for NE-afflicted chickens. The 2-wk-old broiler chickens were preinfected with EM (5 × 103 oocysts) followed by CP infection (around 1 × 109 colony-forming units per chicken). The group of the LLY_Tpel17 isolate with EM coinfection had 25% mortality. No mortality was observed in the groups infected with EM alone, all CP alone, or dual infections of EM/other CP isolates. In this model of EM/CP coinfections, the relative percentages of body weight gain showed statistically significant decreases in all EM/CP groups except the EM/SM101 group when compared with the sham control group. Evident gut lesions were only observed in the three groups of EM/LLY_N11, EM/Del1, and EM/LLY_Tpel17, all of which possessed an essential NE pathogenesis locus in their genomes. Our studies indicate that LLY_Tpel17 is highly pathogenic to induce severe gut lesions and would be a good CP challenge strain for studies investigating pathogenesis and evaluating the protection efficacy for antibiotic alternative approaches.
Collapse
Affiliation(s)
- Liheng Liu
- Animal Bioscience and Biotechnology Laboratory, Agricultural Research Service (ARS), United States Department of Agriculture (USDA), Beltsville, MD 20705.,College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, China
| | - Xianghe Yan
- Environmental Microbial and Food Safety Laboratory, ARS/USDA, Beltsville, MD 20705
| | - Hyun Lillehoj
- Animal Bioscience and Biotechnology Laboratory, Agricultural Research Service (ARS), United States Department of Agriculture (USDA), Beltsville, MD 20705
| | - Zhifeng Sun
- Animal Bioscience and Biotechnology Laboratory, Agricultural Research Service (ARS), United States Department of Agriculture (USDA), Beltsville, MD 20705
| | - Hongyan Zhao
- Animal Bioscience and Biotechnology Laboratory, Agricultural Research Service (ARS), United States Department of Agriculture (USDA), Beltsville, MD 20705.,College of Veterinary Medicine, Yangzhou University, Yangzhou, China
| | - Zhezi Xianyu
- Animal Bioscience and Biotechnology Laboratory, Agricultural Research Service (ARS), United States Department of Agriculture (USDA), Beltsville, MD 20705
| | - Youngsub Lee
- Animal Bioscience and Biotechnology Laboratory, Agricultural Research Service (ARS), United States Department of Agriculture (USDA), Beltsville, MD 20705
| | - Stephen Melville
- Department of Biological Sciences, Virginia Polytech and State University, Blacksburg, VA 24061
| | - Changqin Gu
- Animal Bioscience and Biotechnology Laboratory, Agricultural Research Service (ARS), United States Department of Agriculture (USDA), Beltsville, MD 20705.,College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Yunfei Wang
- Biostatistics Center, Duke Human Vaccine Institute, Durham, NC 27710
| | - Mingmin Lu
- Animal Bioscience and Biotechnology Laboratory, Agricultural Research Service (ARS), United States Department of Agriculture (USDA), Beltsville, MD 20705
| | - Charles Li
- Animal Bioscience and Biotechnology Laboratory, Agricultural Research Service (ARS), United States Department of Agriculture (USDA), Beltsville, MD 20705
| |
Collapse
|
47
|
Sun Y, Ni A, Jiang Y, Li Y, Huang Z, Shi L, Xu H, Chen C, Li D, Han Y, Chen J. Effects of Replacing In-feed Antibiotics with Synergistic Organic Acids on Growth Performance, Health, Carcass, and Immune and Oxidative Statuses of Broiler Chickens Under Clostridium perfringens Type A Challenge. Avian Dis 2021; 64:393-400. [PMID: 33205169 DOI: 10.1637/aviandiseases-d-19-00101] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Accepted: 04/06/2020] [Indexed: 11/05/2022]
Abstract
This study was conducted to investigate the effects of replacing in-feed antibiotics with synergistic organic acids on growth performance, health, carcass, and immune and oxidative statuses of broiler chickens under Clostridium perfringens (CP) type A challenge. Two organic acid products were tested: organic acid 1 (OA1), consisting of butyrate, medium-chain fatty acids, organic acids, and phenolics; and organic acid 2 (OA2), consisting of buffered short-chain fatty acids. Six hundred 1-day-old male Arbor Acres broiler chicks were randomly assigned to one of five treatments: Control 1, basal diet, nonchallenged birds; Control 2, basal diet, with CP challenge; antimicrobial growth promoters (AGP), basal diet supplemented with Aureomycin (chlortetracycline), with CP challenge; OA1, basal diet supplemented with OA1, with CP challenge; and OA1OA2, basal diet supplemented with OA1 and OA2, with CP challenge. Each treatment had eight replicate pens of 15 birds. The experiments lasted for 29 days. The disease challenge was performed on days 15-17, with an oral gavage of 0.5 mL of CP culture (2.0 × 108 colony-forming units [CFU]/mL) for each bird. Body weights (BWs), intestinal lesion scores, immune organ indices, and serum malondialdehyde (MDA) concentrations were measured on days 19, 22, and 29, respectively, in three birds per pen. Carcass characteristics were determined on day 29. No treatment-related differences in mortality were noted before (P = 0.28) or after (P = 0.64) challenge or over the whole study period (days 0-28; P = 0.66). On day 19, the BW of Control 2 was lower than other treatments (P < 0.0001). On day 22, AGP, OA1, and OA1OA2 had higher BW than Control 2 (P = 0.001). The breast muscle yield of OA1 and OA1OA2 was higher than AGP (P < 0.05). The abdominal fat yield of OA1OA2 was lower than AGP and Control 2 (P < 0.05). On day 22, the birds fed OA1OA2 showed lower intestinal lesion scores than OA1 (P < 0.05). No treatment-related differences in immune organ (spleen, thymus, and bursa) indices were noted (P > 0.05). On day 29, the MDA concentration of OA1 and OA1OA2 was lower than those of Control 1 and AGP (P < 0.05). In conclusion, the addition of organic acids may protect broiler chickens from severe intestinal lesions and oxidative stress and may help reduce abdominal fat mass deposition. There is potential for organic acid-based products as alternatives for AGP in preventing necrotic enteritis in broilers.
Collapse
Affiliation(s)
- Yanyan Sun
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Yuanmingyuan West Road 2, Beijing 100193, China
| | - Aixin Ni
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Yuanmingyuan West Road 2, Beijing 100193, China
| | - Ying Jiang
- China Institute of Veterinary Drug Control, Beijing 100081, China
| | - Yunlei Li
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Yuanmingyuan West Road 2, Beijing 100193, China
| | - Ziyan Huang
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Yuanmingyuan West Road 2, Beijing 100193, China
| | - Lei Shi
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Yuanmingyuan West Road 2, Beijing 100193, China
| | - Hong Xu
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Yuanmingyuan West Road 2, Beijing 100193, China
| | - Chao Chen
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Yuanmingyuan West Road 2, Beijing 100193, China
| | - Dongli Li
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Yuanmingyuan West Road 2, Beijing 100193, China
| | - Yanming Han
- Trouw Nutrition R & D, Stationsstraat 77, 3811 MH, Amersfoort, the Netherlands
| | - Jilan Chen
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Yuanmingyuan West Road 2, Beijing 100193, China
| |
Collapse
|
48
|
Crispo M, Stoute ST, Uzal FA, Bickford AA, Shivaprasad HL. Nonenteric Lesions of Necrotic Enteritis in Commercial Chickens in California: 25 Cases (2009-2018). Avian Dis 2021; 64:356-364. [PMID: 33205162 DOI: 10.1637/aviandiseases-d-19-00129] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Accepted: 02/03/2020] [Indexed: 11/05/2022]
Abstract
Necrotic enteritis (NE) is an important enteric disease affecting a wide variety of avian species, including poultry, caused by Clostridium perfringens type G and, rarely, type C. Significant economic losses can result from elevated mortality rates and poor performance, such as decreased weight gain associated with intestinal damage and impaired absorption of nutrients. Additional losses can result from elevated condemnation at the processing plant because of a high incidence of cholangiohepatitis. Nonenteric lesions associated with NE have been rarely reported. This paper describes uncommon presentations of NE in commercial chickens received by the California Animal Health and Food Safety Laboratory (Turlock and Tulare branches) between 2009 and 2018. Overall, extraintestinal lesions associated with C. perfringens were diagnosed in 25 cases of NE involving commercial broiler chickens. The extraintestinal sites most commonly affected included liver, followed by gizzard, bursa of Fabricius, gall bladder, and spleen. The etiology of these lesions, C. perfringens, was confirmed from a combination of gross, bacteriologic, microscopic, and immunohistochemical findings. The most common predisposing factors for NE identified were coccidiosis (56%, 14/25) and immunosuppressive disease agents, including infectious bursal disease virus (16%, 4/25) and fowl adenovirus group 1 (4%, 1/25). Additionally, four cases (16%) had microscopic lesions compatible with cystic enteritis, probably of viral etiology. This study describes the incidence of extraintestinal lesions of NE in chickens, underlying the role of enteric disorders and immunosuppression as major predisposing factors for the development of NE.
Collapse
Affiliation(s)
- Manuela Crispo
- California Animal Health and Food Safety Laboratory System, University of California, Davis, Turlock, CA 95380
| | - Simone T Stoute
- California Animal Health and Food Safety Laboratory System, University of California, Davis, Turlock, CA 95380
| | - Francisco A Uzal
- California Animal Health and Food Safety Laboratory System, University of California, Davis, San Bernardino, CA 92408
| | - Arthur A Bickford
- California Animal Health and Food Safety Laboratory System, University of California, Davis, Turlock, CA 95380
| | - H L Shivaprasad
- California Animal Health and Food Safety Laboratory System, University of California, Davis, Tulare, CA 93274
| |
Collapse
|
49
|
Geier RR, Rehberger TG, Smith AH. Comparative Genomics of Clostridium perfringens Reveals Patterns of Host-Associated Phylogenetic Clades and Virulence Factors. Front Microbiol 2021; 12:649953. [PMID: 34177831 PMCID: PMC8220089 DOI: 10.3389/fmicb.2021.649953] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Accepted: 04/30/2021] [Indexed: 11/23/2022] Open
Abstract
Clostridium perfringens is an opportunistic pathogenic bacterium that infects both animals and humans. Clostridium perfringens genomes encode a diverse array of toxins and virulence proteins, which continues to expand as more genomes are sequenced. In this study, the genomes of 44 C. perfringens strains isolated from intestinal sections of diseased cattle and from broiler chickens from diseased and healthy flocks were sequenced. These newly assembled genomes were compared to 141 publicly available C. perfringens genome assemblies, by aligning known toxin and virulence protein sequences in the assemblies using BLASTp. The genes for alpha toxin, collagenase, a sialidase (nanH), and alpha-clostripain were present in at least 99% of assemblies analyzed. In contrast, beta toxin, epsilon toxin, iota toxin, and binary enterotoxin of toxinotypes B, C, D, and E were present in less than 5% of assemblies analyzed. Additional sequence variants of beta2 toxin were detected, some of which were missing the leader or signal peptide sequences and therefore likely not secreted. Some pore-forming toxins involved in intestinal diseases were host-associated, the netB gene was only found in avian isolates, while netE, netF, and netG were only present in canine and equine isolates. Alveolysin was positively associated with canine and equine strains and only present in a single monophyletic clade. Strains from ruminant were not associated with known virulence factors and, except for the food poisoning associated clade, were present across the phylogenetic diversity identified to date for C. perfringens. Many C. perfringens strains associated with food poisoning lacked the genes for hyaluronidases and sialidases, important for attaching to and digesting complex carbohydrates found in animal tissues. Overall, the diversity of virulence factors in C. perfringens makes these species capable of causing disease in a wide variety of hosts and niches.
Collapse
Affiliation(s)
| | | | - Alexandra H. Smith
- Arm and Hammer Animal and Food Production, Church & Dwight Co., Inc., Waukesha, WI, United States
| |
Collapse
|
50
|
Segmental Intestinal Necrosis: a Typical Presentation of Acute Hemorrhagic Necrotizing Enteritis. Indian J Surg 2021. [DOI: 10.1007/s12262-021-02938-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
|