1
|
Liang J, Zhang K, Hu X, Lv A. Comparative Genomics Analysis of the Fish Pathogen Rahnella aquatilis KCL-5 Reveals Potential Multidrug Resistance and Virulence Properties. Curr Microbiol 2025; 82:158. [PMID: 40014107 DOI: 10.1007/s00284-025-04125-0] [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: 08/29/2024] [Accepted: 02/06/2025] [Indexed: 02/28/2025]
Abstract
The genus Rahnella has been isolated from human, fish, and water environments. We recently reported on the isolation and genomic identification of a novel pathogen R. aquatilis strain KCL-5 from crucian carp Carassius auratus. To investigate the evolution of bacterial virulence and resistance properties of R. aquatilis, comparative genomics analyses were performed for genus Rahnella strains including R. aquatilis, R. variigena, R. bruchi, and R. victoriana. This analysis provides up-to-date information on genus Rahnella genomic diversity, including comparative analyses of virulence and resistance, synteny, single-nucleotide polymorphisms, average nucleotide identity, core-genes, gene families, and genomic islands. The sister species to R. aquatilis is R. victoriana, and closer R. victoriana than with other Rahnella sp. Multiple genes encoding functions that likely contribute to antimicrobial resistance and pathogenic factors were identified by comparative genome analysis, including multidrug resistance efflux pump, adherence, invasion, and secretion systems. To our knowledge, this is the first report to provide a more detailed insight into the comparative genomic characteristics of Rahnella spp., contributing to the understanding of its diversity and evolution, as well as concerning the virulence of R. aquatilis.
Collapse
Affiliation(s)
- Jing Liang
- Tianjin Key Lab of Aqua-Ecology and Aquaculture, College of Fisheries, Tianjin Agricultural University, Tianjin, 300392, China
| | - Kaiyang Zhang
- Tianjin Key Lab of Aqua-Ecology and Aquaculture, College of Fisheries, Tianjin Agricultural University, Tianjin, 300392, China
| | - Xiucai Hu
- Tianjin Key Lab of Aqua-Ecology and Aquaculture, College of Fisheries, Tianjin Agricultural University, Tianjin, 300392, China.
| | - Aijun Lv
- Tianjin Key Lab of Aqua-Ecology and Aquaculture, College of Fisheries, Tianjin Agricultural University, Tianjin, 300392, China.
| |
Collapse
|
2
|
Menon R, Bhattarai SK, Crossette E, Prince AL, Olle B, Silber JL, Bucci V, Faith J, Norman JM. Multi-omic profiling a defined bacterial consortium for treatment of recurrent Clostridioides difficile infection. Nat Med 2025; 31:223-234. [PMID: 39747680 DOI: 10.1038/s41591-024-03337-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 10/02/2024] [Indexed: 01/04/2025]
Abstract
Donor-derived fecal microbiota treatments are efficacious in preventing recurrent Clostridioides difficile infection (rCDI), but they have inherently variable quality attributes, are difficult to scale and harbor the risk of pathogen transfer. In contrast, VE303 is a defined consortium of eight purified, clonal bacterial strains developed for prevention of rCDI. In the phase 2 CONSORTIUM study, high-dose VE303 was well tolerated and reduced the odds of rCDI by more than 80% compared to placebo. VE303 organisms robustly colonized the gut in the high-dose group and were among the top taxa associated with non-recurrence. Multi-omic modeling identified antibiotic history, baseline stool metabolites and serum cytokines as predictors of both on-study CDI recurrence and VE303 colonization. VE303 potentiated early recovery of the host microbiome and metabolites with increases in short-chain fatty acids, secondary bile acids and bile salt hydrolase genes after antibiotic treatment for CDI, which is considered important to prevent CDI recurrences. These results support the idea that VE303 promotes efficacy in rCDI through multiple mechanisms.
Collapse
Affiliation(s)
| | - Shakti K Bhattarai
- Program in Microbiome Dynamics, Department of Microbiology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | | | | | - Bernat Olle
- Vedanta Biosciences, Inc., Cambridge, MA, USA
| | | | - Vanni Bucci
- Program in Microbiome Dynamics, Department of Microbiology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Jeremiah Faith
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | |
Collapse
|
3
|
Fu S, Yu R, Yang B, Han X, Xu Y, Miao J. Hypoxia-inducible lipid droplet-associated protein (HILPDA) and cystathionine β-synthase (CBS) co-contribute to protecting intestinal epithelial cells from Staphylococcus aureus via regulating lipid droplets formation. Biochim Biophys Acta Mol Cell Biol Lipids 2024; 1869:159558. [PMID: 39173873 DOI: 10.1016/j.bbalip.2024.159558] [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: 04/13/2024] [Revised: 08/06/2024] [Accepted: 08/19/2024] [Indexed: 08/24/2024]
Abstract
Despite Staphylococcus aureus (S. aureus) being a highly studied zoontic bacterium, its enteropathogenicity remains elusive. Herein, our findings demonstrated that S. aureus infection led to the accumulation of lipid droplets (LDs) in intestinal epithelial cells, accompanied by marked elevation inflammatory response that ultimately decreases intracellular bacterial load. The aforestated phenomenon may be partly attributed to the up-regulation of hypoxia-inducible lipid droplet-associated protein (HILPDA) and the concomitant down-regulation of cystathionine β-synthase (CBS) protein. Moreover, S. aureus infection up-regulated the expression of HILPDA, thereby promoting LDs accumulation, and down-regulated that of CBS, consequently inhibiting microsomal triglyceride transfer protein (MTTP) expression. This process may suppress the transport of LDs to the extracellular environment, further contributing to the formation of intracellular LDs. In summary, the results of this study provide significant insights into the intricate mechanisms through which the host organism combats pathogens and maintains the balance of sulfur and lipid metabolism. These findings not only enhance our understanding of the host's defense mechanisms but also offer promising avenues for the development of novel strategies to combat intestinal infectious diseases.
Collapse
Affiliation(s)
- Shaodong Fu
- MOE Joint International Research Laboratory of Animal Health and Food Safety, Key Laboratory of Animal Physiology & Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Rui Yu
- MOE Joint International Research Laboratory of Animal Health and Food Safety, Key Laboratory of Animal Physiology & Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Bo Yang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, Key Laboratory of Animal Physiology & Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Xiangan Han
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China
| | - Yuanyuan Xu
- MOE Joint International Research Laboratory of Animal Health and Food Safety, Key Laboratory of Animal Physiology & Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Jinfeng Miao
- MOE Joint International Research Laboratory of Animal Health and Food Safety, Key Laboratory of Animal Physiology & Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China.
| |
Collapse
|
4
|
Shrestha A, Mehdizadeh Gohari I, Li J, Navarro M, Uzal FA, McClane BA. The biology and pathogenicity of Clostridium perfringens type F: a common human enteropathogen with a new(ish) name. Microbiol Mol Biol Rev 2024; 88:e0014023. [PMID: 38864615 PMCID: PMC11426027 DOI: 10.1128/mmbr.00140-23] [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] [Indexed: 06/13/2024] Open
Abstract
SUMMARYIn the 2018-revised Clostridium perfringens typing classification system, isolates carrying the enterotoxin (cpe) and alpha toxin genes but no other typing toxin genes are now designated as type F. Type F isolates cause food poisoning and nonfoodborne human gastrointestinal (GI) diseases, which most commonly involve type F isolates carrying, respectivefooly, a chromosomal or plasmid-borne cpe gene. Compared to spores of other C. perfringens isolates, spores of type F chromosomal cpe isolates often exhibit greater resistance to food environment stresses, likely facilitating their survival in improperly prepared or stored foods. Multiple factors contribute to this spore resistance phenotype, including the production of a variant small acid-soluble protein-4. The pathogenicity of type F isolates involves sporulation-dependent C. perfringens enterotoxin (CPE) production. C. perfringens sporulation is initiated by orphan histidine kinases and sporulation-associated sigma factors that drive cpe transcription. CPE-induced cytotoxicity starts when CPE binds to claudin receptors to form a small complex (which also includes nonreceptor claudins). Approximately six small complexes oligomerize on the host cell plasma membrane surface to form a prepore. CPE molecules in that prepore apparently extend β-hairpin loops to form a β-barrel pore, allowing a Ca2+ influx that activates calpain. With low-dose CPE treatment, caspase-3-dependent apoptosis develops, while high-CPE dose treatment induces necroptosis. Those effects cause histologic damage along with fluid and electrolyte losses from the colon and small intestine. Sialidases likely contribute to type F disease by enhancing CPE action and, for NanI-producing nonfoodborne human GI disease isolates, increasing intestinal growth and colonization.
Collapse
Affiliation(s)
- Archana Shrestha
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Iman Mehdizadeh Gohari
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Jihong Li
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Mauricio Navarro
- Instituto de Patologia Animal, Facultad de Ciencias Veterinarias, Universidad Austral de Chile, Valdivia, Chile
| | - Francisco A Uzal
- California Animal Health and Food Safety Laboratory System, School of Veterinary Medicine, University of California Davis, San Bernardino, California, USA
| | - Bruce A McClane
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
5
|
Zhong S, Yang J, Huang H. The role of single and mixed biofilms in Clostridioides difficile infection and strategies for prevention and inhibition. Crit Rev Microbiol 2024; 50:285-299. [PMID: 36939635 DOI: 10.1080/1040841x.2023.2189950] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 03/06/2023] [Indexed: 03/21/2023]
Abstract
Clostridioides difficile infection (CDI) is a serious disease with a high recurrence rate. The single and mixed biofilms formed by C. difficile in the gut contribute to the formation of recurrent CDI (rCDI). In parallel, other gut microbes influence the formation and development of C. difficile biofilms, also known as symbiotic biofilms. Interactions between members within the symbiotic biofilm are associated with the worsening or alleviation of CDI. These interactions include effects on C. difficile adhesion and chemotaxis, modulation of LuxS/AI-2 quorum sensing (QS) system activity, promotion of cross-feeding by microbial metabolites, and regulation of intestinal bile acid and pyruvate levels. In the process of C. difficile biofilms control, inhibition of C. difficile initial biofilm formation and killing of C. difficile vegetative cells and spores are the main targets of action. The role of symbiotic biofilms in CDI suggested that targeting interventions of C. difficile-promoting gut microbes could indirectly inhibit the formation of C. difficile mixed biofilms and improved the ultimate therapeutic effect. In summary, this review outlines the mechanisms of C. difficile biofilm formation and summarises the treatment strategies for such single and mixed biofilms, aiming to provide new ideas for the prevention and treatment of CDI.
Collapse
Affiliation(s)
- Saiwei Zhong
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, China
- College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Nanjing, China
| | - Jingpeng Yang
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, China
| | - He Huang
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, China
| |
Collapse
|
6
|
Chen B, Yang X, Zhan M, Chen Y, Xu J, Xiao J, Xiao H, Song M. Dietary tangeretin improved antibiotic-associated diarrhea in mice by enhancing the intestinal barrier function, regulating the gut microbiota, and metabolic homeostasis. Food Funct 2023; 14:10731-10746. [PMID: 37933488 DOI: 10.1039/d3fo02998k] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2023]
Abstract
Antibiotic-associated diarrhea is mediated by antibiotic treatment and is usually caused by the disruption of the intestinal barrier, gut microbiota, and metabolic balance. To identify a dietary strategy that can mitigate the side effects of antibiotics, this study investigated the effect of tangeretin on antibiotic-associated diarrhea in C57BL/6 mice. The results revealed that dietary tangeretin significantly ameliorated symptoms of antibiotic-associated diarrhea, as evidenced by the decreased diarrhea status scores, the reduced fecal water content, the decreased caecum/body weight ratio, and the alleviated colonic tissue damage. Dietary tangeretin also exhibited a protective effect on the intestinal barrier function by upregulating the mRNA and protein expression of claudin-1 and ZO-1. Furthermore, analysis of the gut microbiota using 16S rRNA gene sequencing indicated that dietary tangeretin modulated the gut microbiota of mice with antibiotic-associated diarrhea via increasing the gut microbiota diversity and the abundance of beneficial bacteria, e.g., Lactobacillaceae and Ruminococcaceae, and decreasing the abundance of harmful bacteria, e.g., Enterococcus and Terrisporobacter. Additionally, dietary tangeretin restored the levels of short-chain fatty acids and modulated metabolic pathways by enriching purine metabolism, bile acid metabolism, ABC transporters, and choline metabolism in cancer. Collectively, these findings provide a solid scientific basis for the rational use of tangeretin as a preventive and therapeutic agent for antibiotic-associated diarrhea.
Collapse
Affiliation(s)
- Bin Chen
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Guangzhou, 510642, China.
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, 510642, China
- Department of Food Science, University of Massachusetts, Amherst, Massachusetts 01003, USA.
| | - Xun Yang
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Guangzhou, 510642, China.
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, 510642, China
| | - Minmin Zhan
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Guangzhou, 510642, China.
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, 510642, China
| | - Yilu Chen
- Department of Food Science, University of Massachusetts, Amherst, Massachusetts 01003, USA.
| | - Jingyi Xu
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Guangzhou, 510642, China.
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, 510642, China
| | - Jie Xiao
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Guangzhou, 510642, China.
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, 510642, China
| | - Hang Xiao
- Department of Food Science, University of Massachusetts, Amherst, Massachusetts 01003, USA.
| | - Mingyue Song
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Guangzhou, 510642, China.
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, 510642, China
| |
Collapse
|
7
|
Chowdhury G, Das B, Kumar S, Pant A, Mukherjee P, Ghosh D, Koley H, Miyoshi SI, Okamoto K, Paul A, Dutta S, Ramamurthy T, Mukhopadyay AK. Genomic insights into extensively drug-resistant Pseudomonas aeruginosa isolated from a diarrhea case in Kolkata, India. Future Microbiol 2023; 18:173-186. [PMID: 36916516 DOI: 10.2217/fmb-2022-0140] [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: 03/16/2023] Open
Abstract
Aim: To characterize extensively drug-resistant Pseudomonas aeruginosa from a patient with diarrhea. Materials & methods: Antimicrobial susceptibility was tested by the disk diffusion method. The P. aeruginosa genome was sequenced to identify virulence, antibiotic resistance and prophages encoding genes. Results: P. aeruginosa had a wide spectrum of resistance to antibiotics. Genomic analysis of P. aeruginosa revealed 76 genes associated with antimicrobial resistance, xenobiotic degradation and the type three secretion system. Conclusion: This is the first report on diarrhea associated with P. aeruginosa. Since no other organism was identified, the authors assume that the patient had dysbiosis due to antibiotic exposure, leading to antibiotic-associated diarrhea. The in vivo toxicity expressed by the pathogen may be associated with T3SS.
Collapse
Affiliation(s)
- Goutam Chowdhury
- Division of Bacteriology, Indian Council of Medical Research - National Institute of Cholera & Enteric Diseases, Kolkata, 700010, India.,Collaborative Research Center of Okayama University for Infectious Diseases, Indian Council of Medical Research - National Institute of Cholera & Enteric Diseases, Kolkata, 700010, India
| | - Bhabatosh Das
- Infection & Immunology Division, Translational Health Science & Technology Institute, Faridabad, 121001, India
| | - Shakti Kumar
- Infection & Immunology Division, Translational Health Science & Technology Institute, Faridabad, 121001, India
| | - Archana Pant
- Infection & Immunology Division, Translational Health Science & Technology Institute, Faridabad, 121001, India
| | - Priyadarshini Mukherjee
- Division of Bacteriology, Indian Council of Medical Research - National Institute of Cholera & Enteric Diseases, Kolkata, 700010, India
| | - Debjani Ghosh
- Division of Bacteriology, Indian Council of Medical Research - National Institute of Cholera & Enteric Diseases, Kolkata, 700010, India
| | - Hemanta Koley
- Division of Bacteriology, Indian Council of Medical Research - National Institute of Cholera & Enteric Diseases, Kolkata, 700010, India
| | - Shin-Ichi Miyoshi
- Collaborative Research Center of Okayama University for Infectious Diseases, Indian Council of Medical Research - National Institute of Cholera & Enteric Diseases, Kolkata, 700010, India.,Graduate School of Medicine, Dentistry & Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Keinosuke Okamoto
- Collaborative Research Center of Okayama University for Infectious Diseases, Indian Council of Medical Research - National Institute of Cholera & Enteric Diseases, Kolkata, 700010, India
| | - Alapan Paul
- Department of Medicine, Nil Ratan Sircar Medical College & Hospital, Kolkata, 700014, India
| | - Shanta Dutta
- Division of Bacteriology, Indian Council of Medical Research - National Institute of Cholera & Enteric Diseases, Kolkata, 700010, India
| | - Thandavarayan Ramamurthy
- Division of Bacteriology, Indian Council of Medical Research - National Institute of Cholera & Enteric Diseases, Kolkata, 700010, India.,Infection & Immunology Division, Translational Health Science & Technology Institute, Faridabad, 121001, India
| | - Asish K Mukhopadyay
- Division of Bacteriology, Indian Council of Medical Research - National Institute of Cholera & Enteric Diseases, Kolkata, 700010, India
| |
Collapse
|
8
|
Wang Y, Chen J, Wang Y, Zheng F, Qu M, Huang Z, Yan J, Bao F, Li X, Sun C, Zheng Y. Cyanidin-3- O-glucoside extracted from the Chinese bayberry ( Myrica rubra Sieb. et Zucc.) alleviates antibiotic-associated diarrhea by regulating gut microbiota and down-regulating inflammatory factors in NF-κB pathway. Front Nutr 2022; 9:970530. [PMID: 36091245 PMCID: PMC9449314 DOI: 10.3389/fnut.2022.970530] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 07/29/2022] [Indexed: 11/13/2022] Open
Abstract
Chinese bayberry has been used to treat diarrhea in China for more than 2,000 years, but the mechanism is not clear. Due to the extensive use of antibiotics, antibiotic-associated diarrhea (AAD) is becoming more and more common in clinic, but there is no effective drug for the treatment. The present study aimed to explore the therapeutic effect of Chinese bayberry on AAD for the first time, and explained the underlying mechanism from different aspects. The BALB/c mice model was established by intragastric administration of lincomycin (3 g/kg). Successfully modeled mice were treated with purified water, dried bayberry powder suspension (100 mg/kg), C3G suspension (40 mg/kg) and montmorillonite powder suspension (40 mg/kg), respectively. The changes of body weight, diarrhea index, diarrhea status score were recorded and calculated regularly. 16S rRNA gene sequencing, intestinal immunofluorescence and inflammatory factor detection were further performed. The treatment with dried bayberry powder suspension and C3G suspension could rapidly reduce the diarrhea score and diarrhea index, increase food intake and restore body weight gain. The gut microbiota richness and diversity were significantly increased after dried bayberry powder suspension and C3G suspension treatments, typically decreased bacterial genera Enterococcus and Clostridium senus stricto 1. In addition, intake of Chinese bayberry powder and C3G significantly decreased the level of p65 phosphorylation, and up-regulated the expression of intestinal tight junction protein claudin-1 and ZO-1. Chinese bayberry fruit had the effect of alleviating AAD, and C3G was supposed to play the predominant role. The mechanism was indicated to be related with restoring the homeostasis of gut microbiota, inhibiting the level of harmful bacteria and increasing the abundance of beneficial bacteria, down-regulating TNF-α, IL-6, and IL-12 factors to reduce inflammation, restoring intestinal tight junction proteins and reducing intestinal permeability.
Collapse
Affiliation(s)
- Yanshuai Wang
- Department of General Surgery, School of Medicine, The Fourth Affiliated Hospital, Zhejiang University, Yiwu, China
| | - Jiebiao Chen
- Laboratory of Fruit Quality Biology, The State Agriculture Ministry Laboratory of Horticultural Plant Growth, Development, and Quality Improvement, Fruit Science Institute, Zhejiang University, Hangzhou, China
| | - Yue Wang
- Laboratory of Fruit Quality Biology, The State Agriculture Ministry Laboratory of Horticultural Plant Growth, Development, and Quality Improvement, Fruit Science Institute, Zhejiang University, Hangzhou, China
| | - Fanghong Zheng
- Department of General Surgery, School of Medicine, The Fourth Affiliated Hospital, Zhejiang University, Yiwu, China
| | - Meiyu Qu
- Department of Pharmacology, School of Medicine, Zhejiang University, Hangzhou, China
| | - Ziwei Huang
- Department of General Surgery, School of Medicine, The Fourth Affiliated Hospital, Zhejiang University, Yiwu, China
| | - Jialang Yan
- Department of General Surgery, School of Medicine, The Fourth Affiliated Hospital, Zhejiang University, Yiwu, China
| | - Fangping Bao
- Department of Anesthesiology, School of Medicine, The Fourth Affiliated Hospital, Zhejiang University, Yiwu, China
| | - Xian Li
- Laboratory of Fruit Quality Biology, The State Agriculture Ministry Laboratory of Horticultural Plant Growth, Development, and Quality Improvement, Fruit Science Institute, Zhejiang University, Hangzhou, China
| | - Chongde Sun
- Laboratory of Fruit Quality Biology, The State Agriculture Ministry Laboratory of Horticultural Plant Growth, Development, and Quality Improvement, Fruit Science Institute, Zhejiang University, Hangzhou, China
| | - Yixiong Zheng
- Department of General Surgery, School of Medicine, The Fourth Affiliated Hospital, Zhejiang University, Yiwu, China
| |
Collapse
|
9
|
Gulliver EL, Young RB, Chonwerawong M, D'Adamo GL, Thomason T, Widdop JT, Rutten EL, Rossetto Marcelino V, Bryant RV, Costello SP, O'Brien CL, Hold GL, Giles EM, Forster SC. Review article: the future of microbiome-based therapeutics. Aliment Pharmacol Ther 2022; 56:192-208. [PMID: 35611465 PMCID: PMC9322325 DOI: 10.1111/apt.17049] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 03/29/2022] [Accepted: 05/12/2022] [Indexed: 12/12/2022]
Abstract
BACKGROUND From consumption of fermented foods and probiotics to emerging applications of faecal microbiota transplantation, the health benefit of manipulating the human microbiota has been exploited for millennia. Despite this history, recent technological advances are unlocking the capacity for targeted microbial manipulation as a novel therapeutic. AIM This review summarises the current developments in microbiome-based medicines and provides insight into the next steps required for therapeutic development. METHODS Here we review current and emerging approaches and assess the capabilities and weaknesses of these technologies to provide safe and effective clinical interventions. Key literature was identified through Pubmed searches with the following key words, 'microbiome', 'microbiome biomarkers', 'probiotics', 'prebiotics', 'synbiotics', 'faecal microbiota transplant', 'live biotherapeutics', 'microbiome mimetics' and 'postbiotics'. RESULTS Improved understanding of the human microbiome and recent technological advances provide an opportunity to develop a new generation of therapies. These therapies will range from dietary interventions, prebiotic supplementations, single probiotic bacterial strains, human donor-derived faecal microbiota transplants, rationally selected combinations of bacterial strains as live biotherapeutics, and the beneficial products or effects produced by bacterial strains, termed microbiome mimetics. CONCLUSIONS Although methods to identify and refine these therapeutics are continually advancing, the rapid emergence of these new approaches necessitates accepted technological and ethical frameworks for measurement, testing, laboratory practices and clinical translation.
Collapse
Affiliation(s)
- Emily L. Gulliver
- Centre for Innate Immunity and Infectious DiseasesHudson Institute of Medical ResearchClaytonVictoriaAustralia,Department of Molecular and Translational SciencesMonash UniversityClaytonVictoriaAustralia
| | - Remy B. Young
- Centre for Innate Immunity and Infectious DiseasesHudson Institute of Medical ResearchClaytonVictoriaAustralia,Department of Molecular and Translational SciencesMonash UniversityClaytonVictoriaAustralia
| | - Michelle Chonwerawong
- Centre for Innate Immunity and Infectious DiseasesHudson Institute of Medical ResearchClaytonVictoriaAustralia,Department of Molecular and Translational SciencesMonash UniversityClaytonVictoriaAustralia
| | - Gemma L. D'Adamo
- Centre for Innate Immunity and Infectious DiseasesHudson Institute of Medical ResearchClaytonVictoriaAustralia,Department of Molecular and Translational SciencesMonash UniversityClaytonVictoriaAustralia
| | - Tamblyn Thomason
- Centre for Innate Immunity and Infectious DiseasesHudson Institute of Medical ResearchClaytonVictoriaAustralia,Department of Molecular and Translational SciencesMonash UniversityClaytonVictoriaAustralia
| | - James T. Widdop
- Centre for Innate Immunity and Infectious DiseasesHudson Institute of Medical ResearchClaytonVictoriaAustralia,Department of Molecular and Translational SciencesMonash UniversityClaytonVictoriaAustralia
| | - Emily L. Rutten
- Centre for Innate Immunity and Infectious DiseasesHudson Institute of Medical ResearchClaytonVictoriaAustralia,Department of Molecular and Translational SciencesMonash UniversityClaytonVictoriaAustralia
| | - Vanessa Rossetto Marcelino
- Centre for Innate Immunity and Infectious DiseasesHudson Institute of Medical ResearchClaytonVictoriaAustralia,Department of Molecular and Translational SciencesMonash UniversityClaytonVictoriaAustralia
| | - Robert V. Bryant
- Department of GastroenterologyThe Queen Elizabeth HospitalWoodvilleSouth AustraliaAustralia,School of MedicineUniversity of AdelaideAdelaideSouth AustraliaAustralia
| | - Samuel P. Costello
- Department of GastroenterologyThe Queen Elizabeth HospitalWoodvilleSouth AustraliaAustralia,School of MedicineUniversity of AdelaideAdelaideSouth AustraliaAustralia
| | | | - Georgina L. Hold
- Microbiome Research Centre, St George & Sutherland Clinical SchoolUniversity of New South WalesSydneyNew South WalesAustralia
| | - Edward M. Giles
- Centre for Innate Immunity and Infectious DiseasesHudson Institute of Medical ResearchClaytonVictoriaAustralia,Department of Molecular and Translational SciencesMonash UniversityClaytonVictoriaAustralia,Department of PaediatricsMonash UniversityClaytonVictoriaAustralia
| | - Samuel C. Forster
- Centre for Innate Immunity and Infectious DiseasesHudson Institute of Medical ResearchClaytonVictoriaAustralia,Department of Molecular and Translational SciencesMonash UniversityClaytonVictoriaAustralia
| |
Collapse
|
10
|
Xu B, Liang S, Zhao J, Li X, Guo J, Xin B, Li B, Huo G, Ma W. Bifidobacterium animalis subsp. lactis XLTG11 improves antibiotic-related diarrhea by alleviating inflammation, enhancing intestinal barrier function and regulating intestinal flora. Food Funct 2022; 13:6404-6418. [PMID: 35616024 DOI: 10.1039/d1fo04305f] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Antibiotic-associated diarrhea (AAD) is a common side effect during antibiotic treatment. In this study, we evaluated the regulatory effect of Bifidobacterium animalis subsp. lactis XLTG11 on mouse diarrhea caused by antibiotic-induced intestinal flora disturbance. Then, two strains of Bifidobacterium animalis subsp. lactis XLTG11 and Bifidobacterium animalis subsp. lactis BB-12 were administered to AAD mice. We found that the recovery effect of using B. lactis XLTG11 was better than that of B. lactis BB-12. B. lactis XLTG11 reduced the pathological characteristics of the intestinal tract, and significantly reduced the levels of lipopolysaccharide (LPS), D-lactic acid (D-LA) and diamine oxidase (DAO) to decrease intestinal permeability. In addition, these two strains significantly increased the expression of aquaporin and tight junction proteins, and inhibited toll-like receptor 4 (TLR4)/activation of the nuclear factor-κB (NF-κB) signaling pathway, significantly increased the levels of anti-inflammatory cytokines and decreased levels of pro-inflammatory cytokines. Moreover, after treatment with B. lactis XLTG11, the contents of acetic acid, propionic acid, butyric acid and total short-chain fatty acids were significantly increased. Compared with the MC group, B. lactis XLTG11 increased the abundance and diversity of the intestinal flora and changed the composition of the intestinal flora. We found that B. lactis XLTG11 can promote the recovery of intestinal flora and mucosal barrier function, thereby effectively improving AAD-related symptoms, providing a scientific basis for future clinical applications.
Collapse
Affiliation(s)
- Baofeng Xu
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin 150030, China. .,Food College, Northeast Agricultural University, Harbin 150030, China. .,Heilongjiang Key Laboratory of Genetic and Metabolic Engineering of Lactic Acid Bacteria, Harbin 150030, China
| | - Shengnan Liang
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin 150030, China. .,Food College, Northeast Agricultural University, Harbin 150030, China. .,Heilongjiang Key Laboratory of Genetic and Metabolic Engineering of Lactic Acid Bacteria, Harbin 150030, China
| | - Jiayi Zhao
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin 150030, China. .,Food College, Northeast Agricultural University, Harbin 150030, China. .,Heilongjiang Key Laboratory of Genetic and Metabolic Engineering of Lactic Acid Bacteria, Harbin 150030, China
| | - Xuetong Li
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin 150030, China. .,Food College, Northeast Agricultural University, Harbin 150030, China. .,Heilongjiang Key Laboratory of Genetic and Metabolic Engineering of Lactic Acid Bacteria, Harbin 150030, China
| | - Jiayao Guo
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin 150030, China. .,Food College, Northeast Agricultural University, Harbin 150030, China. .,Heilongjiang Key Laboratory of Genetic and Metabolic Engineering of Lactic Acid Bacteria, Harbin 150030, China
| | - Bowen Xin
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin 150030, China. .,Food College, Northeast Agricultural University, Harbin 150030, China. .,Heilongjiang Key Laboratory of Genetic and Metabolic Engineering of Lactic Acid Bacteria, Harbin 150030, China
| | - Bailiang Li
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin 150030, China. .,Food College, Northeast Agricultural University, Harbin 150030, China. .,Heilongjiang Key Laboratory of Genetic and Metabolic Engineering of Lactic Acid Bacteria, Harbin 150030, China
| | - Guicheng Huo
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin 150030, China. .,Food College, Northeast Agricultural University, Harbin 150030, China. .,Heilongjiang Key Laboratory of Genetic and Metabolic Engineering of Lactic Acid Bacteria, Harbin 150030, China
| | - Weiwei Ma
- College of Pharmacy, Heilongjiang University of Chinese Medicine, Haerbin 150000, China.
| |
Collapse
|
11
|
Gu X, Sim JX, Lee WL, Cui L, Chan YF, Chang ED, Teh YE, Zhang AN, Armas F, Chandra F, Chen H, Zhao S, Lee Z, Thompson JR, Ooi EE, Low JG, Alm EJ, Kalimuddin S. Gut Ruminococcaceae levels at baseline correlate with risk of antibiotic-associated diarrhea. iScience 2022; 25:103644. [PMID: 35005566 PMCID: PMC8718891 DOI: 10.1016/j.isci.2021.103644] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 11/19/2021] [Accepted: 12/13/2021] [Indexed: 12/11/2022] Open
Abstract
Antibiotic-associated diarrhea (AAD) affects a significant proportion of patients receiving antibiotics. We sought to understand if differences in the gut microbiome would influence the development of AAD. We administered a 3-day course of amoxicillin-clavulanate to 30 healthy adult volunteers, and analyzed their stool microbiome, using 16S rRNA gene sequencing, at baseline and up to 4 weeks post antibiotic administration. Lower levels of gut Ruminococcaceae were significantly and consistently observed from baseline until day 7 in participants who developed AAD. Overall, participants who developed AAD experienced a greater decrease in microbial diversity. The probability of AAD could be predicted based on qPCR-derived levels of Faecalibacterium prausnitzii at baseline. Our findings suggest that a lack of gut Ruminococcaceae influences development of AAD. Quantification of F. prausnitzii in stool prior to antibiotic administration may help identify patients at risk of AAD, and aid clinicians in devising individualized treatment regimens to minimize such adverse effects.
Collapse
Affiliation(s)
- Xiaoqiong Gu
- Antimicrobial Resistance Interdisciplinary Research Group, Singapore-MIT Alliance for Research and Technology, 1 Create Way, Singapore 138602, Singapore
- Campus for Research Excellence and Technological Enterprise (CREATE), Singapore 138602, Singapore
| | - Jean X.Y. Sim
- Department of Infectious Diseases, Singapore General Hospital, Academia Level 3, 20 College Road, Singapore 169856, Singapore
| | - Wei Lin Lee
- Antimicrobial Resistance Interdisciplinary Research Group, Singapore-MIT Alliance for Research and Technology, 1 Create Way, Singapore 138602, Singapore
- Campus for Research Excellence and Technological Enterprise (CREATE), Singapore 138602, Singapore
| | - Liang Cui
- Antimicrobial Resistance Interdisciplinary Research Group, Singapore-MIT Alliance for Research and Technology, 1 Create Way, Singapore 138602, Singapore
- Campus for Research Excellence and Technological Enterprise (CREATE), Singapore 138602, Singapore
| | - Yvonne F.Z. Chan
- Department of Infectious Diseases, Singapore General Hospital, Academia Level 3, 20 College Road, Singapore 169856, Singapore
| | - Ega Danu Chang
- Antimicrobial Resistance Interdisciplinary Research Group, Singapore-MIT Alliance for Research and Technology, 1 Create Way, Singapore 138602, Singapore
- Campus for Research Excellence and Technological Enterprise (CREATE), Singapore 138602, Singapore
| | - Yii Ean Teh
- Department of Infectious Diseases, Singapore General Hospital, Academia Level 3, 20 College Road, Singapore 169856, Singapore
| | - An-Ni Zhang
- Department of Biological Engineering, Massachusetts Institute of Technology, 21 Ames Street, Cambridge, MA 02142, USA
| | - Federica Armas
- Antimicrobial Resistance Interdisciplinary Research Group, Singapore-MIT Alliance for Research and Technology, 1 Create Way, Singapore 138602, Singapore
- Campus for Research Excellence and Technological Enterprise (CREATE), Singapore 138602, Singapore
| | - Franciscus Chandra
- Antimicrobial Resistance Interdisciplinary Research Group, Singapore-MIT Alliance for Research and Technology, 1 Create Way, Singapore 138602, Singapore
- Campus for Research Excellence and Technological Enterprise (CREATE), Singapore 138602, Singapore
| | - Hongjie Chen
- Antimicrobial Resistance Interdisciplinary Research Group, Singapore-MIT Alliance for Research and Technology, 1 Create Way, Singapore 138602, Singapore
- Campus for Research Excellence and Technological Enterprise (CREATE), Singapore 138602, Singapore
| | - Shijie Zhao
- Department of Biological Engineering, Massachusetts Institute of Technology, 21 Ames Street, Cambridge, MA 02142, USA
| | - Zhanyi Lee
- Antimicrobial Resistance Interdisciplinary Research Group, Singapore-MIT Alliance for Research and Technology, 1 Create Way, Singapore 138602, Singapore
- Campus for Research Excellence and Technological Enterprise (CREATE), Singapore 138602, Singapore
| | - Janelle R. Thompson
- Singapore Centre for Environmental Life Sciences Engineering, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore
- Asian School of the Environment, Nanyang Technological University, 62 Nanyang Drive, Singapore 637459, Singapore
| | - Eng Eong Ooi
- Program in Emerging Infectious Diseases, Duke-NUS Medical School, 8 College Road, Singapore 169857, Singapore
- Viral Research and Experimental Medicine Center, SingHealth Duke-NUS Academic Medical Centre (ViREMiCS), 20 College Road, Singapore 169856, Singapore
- Saw Swee Hock School of Public Health, National University of Singapore, 12 Science Drive 2, Singapore 117549, Singapore
| | - Jenny G. Low
- Department of Infectious Diseases, Singapore General Hospital, Academia Level 3, 20 College Road, Singapore 169856, Singapore
- Program in Emerging Infectious Diseases, Duke-NUS Medical School, 8 College Road, Singapore 169857, Singapore
- Viral Research and Experimental Medicine Center, SingHealth Duke-NUS Academic Medical Centre (ViREMiCS), 20 College Road, Singapore 169856, Singapore
| | - Eric J. Alm
- Antimicrobial Resistance Interdisciplinary Research Group, Singapore-MIT Alliance for Research and Technology, 1 Create Way, Singapore 138602, Singapore
- Campus for Research Excellence and Technological Enterprise (CREATE), Singapore 138602, Singapore
- Department of Biological Engineering, Massachusetts Institute of Technology, 21 Ames Street, Cambridge, MA 02142, USA
- Center for Microbiome Informatics and Therapeutics, Massachusetts Institute of Technology, Building E25-321, Cambridge, MA 02139, USA
- Broad Institute of MIT and Harvard, 415 Main Street, Cambridge, MA 02142, USA
| | - Shirin Kalimuddin
- Department of Infectious Diseases, Singapore General Hospital, Academia Level 3, 20 College Road, Singapore 169856, Singapore
- Program in Emerging Infectious Diseases, Duke-NUS Medical School, 8 College Road, Singapore 169857, Singapore
| |
Collapse
|
12
|
Markham NO, Bloch SC, Shupe JA, Laubacher EN, Thomas AK, Kroh HK, Childress KO, Peritore-Galve FC, Washington MK, Coffey RJ, Lacy DB. Murine Intrarectal Instillation of Purified Recombinant Clostridioides difficile Toxins Enables Mechanistic Studies of Pathogenesis. Infect Immun 2021; 89:e00543-20. [PMID: 33468584 PMCID: PMC8090962 DOI: 10.1128/iai.00543-20] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Accepted: 01/09/2021] [Indexed: 12/15/2022] Open
Abstract
Clostridioides difficile is linked to nearly 225,000 antibiotic-associated diarrheal infections and almost 13,000 deaths per year in the United States. Pathogenic strains of C. difficile produce toxin A (TcdA) and toxin B (TcdB), which can directly kill cells and induce an inflammatory response in the colonic mucosa. Hirota et al. (S. A. Hirota et al., Infect Immun 80:4474-4484, 2012) first introduced the intrarectal instillation model of intoxication using TcdA and TcdB purified from VPI 10463 (VPI 10463 reference strain [ATCC 43255]) and 630 C. difficile strains. Here, we expand this technique by instilling purified, recombinant TcdA and TcdB, which allows for the interrogation of how specifically mutated toxins affect tissue. Mouse colons were processed and stained with hematoxylin and eosin for blinded evaluation and scoring by a board-certified gastrointestinal pathologist. The amount of TcdA or TcdB needed to produce damage was lower than previously reported in vivo and ex vivo Furthermore, TcdB mutants lacking either endosomal pore formation or glucosyltransferase activity resemble sham negative controls. Immunofluorescent staining revealed how TcdB initially damages colonic tissue by altering the epithelial architecture closest to the lumen. Tissue sections were also immunostained for markers of acute inflammatory infiltration. These staining patterns were compared to slides from a human C. difficile infection (CDI). The intrarectal instillation mouse model with purified recombinant TcdA and/or TcdB provides the flexibility needed to better understand structure/function relationships across different stages of CDI pathogenesis.
Collapse
Affiliation(s)
- Nicholas O Markham
- Division of Gastroenterology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Epithelial Biology Center, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Sarah C Bloch
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - John A Shupe
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Erin N Laubacher
- Division of Gastroenterology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Epithelial Biology Center, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Audrey K Thomas
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Heather K Kroh
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Kevin O Childress
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - F Christopher Peritore-Galve
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - M Kay Washington
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Robert J Coffey
- Division of Gastroenterology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Epithelial Biology Center, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - D Borden Lacy
- Epithelial Biology Center, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Department of Veterans Affairs Medical Center, Nashville, Tennessee, USA
| |
Collapse
|
13
|
Yang L, Zhang Q, Huang J, Liu D, Lan Y, Yuan L, Chen Q. Xianglian Pill ameliorates antibiotic-associated diarrhea by restoring intestinal microbiota and attenuating mucosal damage. JOURNAL OF ETHNOPHARMACOLOGY 2021; 264:113377. [PMID: 32920136 DOI: 10.1016/j.jep.2020.113377] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Revised: 08/13/2020] [Accepted: 09/06/2020] [Indexed: 06/11/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Xianglian Pill (XLP), a traditional Chinese pharmaceutical preparation for the treatment of gastrointestinal disease, possessing anti-inflammatory, anti-microbial and analgesic activities, may represent a promising candidate for the treatment of antibiotic-associated diarrhea (AAD). AIM OF THE STUDY This study aimed to unravel the underlying mechanism of XLP on the amelioration of AAD. MATERIALS AND METHODS AAD was induced by intragastric administration of a mixture of cefuroxime and levofoxacin (300 mg/kg. bw + 200 mg/kg. bw) for five consecutive days. Then AAD mice were treated with XLP at the dose of 500, 1000 and 2000 mg/kg. bw, respectively for 5 days. The physical manifestations, diarrhea status were monitored during the drug delivery. Histopathology of colon, intestinal microbiota, inflammatory cytokines, tight junction protein and short chain fat acids (SCFAs) were determined. RESULTS Mice received cefuroxime and levofoxacin for 5 days developed medium to severe diarrhea. XLP treatment, however, mitigated the diarrhea status. Further evaluation revealed that XLP promoted the recovery of mucosa, maintained the integrity of tight junction, attenuated the inflammatory disorders, restored intestinal microbiota and increased SCFAs level in feces. CONCLUSION XLP ameliorates AAD by restoring intestinal microbiota and attenuating mucosal damage.
Collapse
Affiliation(s)
- Lujia Yang
- College of Pharmaceutical Sciences, Southwest University, Chongqing, 400715, PR China
| | - Qian Zhang
- College of Pharmaceutical Sciences, Southwest University, Chongqing, 400715, PR China
| | - Jieyao Huang
- College of Pharmaceutical Sciences, Southwest University, Chongqing, 400715, PR China
| | - Danning Liu
- College of Pharmaceutical Sciences, Southwest University, Chongqing, 400715, PR China
| | - Yunfei Lan
- College of Pharmaceutical Sciences, Southwest University, Chongqing, 400715, PR China
| | - Lujiang Yuan
- College of Pharmaceutical Sciences, Southwest University, Chongqing, 400715, PR China.
| | - Qianfeng Chen
- College of Pharmaceutical Sciences, Southwest University, Chongqing, 400715, PR China.
| |
Collapse
|
14
|
Ma ZJ, Wang HJ, Ma XJ, Li Y, Yang HJ, Li H, Su JR, Zhang CE, Huang LQ. Modulation of gut microbiota and intestinal barrier function during alleviation of antibiotic-associated diarrhea with Rhizoma Zingiber officinale (Ginger) extract. Food Funct 2020; 11:10839-10851. [PMID: 33241234 DOI: 10.1039/d0fo01536a] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Antibiotic-associated diarrhea (AAD) is typically mediated by antibiotic therapy, which has increased in prevalence in recent years. Previous studies have suggested that ginger, a common spice and herbal medicine, can modulate the composition of gut microbiota and is beneficial against gastrointestinal disease. This study investigates the therapeutic effects of fresh ginger extract on AAD in a rat model. Gut microbiota and intestinal barrier function were also studied. Ginger was administered to rats with AAD. Diarrhea symptoms were assessed, and 16s rRNA sequencing analysis of gut microbiota was performed. An AAD model was successfully established, and ginger was found to effectively ameliorate AAD-related diarrhea symptoms. After the intervention of ginger decoction, the diversity (rather than richness) of gut microbiota was significantly improved, and the gut microbiota recovery was accelerated. At the genus level, Escherichia_Shigella and Bacteroides levels decreased and increased the most, respectively. Additionally, these changes were demonstrated to be coincidental with the moderate restoration of intestinal barrier function, especially the restoration of tight junction protein ZO-1. Our data indicate that ginger could restore gut microbiota and intestinal barrier function during alleviation of AAD.
Collapse
Affiliation(s)
- Zhi-Jie Ma
- Center for Post-doctoral Research, Resource Center for Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China.
| | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Jaguezeski AM, Glombowsky P, Galli GM, da Rosa G, Araújo DN, Campigotto G, Horn VW, Sareta L, Mendes RE, Da Silva AS. Daily consumption of a homeopathic product decreases intestinal damage and stool bacterial counts in mice challenged with Escherichia coli. Microb Pathog 2020; 147:104269. [PMID: 32439564 DOI: 10.1016/j.micpath.2020.104269] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 04/04/2020] [Accepted: 05/13/2020] [Indexed: 01/16/2023]
Abstract
Escherichia coli is a bacterium normally found in the gastrointestinal tract of domestic animals that can usually control the infection. Nevertheless, some factors (high exposure, stress conditions, animal category, among others) can favor the exacerbation of E. coli infection and cause of disease. Because it is a zoonotic bacterium, it is important to control the infection, avoiding contamination of home interiors in the case of pets. There are various forms of treatment for E. coli; nevertheless, there are few options for prevention. In the present study, we evaluated homeopathy. Thus, the objective of this study was to determine whether administration of a prophylactic homeopathic in water would minimize the negative effects of E. coli infection, as well as reducing bacterial counts in the feces of a experimental model. Forty mice were divided into four experimental groups (n = 10/group). Groups NC (negative control) and PC (positive control) were not treated; in group T1, the animals received 0.002 mL/day/animal of the homeopathic in water, and animals in group T2 0.004 mL/day/animal. The experiment lasted 54 days, and on the 31st day, mice of T1, T2 and PC groups were infected orally a 0.2 mL inoculum of 1.5 × 108 CFU of E. coli. Euthanasia and sample collection were performed on the 40th and 54th days of the experiment (n = 5/group/time point). Blood, liver, spleen, intestine, and feces samples were collected from the final portion of the intestine. There was no significant difference in animal weight between groups at the end of the experiment. Neutrophil count was lower in PC group animals on day 40, while on day 54, the counts were lower in T2 and PC. Lymphocyte counts were lower only in the PC group than in the NC group on day 54. Globulins were lower in the NC and PC groups than in T1 and T2 on day 40, remaining lower the PC group and higher in T1 on day 54; levels of immunoglobulin IgG and IgM were higher in groups T1 and T2, which differed from PC and NC. TNF-α levels were higher in the T1 and T2 groups at 40 and 54 days. INF-γ levels were higher in T1, T2, and PC compared to NC on day 40, remaining higher than NC in groups T1 and T2 on day 54. Total bacterial count, total coliforms and E. coli counts were lower in group T1 and higher in NC and PC on days 40 and 54, when they were lower for T1 and T2. Histologically, no lesions were observed in extra-intestinal tissues; however the height of intestinal crypts in the PC group was smaller than the others on day 40. On day 54, villi and crypts of all infected groups were larger in T1 and T2 than in NC; sizes in the PC group were higher than those of all other groups. These data suggest that the homeopathic agent in the drinking water improved health of the mice.
Collapse
Affiliation(s)
- Antonise M Jaguezeski
- Department of Toxicological Biochemistry, Universidade Federal de Santa Maria (UFSM), Santa Maria, RS, Brazil
| | - Patricia Glombowsky
- Graduate Program in Animal Science, Universidade do Estado de Santa Catarina (UDESC), Chapecó, SC, Brazil
| | - Gabriela M Galli
- Graduate Program in Animal Science, Universidade do Estado de Santa Catarina (UDESC), Chapecó, SC, Brazil
| | - Gilneia da Rosa
- Graduate Program in Animal Science, Universidade do Estado de Santa Catarina (UDESC), Chapecó, SC, Brazil
| | - Denise N Araújo
- Graduate Program in Animal Science, Universidade do Estado de Santa Catarina (UDESC), Chapecó, SC, Brazil; Department of Animal Science, Universidade do Estado de Santa Catarina (UDESC), Chapecó, SC, Brazil
| | - Gabriela Campigotto
- Graduate Program in Animal Science, Universidade do Estado de Santa Catarina (UDESC), Chapecó, SC, Brazil
| | - Vitor W Horn
- Department of Veterinary Pathology, Instituto Federal Catarinense (IFC), Concórdia, SC, Brazil
| | - Laércio Sareta
- Department of Veterinary Pathology, Instituto Federal Catarinense (IFC), Concórdia, SC, Brazil
| | - Ricardo E Mendes
- Department of Veterinary Pathology, Instituto Federal Catarinense (IFC), Concórdia, SC, Brazil
| | - Aleksandro S Da Silva
- Department of Toxicological Biochemistry, Universidade Federal de Santa Maria (UFSM), Santa Maria, RS, Brazil; Graduate Program in Animal Science, Universidade do Estado de Santa Catarina (UDESC), Chapecó, SC, Brazil; Department of Animal Science, Universidade do Estado de Santa Catarina (UDESC), Chapecó, SC, Brazil.
| |
Collapse
|
16
|
Larcombe S, Jiang JH, Hutton ML, Abud HE, Peleg AY, Lyras D. A mouse model of Staphylococcus aureus small intestinal infection. J Med Microbiol 2020; 69:290-297. [PMID: 32004137 PMCID: PMC7431102 DOI: 10.1099/jmm.0.001163] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Introduction Staphylococcus aureus is a recognised cause of foodborne intoxication and antibiotic-associated diarrhoea (AAD), which are both mediated by staphylococcal enterotoxins. However, unlike foodborne intoxication, AAD appears to require infection of the host. While S. aureus intoxication is widely studied, little is known about S. aureus pathogenesis in the context of gastrointestinal infection. Aim To develop a mouse model of S. aureus gastrointestinal infection. Methodology An established AAD mouse model was adapted for S. aureus infection, and damage observed via histopathological analysis and immunostaining of intestinal tissues. Results Various strains colonised the mouse model, and analysis showed that although clinical signs of disease were not seen, S. aureus infection induced damage in the small intestine, disrupting host structures essential for epithelial integrity. Studies using a staphylococcal enterotoxin B mutant showed that this toxin may contribute to damage during gastrointestinal infection. Conclusion This work presents a new mouse model of S. aureus gastrointestinal infection, while also providing insight into the pathogenesis of S. aureus in the gut.
Collapse
Affiliation(s)
- Sarah Larcombe
- Infection and Immunity Program, Monash Biomedicine Discovery Institute and Department of Microbiology, Monash University, Clayton, Victoria, Australia
| | - Jhih-Hang Jiang
- Infection and Immunity Program, Monash Biomedicine Discovery Institute and Department of Microbiology, Monash University, Clayton, Victoria, Australia
| | - Melanie L. Hutton
- Infection and Immunity Program, Monash Biomedicine Discovery Institute and Department of Microbiology, Monash University, Clayton, Victoria, Australia
| | - Helen E. Abud
- Cancer Program, Monash Biomedicine Discovery Institute and Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, Australia
| | - Anton Y. Peleg
- Infection and Immunity Program, Monash Biomedicine Discovery Institute and Department of Microbiology, Monash University, Clayton, Victoria, Australia
- Department of Infectious Diseases, The Alfred Hospital and Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Dena Lyras
- Infection and Immunity Program, Monash Biomedicine Discovery Institute and Department of Microbiology, Monash University, Clayton, Victoria, Australia
- *Correspondence: Dena Lyras,
| |
Collapse
|