1
|
Mukherjee SD, Batagello C, Adler A, Agudelo J, Zampini A, Suryavanshi M, Nguyen A, Orr T, Dearing D, Monga M, Miller AW. Complex system modeling reveals oxalate homeostasis is driven by diverse oxalate-degrading bacteria. eLife 2025; 14:RP104121. [PMID: 40310467 PMCID: PMC12045624 DOI: 10.7554/elife.104121] [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: 05/02/2025] Open
Abstract
Decades of research have made clear that host-associated microbiomes touch all facets of health. However, effective therapies that target the microbiome have been elusive given its inherent complexity. Here, we experimentally examined diet-microbe-host interactions through a complex systems framework, centered on dietary oxalate. Using multiple, independent molecular, rodent, and in vitro experimental models, we found that microbiome composition influenced multiple oxalate-microbe-host interfaces. Importantly, the administration of the oxalate-degrading specialist, Oxalobacter formigenes, was only effective against a poor oxalate-degrading microbiota background and gives critical new insights into why clinical intervention trials with this species exhibit variable outcomes. Data suggest that, while heterogeneity in the microbiome impacts multiple diet-host-microbe interfaces, metabolic redundancy among diverse microorganisms in specific diet-microbe axes is a critical variable that may impact the efficacy of bacteriotherapies, which can help guide patient and probiotic selection criteria in probiotic clinical trials.
Collapse
Affiliation(s)
- Sromona D Mukherjee
- Department of Cardiovascular and Metabolic Sciences, Cleveland ClinicClevelandUnited States
| | - Carlos Batagello
- Division of Urology, Hospital das Clínicas, University of Sao Paulo Medical SchoolSao PauloBrazil
| | - Ava Adler
- Department of Urology, Glickman Urological and Kidney Institute, Cleveland ClinicClevelandUnited States
| | - Jose Agudelo
- Department of Cardiovascular and Metabolic Sciences, Cleveland ClinicClevelandUnited States
| | - Anna Zampini
- Department of Urology, Glickman Urological and Kidney Institute, Cleveland ClinicClevelandUnited States
| | - Mangesh Suryavanshi
- Department of Cardiovascular and Metabolic Sciences, Cleveland ClinicClevelandUnited States
| | - Andrew Nguyen
- M Health Fairview Southdale HospitalEdinaUnited States
| | - Terry Orr
- Department of Biology, New Mexico State UniversityLas CrucesUnited States
| | - Denise Dearing
- School of Biological Sciences, University of UtahSalt Lake CityUnited States
| | - Manoj Monga
- Department of Urology, University of California San DiegoSan DiegoUnited States
| | - Aaron W Miller
- Department of Cardiovascular and Metabolic Sciences, Cleveland ClinicClevelandUnited States
- Department of Urology, Glickman Urological and Kidney Institute, Cleveland ClinicClevelandUnited States
| |
Collapse
|
2
|
Wolfe TM, Jo J, Pinkham NV, Garey KW, Walk ST. The impact of ibezapolstat and other Clostridioides difficile infection-relevant antibiotics on the microbiome of humanized mice. Antimicrob Agents Chemother 2025; 69:e0160424. [PMID: 39998294 PMCID: PMC11963539 DOI: 10.1128/aac.01604-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 01/29/2025] [Indexed: 02/26/2025] Open
Abstract
Ibezapolstat (IBZ) is a competitive inhibitor of the bacterial Pol IIIC enzyme in clinical development for the treatment of Clostridioides difficile infection (CDI). Previous studies demonstrated that IBZ carries a favorable microbiome diversity profile compared to vancomycin (VAN). However, head-to-head comparisons with other CDI antibiotics have not been done. The purpose of this study was to compare microbiome changes associated with IBZ to other clinically used CDI antibiotics. Groups of germ-free (GF) mice received a fecal microbiota transplant from one of two healthy human donors and were subsequently exposed to either IBZ, VAN, fidaxomicin (FDX), metronidazole (MET), or no antibiotic (control). 16S rRNA encoding gene sequencing of temporally collected stool samples was used to compare the gut microbiome perturbations between treatment and no-drug control groups. Among the tested antibiotics, the most significant change in microbiome diversity was observed in MET-treated mice. Each antibiotic had a unique effect, but changes in alpha and beta diversities following FDX- and IBZ-treated groups were less pronounced than those observed in VAN- or MET-treated groups. By the end of therapy, both IBZ and FDZ increased the relative abundance of Bacteroidota (phylum), with IBZ additionally increasing the relative abundance of Actinomycetota (phylum). In microbiome-humanized mice, IBZ and FDX had smaller effects on gut microbiome diversity than VAN and MET. Notable differences were observed between the microbiome of IBZ- and FDX-treated groups, which may allow for differentiation of these two antibiotics in future studies.
Collapse
Affiliation(s)
- Trenton M. Wolfe
- Department of Microbiology and Cell Biology, Montana State University, Bozeman, Montana, USA
| | - Jinhee Jo
- Department of Pharmacy Practice and Translational Research, University of Houston College of Pharmacy, Houston, Texas, USA
| | - Nick V. Pinkham
- Department of Microbiology and Cell Biology, Montana State University, Bozeman, Montana, USA
| | - Kevin W. Garey
- Department of Pharmacy Practice and Translational Research, University of Houston College of Pharmacy, Houston, Texas, USA
| | - Seth T. Walk
- Department of Microbiology and Cell Biology, Montana State University, Bozeman, Montana, USA
| |
Collapse
|
3
|
Ozma MA, Fadaee M, Hosseini HM, Ataee MH, Mirhosseini SA. A Critical Review of Postbiotics as Promising Novel Therapeutic Agents for Clostridial Infections. Probiotics Antimicrob Proteins 2025; 17:656-667. [PMID: 39546182 DOI: 10.1007/s12602-024-10406-0] [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] [Accepted: 11/11/2024] [Indexed: 11/17/2024]
Abstract
Clostridial infections, known for their severity and rapid progression, present significant challenges in both clinical and veterinary fields. These bacteria, which can survive without oxygen and produce protective spores, cause many diseases, ranging from simple gastrointestinal disorders to severe and potentially fatal infections including botulism, tetanus, and gas gangrene. The rising occurrence of antibiotic-resistant strains and the repetitive character of some Clostridial illnesses, including Clostridioides difficile infections (CDI), highlight the immediate need for alternate treatment approaches. Postbiotics, which are metabolites derived from probiotics, are showing great potential as effective agents against these diseases. The current study offers a comprehensive investigation of the potential of postbiotics as therapeutic agents for treating Clostridial infections, including C. difficile, Clostridium perfringens, Clostridium botulinum, and Clostridium tetani. It also examines the processes by which postbiotics exert their effects. Preliminary investigations have shown that postbiotics have promising antibacterial and antibiofilm properties, indicating their potential as adjunct agents in methods for controlling microbial growth. Nevertheless, more study is required to thoroughly demonstrate their medicinal uses.
Collapse
Affiliation(s)
- Mahdi Asghari Ozma
- Student Research Committee, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Manouchehr Fadaee
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hamideh Mahmoodzadeh Hosseini
- Applied Microbiology Research Center, Biomedicine Technologies Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Mohammad Hossein Ataee
- Applied Microbiology Research Center, Biomedicine Technologies Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Seyed Ali Mirhosseini
- Applied Microbiology Research Center, Biomedicine Technologies Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
4
|
Puerta-Alcalde P, O'Keefe J, Woolstencroft R, Kaul S, López N, Cronin K, Lim A, Garcia-Pouton N, Álvarez M, Chee L, Espasa M, Grafia I, Suárez-Lledó M, Smibert O, Garcia-Vidal C, Slavin MA, Yong MK, Soriano A, Worth LJ. Clostridioides difficile infection and recurrence in cancer patients (CIRCA): A multicentre, international study. Int J Infect Dis 2025; 153:107785. [PMID: 39818267 DOI: 10.1016/j.ijid.2025.107785] [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: 11/04/2024] [Revised: 12/21/2024] [Accepted: 01/12/2025] [Indexed: 01/18/2025] Open
Abstract
OBJECTIVES We aimed to describe the characteristics of Clostridioides difficile infection (CDI) in cancer patients, analysing risk factors for 90-day recurrence and attributable mortality. METHODS Retrospective analysis on all CDI episodes from 2020 to 2022 in three Australian hospitals and one Spanish hospital. Logistic regression analyses were performed. RESULTS A total of 547 CDI episodes in cancer patients were documented. Treatment predominantly involved vancomycin (81.5%), followed by metronidazole (15.0%) and fidaxomicin (9.1%). Combined antibiotics were used in 61 (11.2%) episodes. The 90-day recurrence rate was 15.6%. Independent risk factors for CDI recurrence were female sex (OR 2.26, 95% CI 1.13-4.52), age >75 years (OR 2.69, 95% CI 1.30-5.59), dialysis (OR 5.15, 95% CI 1.45-18.27), vomiting at presentation (OR 0.06, 95% CI 0.01-0.55), colonic wall thickening in the CT abdomen (OR 2.42, 95% CI 1.06-5.49) and vancomycin therapy (OR 4.60, 95% CI 1.34-15.84). Overall, 90-day mortality was 22.3%, but attributable mortality was 4.9%. Risk factors for mortality attributed to CDI were age >65 years (OR 15.91, 95% CI 2.64-95.80), previous cerebrovascular disease (OR 20.27, 95% CI 3.12-131.84), antibiotic therapy within the last 30 days (OR 0.17, 95% CI 0.05-0.54), high-output diarrhoea (OR 6.68, 95% CI 1.68-26.56), high CRP-levels (OR 11.60, 95% CI 1.90-70.81) and need for treatment change (OR 6.65, 95% CI 2.20-20.08). CONCLUSIONS CDI recurrence rates among cancer patients remain significant. Nonetheless, fidaxomicin and other preventive strategies are seldom used. We identified several factors that could inform the implementation of these strategies in cancer patients.
Collapse
Affiliation(s)
- Pedro Puerta-Alcalde
- Department of Infectious Diseases, Hospital Clínic-IDIBAPS, Barcelona, Spain; Universitat de Barcelona (UB), Barcelona, Spain.
| | - Jessica O'Keefe
- Victorian Infectious Diseases Service, Royal Melbourne Hospital, Melbourne, VIC, Australia
| | - Rachel Woolstencroft
- Department of Infectious Diseases and National Center for Infection, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Shipraa Kaul
- Department of Infectious Diseases and Microbiology, Austin Health, Melbourne, VIC, Australia
| | - Néstor López
- Department of Infectious Diseases, Hospital Clínic-IDIBAPS, Barcelona, Spain
| | - Katie Cronin
- Microbiology Department, Royal Melbourne Hospital, Melbourne, VIC, Australia
| | - Andrew Lim
- Haematology Department, Austin Health, Melbourne, VIC, Australia
| | | | - Míriam Álvarez
- Microbiology Department, Centre Diagnòstic Biomèdic, Hospital Clínic, Barcelona, Spain
| | - Lynette Chee
- Haematology Department, Royal Melbourne Hospital, Melbourne, and Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Mateu Espasa
- Microbiology Department, Centre Diagnòstic Biomèdic, Hospital Clínic, Barcelona, Spain
| | - Ignacio Grafia
- Medical Oncology Department, Hospital Clínic-IDIBAPS, Barcelona, Spain
| | | | - Olivia Smibert
- Department of Infectious Diseases and Microbiology, Austin Health, Melbourne, VIC, Australia
| | - Carolina Garcia-Vidal
- Department of Infectious Diseases, Hospital Clínic-IDIBAPS, Barcelona, Spain; Universitat de Barcelona (UB), Barcelona, Spain; CIBERINF, CIBER in Infectious Diseases, Spain
| | - Monica A Slavin
- Victorian Infectious Diseases Service, Royal Melbourne Hospital, Melbourne, VIC, Australia; Department of Infectious Diseases and National Center for Infection, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia; National Centre for Infections in Cancer, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, VIC, Australia
| | - Michelle K Yong
- Victorian Infectious Diseases Service, Royal Melbourne Hospital, Melbourne, VIC, Australia; Department of Infectious Diseases and National Center for Infection, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia; National Centre for Infections in Cancer, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Alex Soriano
- Department of Infectious Diseases, Hospital Clínic-IDIBAPS, Barcelona, Spain; Universitat de Barcelona (UB), Barcelona, Spain; CIBERINF, CIBER in Infectious Diseases, Spain
| | - Leon J Worth
- Victorian Infectious Diseases Service, Royal Melbourne Hospital, Melbourne, VIC, Australia; Department of Infectious Diseases and National Center for Infection, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia; National Centre for Infections in Cancer, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, VIC, Australia
| |
Collapse
|
5
|
Fernández-Barrantes C, Ramos-Esquivel A, Hernández-Soto LE, Ramírez-Cardoce M, Garro-Zamora LD, Cordero JC, Grau S. Association Between Antimicrobials and Pump Proton Inhibitors Consumption with the Incidence of Nosocomial Clostridiodes difficile Infection in High Complexity Hospitals in Costa Rica. Antibiotics (Basel) 2025; 14:350. [PMID: 40298558 PMCID: PMC12024204 DOI: 10.3390/antibiotics14040350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2025] [Revised: 03/25/2025] [Accepted: 03/25/2025] [Indexed: 04/30/2025] Open
Abstract
Background: Exposure to antimicrobials and Proton Pump Inhibitors (PPIs) are modifiable risk factors for nosocomial Clostridiodes difficile infection (CDI). We investigated the association between these agents and nosocomial CDI over five years. Methods: Nosocomial CDI from January 2017 to December 2021 were included. Consumption trends were analyzed using a simple linear regression model. A correlation analysis was performed using Spearman's test in two ways: without a time interval and with 1-month interval matching. An interrupted time-series method to evaluate the impact of three key temporal breakpoints on CDI incidence rate was performed using the Poisson regression model. Results: A downward trend for cephalexin, ceftriaxone, clindamycin, gentamicin, macrolides, metronidazole, and penicillin sodium was identified. In contrast, an upward trend was recognized for amoxicillin, ceftazidime/avibactam, ertapenem, fluconazole, ketoconazole, levofloxacin, and tigecycline. Among the antimicrobials that showed a positive association between consumption and the incidence of CDI are clindamycin and cephalosporins after immediate consumption. Moreover, macrolides and metronidazole presented a positive correlation, in both immediate and delayed consumption. PPIs consumption did not show changes and was not associated with nosocomial CDI incidence. The interrupted time series analysis showed no changes at the breakpoints selected. Conclusions: Consumption of clindamycin, cephalosporins, and macrolides showed positive association with CDI, despite having a downtrend in consumption. Specific events, such as the COVID-19 pandemic and the implementation of ASP, have had no correlation with CDI. Further analysis is required in Latin America to advance our understanding of risk factors associated with CDI.
Collapse
Affiliation(s)
- Cristina Fernández-Barrantes
- Department of Pharmacology, Faculty of Medicine, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain;
- Department of Pharmacy, Hospital San Juan de Dios, Caja Costarricense de Seguro Social, San José 10103, Costa Rica
| | | | | | - Manuel Ramírez-Cardoce
- Infectious Diseases Unit, Hospital San Juan de Dios, Caja Costarricense de Seguro Social, San José 10103, Costa Rica;
| | - Luis David Garro-Zamora
- Department of Pharmacy, Hopital México, Caja Costarricense de Seguro Social, San José 10107, Costa Rica;
| | - José Castro Cordero
- Infectious Diseases Unit, Hospital México, Caja Costarricense de Seguro Social, San José 10107, Costa Rica;
| | - Santiago Grau
- Department of Pharmacy, Hospital del Mar, Parc de Salut Mar, 08003 Barcelona, Spain
| |
Collapse
|
6
|
Niu Q, Yang K, Zhou Z, Huang Q, Wang J. Intergenerational Transmission of Gut Microbiome from Infected and Non-Infected Salmonella pullorum Hens. Microorganisms 2025; 13:640. [PMID: 40142532 PMCID: PMC11946299 DOI: 10.3390/microorganisms13030640] [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: 02/20/2025] [Revised: 03/04/2025] [Accepted: 03/07/2025] [Indexed: 03/28/2025] Open
Abstract
Pullorum disease (PD) is one of the common infectious diseases in the poultry industry in the world. Our previous study showed that gut bacterial structure has a significant difference between positive and negative hens. However, the gut bacterial basis of intergenerational transmission of PD continues to elude a scientific explanation. The present study carried out fecal microbiota transplantation (FMT) in chicks of a negative group, then fecal samples of the chicks in the control team (CT), Salmonella pullorum (S. pullorum)-negative transplantation team (PN) and S. pullorum-positive transplantation team (PP) were separately collected to be analyzed for microbial structure and prediction functions. Microbial diversity results revealed that there was a large difference in the gut microbiota of these three groups. Prevotella and Parasutterella with higher abundance in PN (p < 0.05) were transplanted from gut bacteria of S. pullorum-negative hens. Furthermore, the differences of the most major microbial functions (top 100) were similar in hens and chicks, including a pentose phosphate pathway and oxidative phosphorylation. The data provided a reference for exploring the intergenerational transmission and genetic mechanisms of gut microbiota associated with S. pullorum in poultry, as well as a theoretical basis for improving intestinal health through the rational regulation of microbiota-host interactions.
Collapse
Affiliation(s)
- Qing Niu
- Animal Husbandry and Veterinary Research Institute, Shanghai Academy of Agricultural Science, 2901 Beidi Road, Shanghai 201106, China; (Q.N.); (K.Y.); (Z.Z.); (Q.H.)
- Zhuanghang Comprehensive Test Station, Shanghai Academy of Agricultural Science, 888 Yezhuang Road, Shanghai 201415, China
| | - Kaixuan Yang
- Animal Husbandry and Veterinary Research Institute, Shanghai Academy of Agricultural Science, 2901 Beidi Road, Shanghai 201106, China; (Q.N.); (K.Y.); (Z.Z.); (Q.H.)
- Zhuanghang Comprehensive Test Station, Shanghai Academy of Agricultural Science, 888 Yezhuang Road, Shanghai 201415, China
| | - Zhenxiang Zhou
- Animal Husbandry and Veterinary Research Institute, Shanghai Academy of Agricultural Science, 2901 Beidi Road, Shanghai 201106, China; (Q.N.); (K.Y.); (Z.Z.); (Q.H.)
- Zhuanghang Comprehensive Test Station, Shanghai Academy of Agricultural Science, 888 Yezhuang Road, Shanghai 201415, China
| | - Qizhong Huang
- Animal Husbandry and Veterinary Research Institute, Shanghai Academy of Agricultural Science, 2901 Beidi Road, Shanghai 201106, China; (Q.N.); (K.Y.); (Z.Z.); (Q.H.)
- Zhuanghang Comprehensive Test Station, Shanghai Academy of Agricultural Science, 888 Yezhuang Road, Shanghai 201415, China
| | - Junliang Wang
- Zhuanghang Comprehensive Test Station, Shanghai Academy of Agricultural Science, 888 Yezhuang Road, Shanghai 201415, China
| |
Collapse
|
7
|
Gregory AL, Bussan HE, Topf MA, Hryckowian AJ. Impacts of perR on oxygen sensitivity, gene expression, and murine infection in Clostridioides difficile 630∆ erm. J Bacteriol 2025; 207:e0046824. [PMID: 39846733 PMCID: PMC11841134 DOI: 10.1128/jb.00468-24] [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: 10/28/2024] [Accepted: 12/23/2024] [Indexed: 01/24/2025] Open
Abstract
Clostridioides difficile infection (CDI), characterized by colitis and diarrhea, afflicts approximately half a million people in the USA every year, burdening both individuals and the healthcare system. C. difficile 630Δerm is an erythromycin-sensitive variant of the clinical isolate C. difficile 630 and is commonly used in the C. difficile research community due to its genetic tractability. 630Δerm possesses a point mutation in perR, an autoregulated transcriptional repressor that regulates oxidative stress resistance genes. This point mutation results in a constitutively de-repressed PerR operon in 630Δerm. To address the impacts of perR on phenotypes relevant for oxygen tolerance and relevant to a murine model of CDI, we corrected the point mutant to restore PerR function in 630∆erm (herein, 630∆erm perRWT). We demonstrate that there is no difference in growth between 630Δerm and 630Δerm perRWT under anaerobic conditions or when exposed to concentrations of O2 that mimic those found near the surface of the colonic epithelium. However, 630∆erm perRWT is more sensitive to ambient oxygen than 630∆erm, which coincides with alterations in expression of a variety of perR-dependent and perR-independent genes. Finally, we show that 630∆erm and 630∆erm perRWT do not differ in their ability to infect and cause disease in a well-established murine model of CDI. Together, these data support the hypothesis that the perR mutation in 630∆erm arose as a result of exposure to ambient oxygen and that the perR mutation in 630∆erm is unlikely to impact CDI-relevant phenotypes in laboratory studies.IMPORTANCEClostridioides difficile is a diarrheal pathogen and a major public health concern. To improve humans' understanding of C. difficile, a variety of C. difficile isolates are used in research, including C. difficile 630Δerm. 630Δerm is a derivative of the clinical isolate 630 and is commonly studied because it is genetically manipulable. Previous work showed that a mutation in perR in 630Δerm results in a dysregulated oxidative stress response, but no work has been done to characterize perR-dependent effects on the transcriptome or to determine impacts of perR during infection. Here, we identify transcriptomic differences between 630∆erm and 630∆erm perRWT exposed to ambient oxygen and demonstrate that there is no strain-based difference in burdens in murine C. difficile infection.
Collapse
Affiliation(s)
- Anna L. Gregory
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
- Department of Medical Microbiology & Immunology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
- Microbiology Doctoral Training Program, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Hailey E. Bussan
- Microbiology Doctoral Training Program, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Department of Bacteriology, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Madeline A. Topf
- Department of Medical Microbiology & Immunology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
- Microbiology Doctoral Training Program, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Andrew J. Hryckowian
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
- Department of Medical Microbiology & Immunology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| |
Collapse
|
8
|
Mukherjee SD, Batagello CA, Adler A, Agudelo J, Zampini A, Suryavanshi M, Nguyen A, Orr T, Dearing D, Monga M, Miller AW. Complex system modelling reveals oxalate homeostasis is driven by diverse oxalate-degrading bacteria. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.10.28.620613. [PMID: 39553961 PMCID: PMC11565779 DOI: 10.1101/2024.10.28.620613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2024]
Abstract
Decades of research have made clear that host-associated microbiomes touch all facets of health. However, effective therapies that target the microbiome have been elusive given its inherent complexity. Here, we experimentally examined diet-microbe-host interactions through a complex systems framework, centered on dietary oxalate. Using multiple, independent molecular, animal, and in vitro experimental models, we found that microbiome composition influenced multiple oxalate-microbe-host interfaces. Importantly, administration of the oxalate-degrading specialist, Oxalobacter formigenes, was only effective against a poor oxalate-degrading microbiota background and gives critical new insights into why clinical intervention trials with this species exhibit variable outcomes. Data suggest that, while heterogeneity in the microbiome impacts multiple diet-host-microbe interfaces, metabolic redundancy among diverse microorganisms in specific diet-microbe axes is a critical variable that may impact the efficacy of bacteriotherapies, which can help guide patient and probiotic selection criteria in probiotic clinical trials.
Collapse
Affiliation(s)
- Sromona D. Mukherjee
- Department of Cardiovascular and Metabolic Sciences, Cleveland Clinic, Cleveland, OH, USA
| | - Carlos A. Batagello
- Division of Urology, Hospital das Clínicas, University of Sao Paulo Medical School, Sao Paulo, Brazil
| | - Ava Adler
- Department of Urology, Glickman Urological and Kidney Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Jose Agudelo
- Department of Cardiovascular and Metabolic Sciences, Cleveland Clinic, Cleveland, OH, USA
| | - Anna Zampini
- Department of Urology, Glickman Urological and Kidney Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Mangesh Suryavanshi
- Department of Cardiovascular and Metabolic Sciences, Cleveland Clinic, Cleveland, OH, USA
| | - Andrew Nguyen
- M Health Fairview Southdale Hospital, Edina, MN, USA
| | - Teri Orr
- Department of Biology, New Mexico State University, Las Cruces, NM, USA
| | - Denise Dearing
- School of Biological Sciences, University of Utah, Salt Lake City, UT, USA
| | - Manoj Monga
- Department of Urology, University of California San Diego, San Diego, CA, USA
| | - Aaron W. Miller
- Department of Cardiovascular and Metabolic Sciences, Cleveland Clinic, Cleveland, OH, USA
- Department of Urology, Glickman Urological and Kidney Institute, Cleveland Clinic, Cleveland, OH, USA
| |
Collapse
|
9
|
Yang JL, Zhu H, Sadh P, Aumiller K, Guvener ZT, Ludington WB. Commensal acidification of specific gut regions produces a protective priority effect against enteropathogenic bacterial infection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.12.637843. [PMID: 39990475 PMCID: PMC11844456 DOI: 10.1101/2025.02.12.637843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/25/2025]
Abstract
The commensal microbiome has been shown to protect against newly introduced enteric pathogens in multiple host species, a phenomenon known as a priority effect. Multiple mechanisms can contribute to this protective priority effect, including antimicrobial compounds, nutrient competition, and pH changes. In Drosophila melanogaster , Lactiplantibacillus plantarum has been shown to protect against enteric pathogens. However, the strains of L. plantarum studied were derived from laboratory flies or non-fly environments and have been found to be unstable colonizers of the fly gut that mainly reside on the food. To study the priority effect using a naturally occurring microbial relationship, we isolated a wild-fly derived strain of L. plantarum that stably colonizes the fly gut in conjunction with a common enteric pathogen, Serratia marcescens . Flies stably associated with the L. plantarum strain were more resilient to oral Serratia marcescens infection as seen by longer lifespan and lower S. marcescens load in the gut. Through in vitro experiments, we found that L. plantarum inhibits S. marcescens growth due to acidification. We used gut imaging with pH-indicator dyes to show that L. plantarum reduces the gut pH to levels that restrict S. marcescens growth in vivo . In flies colonized with L. plantarum prior to S. marcescens infection, L. plantarum and S. marcescens are spatially segregated in the gut and S. marcescens is less abundant where L. plantarum heavily colonizes, indicating that acidification of specific gut regions is a mechanism of a protective priority effect.
Collapse
|
10
|
Henigman U, Kušar D, Biasizzo M, Vadnjal S, Avberšek J, Papić B. Genomic analysis reveals the presence of hypervirulent and fluoroquinolone-resistant Clostridioides difficile in farmed mussels (Mytilus galloprovincialis) in Slovenia. Int J Food Microbiol 2025; 428:110982. [PMID: 39580990 DOI: 10.1016/j.ijfoodmicro.2024.110982] [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: 09/12/2024] [Revised: 11/13/2024] [Accepted: 11/13/2024] [Indexed: 11/26/2024]
Abstract
Clostridioides difficile is one of the leading causes of antibiotic-associated diarrhea. In this study, we characterized 76C. difficile isolates, obtained from three Mediterranean mussel (Mytilus galloprovincialis) farms in Slovenia from November 2014 to October 2015 (sampling period 1) and from January to December 2021 (sampling period 2). The overall isolation rate of C. difficile from all the examined mussels was 59.8 %. A statistically significant trend of seasonal variation was observed, with a higher isolation rate in the colder months of the year (87.9 %; sea temperature ≤ 15 °C) compared with the warmer months (31.8 %; sea temperature > 15 °C). Whole-genome sequencing (WGS) revealed that the isolates belonged to 31 different sequence types (STs), which were associated with three clades (1, 4, and 5) and two cryptic clades (C-II and C-III). Five isolates, which belonged to ST11 (clade 5), harbored all the main toxin genes (A+B+CDT+) and chromosomal mutations conferring fluoroquinolone resistance. Core genome multilocus sequence typing (cgMLST) revealed four clusters of 2-3 isolates, three of which included isolates from different farms, suggesting that clonal C. difficile strains are circulating among the Slovenian mussel farms. The results highlight the presence of hypervirulent strains in mussels; therefore, at-risk population groups should be alerted to the risks associated with consuming shellfish.
Collapse
Affiliation(s)
- Urška Henigman
- University of Ljubljana, Veterinary Faculty, National Veterinary Institute, Institute of Food Safety, Feed and Environment, Gerbičeva 60, 1000 Ljubljana, Slovenia.
| | - Darja Kušar
- University of Ljubljana, Veterinary Faculty, Institute of Microbiology and Parasitology, Gerbičeva 60, 1000 Ljubljana, Slovenia
| | - Majda Biasizzo
- University of Ljubljana, Veterinary Faculty, National Veterinary Institute, Institute of Food Safety, Feed and Environment, Gerbičeva 60, 1000 Ljubljana, Slovenia
| | - Stanka Vadnjal
- University of Ljubljana, Veterinary Faculty, National Veterinary Institute, Institute of Food Safety, Feed and Environment, Gerbičeva 60, 1000 Ljubljana, Slovenia
| | - Jana Avberšek
- University of Ljubljana, Veterinary Faculty, Institute of Microbiology and Parasitology, Gerbičeva 60, 1000 Ljubljana, Slovenia
| | - Bojan Papić
- University of Ljubljana, Veterinary Faculty, Institute of Microbiology and Parasitology, Gerbičeva 60, 1000 Ljubljana, Slovenia
| |
Collapse
|
11
|
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
|
12
|
Shiraishi C, Kato H, Ogura T, Iwamoto T. Association between age and onset of daptomycin-induced adverse events using the U.S. food and drug administration adverse event reporting system. J Infect Chemother 2025; 31:102501. [PMID: 39209260 DOI: 10.1016/j.jiac.2024.08.016] [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: 06/06/2024] [Revised: 08/20/2024] [Accepted: 08/25/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND Daptomycin is a lipopeptide antibiotic with a broad spectrum of activity against gram-positive bacteria. Although information on daptomycin-induced adverse events can be found in clinical trials, data regarding the impact of age on these events are insufficient. Therefore, we evaluated whether age affects the occurrence of daptomycin-induced adverse events using adverse drug event reports in post-marketing stages provided by the U.S. Food and Drug Administration (FDA). METHODS A total dataset of 7307 reports of patients treated with daptomycin in the FDA's Adverse Event Reporting System were analyzed. The patients were divided into seven age groups: 0-28 days, >28 days-23 months, 2-11 years, 12-17 years, 18-64 years, 65-80 years, and >80 years. A disproportionality analysis was conducted to calculate the reporting odds ratio, with a 95 % confidence interval. The univariate regression analysis was conducted using the percentage of each adverse event and age groups. RESULTS Compared with the number of reports aged 18-64 years, there were significantly increased reports of eosinophilic pneumonia in patients aged 65-80 years and >80 years, anaphylactic reaction and pseudomembranous colitis in patients aged 12-17 years, and acute renal failure in patients aged 65-80 years. The regression coefficient for the reporting proportion of eosinophilic pneumonia was significantly positive. CONCLUSIONS Our findings revealed age-related trends in daptomycin-induced adverse events, supporting the idea that implementing age-dependent follow-up and supportive care helps in the continuation of daptomycin therapy.
Collapse
Affiliation(s)
- Chihiro Shiraishi
- Department of Pharmacy, Mie University Hospital, Mie, Japan; Department of Clinical Pharmaceutics, Division of Clinical Medical Science, Mie University Graduate School of Medicine, Mie, Japan
| | - Hideo Kato
- Department of Pharmacy, Mie University Hospital, Mie, Japan; Department of Clinical Pharmaceutics, Division of Clinical Medical Science, Mie University Graduate School of Medicine, Mie, Japan.
| | - Toru Ogura
- Clinical Research Support Center, Mie University Hospital, Mie, Japan
| | - Takuya Iwamoto
- Department of Pharmacy, Mie University Hospital, Mie, Japan; Department of Clinical Pharmaceutics, Division of Clinical Medical Science, Mie University Graduate School of Medicine, Mie, Japan
| |
Collapse
|
13
|
Morado F, Nanda N. A Review of Therapies for Clostridioides difficile Infection. Antibiotics (Basel) 2024; 14:17. [PMID: 39858303 PMCID: PMC11762378 DOI: 10.3390/antibiotics14010017] [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: 11/25/2024] [Revised: 12/20/2024] [Accepted: 12/23/2024] [Indexed: 01/27/2025] Open
Abstract
Clostridioides difficile is an urgent public health threat that affects approximately half a million patients annually in the United States. Despite concerted efforts aimed at the prevention of Clostridioides difficile infection (CDI), it remains a leading cause of healthcare-associated infections. CDI is associated with significant clinical, social, and economic burdens. Therefore, it is imperative to provide optimal and timely therapy for CDI. We conducted a systematic literature review and offer treatment recommendations based on available evidence for the treatment and prevention of CDI.
Collapse
Affiliation(s)
- Faiza Morado
- Department of Pharmacy, Keck Medical Center, University of Southern California, Los Angeles, CA 90033, USA;
| | - Neha Nanda
- Division of Infectious Diseases, Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| |
Collapse
|
14
|
Pelet Del Toro NM, Strunk A, Wu JJ, Stein Gold L, Del Rosso JQ, Brodell RT, Han G. Topical clindamycin for acne vulgaris: analysis of gastrointestinal events. J DERMATOL TREAT 2024; 35:2325603. [PMID: 38568005 DOI: 10.1080/09546634.2024.2325603] [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: 12/19/2023] [Accepted: 02/25/2024] [Indexed: 04/05/2024]
Abstract
Purpose: Topical clindamycin, a lincosamide antibiotic, is commonly combined with benzoyl peroxide or a retinoid for acne vulgaris (AV) treatment. While oral and topical clindamycin carry warnings/contraindications regarding gastrointestinal (GI) adverse events (AEs), real-world incidence of GI AEs with topical clindamycin is unknown. This review provides background information and an overview of safety data of topical clindamycin for treating AV.Materials and Methods: Available safety data from published literature, previously unpublished worldwide pharmacovigilance data, and two retrospective cohort studies were reviewed.Results and Conclusions: According to pharmacovigilance data, the rate of GI adverse drug reactions with topical clindamycin-containing products was 0.000045% (64/141,084,533). Results from two retrospective medical record studies of patients with AV indicated that physicians prescribe topical clindamycin equally to patients with or without inflammatory bowel disease history, and that rates of pseudomembranous colitis in these patients were low. In 8 published pivotal clinical trials of topical clindamycin for AV, GI AEs were reported in 1.4% of participants. Limitations include under/inaccurate reporting of AEs or prescription data and limited generalizability. This review of published case reports, worldwide pharmacovigilance data, retrospective US prescription data, and clinical trials safety data demonstrates that the incidence of colitis in patients exposed to topical clindamycin is extremely low.
Collapse
Affiliation(s)
- Natalia M Pelet Del Toro
- Department of Dermatology, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, New York, NY, USA
| | - Andrew Strunk
- Department of Dermatology, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, New York, NY, USA
| | - Jashin J Wu
- Department of Dermatology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Linda Stein Gold
- Department of Dermatology, Henry Ford Hospital, Detroit, MI, USA
| | - James Q Del Rosso
- JDR Dermatology Research/Thomas Dermatology, Las Vegas, NV, USA
- Advanced Dermatology and Cosmetic Surgery, Maitland, FL, USA
- Department of Dermatology, Touro University Nevada, Henderson, NV, USA
| | - Robert T Brodell
- Department of Dermatology, University of Mississippi Medical Center, Jackson, MS, USA
| | - George Han
- Department of Dermatology, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, New York, NY, USA
| |
Collapse
|
15
|
Cabezas-Cruz A, Bermúdez-Humarán LG. Exploring the relationship between Faecalibacterium duncaniae and Escherichia coli in inflammatory bowel disease (IBD): Insights and implications. Comput Struct Biotechnol J 2024; 23:1-9. [PMID: 38094217 PMCID: PMC10716368 DOI: 10.1016/j.csbj.2023.11.027] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 11/14/2023] [Accepted: 11/14/2023] [Indexed: 03/04/2025] Open
Abstract
Inflammatory bowel disease (IBD) is a group of disorders characterized by an inflammation of the gastrointestinal tract (GIT) and represents a major social and economic burden. Despite ongoing research into the etiology and pathophysiology of this multifactorial disease, treatment options remain limited. From this perspective, the gut microbiota has emerged as a potential player in the pathogenesis of IBD, and animal and human studies support this hypothesis. Indeed, the human gut is one of the most complex ecological communities (composed of 1013-1014 microorganisms) that plays a critical role in human health by influencing normal physiology and disease susceptibility through its collective metabolic activities and host interactions. In addition, live probiotic bacteria present in some food products (which transit through the GIT) have been shown to interact with the host immune system and confer several health benefits. The aim of this review is to provide an overview of the link between Faecalibacterium duncaniae and Escherichia coli and IBD, highlighting the main areas of research in this field. An ecological perspective on the gut microbiota may offer new insights for the development of clinical therapies targeting this bacterial community to improve human health.
Collapse
Affiliation(s)
- Alejandro Cabezas-Cruz
- Anses, INRAE, Ecole Nationale Vétérinaire d’Alfort, UMR BIPAR, Laboratoire de Santé Animale, Maisons-Alfort F-94700, France
| | | |
Collapse
|
16
|
Wolfe TM, Jo J, Pinkham NV, Garey KW, Walk ST. Microbiome impact of ibezapolstat and other Clostridioides difficile infection relevant antibiotics using humanized mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.06.622322. [PMID: 39574673 PMCID: PMC11580883 DOI: 10.1101/2024.11.06.622322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Background Ibezapolstat (IBZ) is a competitive inhibitor of the bacterial Pol IIIC enzyme in clinical development for treatment of Clostridioides difficile infection (CDI). Previous studies demonstrated IBZ carries a favorable microbiome diversity profile compared to vancomycin (VAN). However, head-to-head comparisons with other CDI antibiotics have not been done. The purpose of this study was to compare microbiome changes associated with IBZ to other clinically used CDI antibiotics. Methods Groups of germ-free (GF) mice received a fecal microbiota transplant from one of two healthy human donors and were subsequently exposed to either IBZ, VAN, fidaxomicin (FDX), metronidazole (MET), or no antibiotic (control). 16S rRNA encoding gene sequencing of temporally collected stool samples was used to compare gut microbiome perturbation between treatment and no-drug control groups. Results Among the tested antibiotics, the most significant change in microbiome diversity was observed in MET-treated mice. Each antibiotic had a unique effect, but changes in alpha and beta diversity following FDX- and IBZ-treated groups were less pronounced compared to those observed in VAN-or MET-treated groups. By the end of therapy, both IBZ and FDZ increased the relative abundance of Bacteroidota (phylum), with IBZ additionally increasing the relative abundance of Actinomycetota (phylum). Conclusion In microbiome-humanized mice, IBZ and FDX had smaller effects on gut microbiome diversity compared to VAN and MET. Notable differences were observed between the microbiome of IBZ- and FDX-treated groups, which may allow for differentiation of these two antibiotics in future studies.
Collapse
|
17
|
Van Den Ham KM, Bower LK, Li S, Lorenzi H, Doumbo S, Doumtabe D, Kayentao K, Ongoiba A, Traore B, Crompton PD, Schmidt NW. The gut microbiome is associated with susceptibility to febrile malaria in Malian children. Nat Commun 2024; 15:9525. [PMID: 39500866 PMCID: PMC11538534 DOI: 10.1038/s41467-024-52953-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Accepted: 09/23/2024] [Indexed: 11/08/2024] Open
Abstract
Malaria is a major public health problem, but many of the factors underlying the pathogenesis of this disease are not well understood, including protection from the development of febrile symptoms, which is observed in individuals residing in areas with moderate-to-high transmission by early adolescence. Here, we demonstrate that susceptibility to febrile malaria following Plasmodium falciparum infection is associated with the composition of the gut microbiome prior to the malaria season in 10-year-old Malian children, but not in younger children. Gnotobiotic mice colonized with the fecal samples of malaria-susceptible children were shown to have a significantly higher parasite burden following Plasmodium infection compared to gnotobiotic mice colonized with the fecal samples of malaria-resistant children. The fecal microbiome of the susceptible children was determined to be enriched for bacteria associated with inflammation, mucin degradation and gut permeability, and to have increased levels of nitric oxide-derived DNA adducts and lower levels of mucus phospholipids compared to the resistant children. Overall, these results indicate that the composition of the gut microbiome is associated with the prospective risk of febrile malaria in Malian children and suggest that modulation of the gut microbiome could decrease malaria morbidity in endemic areas.
Collapse
Affiliation(s)
- Kristin M Van Den Ham
- Ryan White Center for Pediatric Infectious Diseases and Global Health, Herman B Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Layne K Bower
- Ryan White Center for Pediatric Infectious Diseases and Global Health, Herman B Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Shanping Li
- Malaria Infection Biology and Immunity Section, Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Hernan Lorenzi
- Infectious Diseases Group, J. Craig Venter Institute, Bethesda, MD, USA
| | - Safiatou Doumbo
- Mali International Center of Excellence in Research; Malaria Research and Training Center, University of Sciences, Techniques and Technologies of Bamako, Bamako, Mali
| | - Didier Doumtabe
- Mali International Center of Excellence in Research; Malaria Research and Training Center, University of Sciences, Techniques and Technologies of Bamako, Bamako, Mali
| | - Kassoum Kayentao
- Mali International Center of Excellence in Research; Malaria Research and Training Center, University of Sciences, Techniques and Technologies of Bamako, Bamako, Mali
| | - Aissata Ongoiba
- Mali International Center of Excellence in Research; Malaria Research and Training Center, University of Sciences, Techniques and Technologies of Bamako, Bamako, Mali
| | - Boubacar Traore
- Mali International Center of Excellence in Research; Malaria Research and Training Center, University of Sciences, Techniques and Technologies of Bamako, Bamako, Mali
| | - Peter D Crompton
- Malaria Infection Biology and Immunity Section, Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Nathan W Schmidt
- Ryan White Center for Pediatric Infectious Diseases and Global Health, Herman B Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA.
| |
Collapse
|
18
|
Khan IM, Nassar N, Chang H, Khan S, Cheng M, Wang Z, Xiang X. The microbiota: a key regulator of health, productivity, and reproductive success in mammals. Front Microbiol 2024; 15:1480811. [PMID: 39633815 PMCID: PMC11616035 DOI: 10.3389/fmicb.2024.1480811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Accepted: 10/15/2024] [Indexed: 12/07/2024] Open
Abstract
The microbiota, intensely intertwined with mammalian physiology, significantly impacts health, productivity, and reproductive functions. The normal microbiota interacts with the host through the following key mechanisms: acting as a protective barrier against pathogens, maintain mucosal barrier integrity, assisting in nutrient metabolism, and modulating of the immune response. Therefore, supporting growth and development of host, and providing protection against pathogens and toxic substances. The microbiota significantly influences brain development and behavior, as demonstrated by comprehensive findings from controlled laboratory experiments and human clinical studies. The prospects suggested that gut microbiome influence neurodevelopmental processes, modulate stress responses, and affect cognitive function through the gut-brain axis. Microbiota in the gastrointestinal tract of farm animals break down and ferment the ingested feed into nutrients, utilize to produce meat and milk. Among the beneficial by-products of gut microbiota, short-chain fatty acids (SCFAs) are particularly noteworthy for their substantial role in disease prevention and the promotion of various productive aspects in mammals. The microbiota plays a pivotal role in the reproductive hormonal systems of mammals, boosting reproductive performance in both sexes and fostering the maternal-infant connection, thereby becoming a crucial factor in sustaining mammalian existence. The microbiota is a critical factor influencing reproductive success and production traits in mammals. A well-balanced microbiome improves nutrient absorption and metabolic efficiency, leading to better growth rates, increased milk production, and enhanced overall health. Additionally, it regulates key reproductive hormones like estrogen and progesterone, which are essential for successful conception and pregnancy. Understanding the role of gut microbiota offers valuable insights for optimizing breeding and improving production outcomes, contributing to advancements in agriculture and veterinary medicine. This study emphasizes the critical ecological roles of mammalian microbiota, highlighting their essential contributions to health, productivity, and reproductive success. By integrating human and veterinary perspectives, it demonstrates how microbial communities enhance immune function, metabolic processes, and hormonal regulation across species, offering insights that benefit both clinical and agricultural advancements.
Collapse
Affiliation(s)
| | - Nourhan Nassar
- College of Life Science, Anhui Agricultural University, Hefei, China
- Department of Clinical Pathology, Faculty of Veterinary Medicine, Benha University, Moshtohor, Egypt
| | - Hua Chang
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming, China
| | - Samiullah Khan
- The Scientific Observing and Experimental Station of Crop Pest in Guiyang, Ministry of Agriculture, Institute of Entomology, Guizhou University, Guiyang, China
| | - Maoji Cheng
- Fisugarpeptide Biology Engineering Co. Ltd., Lu’an, China
| | - Zaigui Wang
- College of Life Science, Anhui Agricultural University, Hefei, China
| | - Xun Xiang
- College of Animal Science and Technology, Yunnan Agricultural University, Kunming, China
| |
Collapse
|
19
|
Kwoji ID, Okpeku M, Aiyegoro OA, Adeleke MA. Metabolic interactions of Limosilactobacillus reuteri ZJ625 and Ligilactobacillus salivarius ZJ614 in co-culture: implications for multi-strain probiotics. J Appl Microbiol 2024; 135:lxae264. [PMID: 39510973 DOI: 10.1093/jambio/lxae264] [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: 02/18/2024] [Revised: 08/31/2024] [Accepted: 11/05/2024] [Indexed: 11/15/2024]
Abstract
AIMS Limosilactobacillus reuteri ZJ625 and Ligilactobacillus salivarius ZJ614 are potential probiotic bacteria. The mechanisms of enhanced benefits by muti-strain probiotics are yet fully understood. We elucidated the influence of co-culturing on the metabolite profiles of Limosilactobacillus reuteri ZJ625 and Ligilactobacillus salivarius ZJ614 to decipher the impacts of co-culturing on metabolic interactions between the strains. METHODS AND RESULTS Limosilactobacillus reuteri ZJ625 and Ligilactobacillus salivarius ZJ614 were grown in single and co-cultures in defined media. Bacterial cell metabolites were extracted at the mid-stationary growth phase and analysed using two-dimensional gas chromatography-time-of-flight mass spectrometry (GC × GC-TOFMS). Mass-spectral data were preprocessed and analysed using unsupervised and supervised methods based on the group allocations. A total of 1387 metabolites were identified, with 18.31% significant metabolites (P < 0.05) and 10.17% differential metabolites (P < 0.05, variable importance on projection > 1). The differential metabolites identified include arabinofuranose, methyl-galactoside, N-acetylglutamic acid, phosphoric acid, and decanoic acid. The metabolites impacted carbohydrate and amino-sugar metabolism. CONCLUSION Co-culturing of Limosilactobacillus reuteri ZJ625 and Ligilactobacillus salivarius ZJ614 influenced the metabolite profiles of the strains and impacted metabolic/biosynthetic pathways, indicating cell-to-cell interactions between the strains.
Collapse
Affiliation(s)
- Iliya Dauda Kwoji
- Discipline of Genetics, School of Life Sciences, College of Agriculture, Engineering and Sciences, University of KwaZulu-Natal, 4090 Durban, South Africa
| | - Moses Okpeku
- Discipline of Genetics, School of Life Sciences, College of Agriculture, Engineering and Sciences, University of KwaZulu-Natal, 4090 Durban, South Africa
| | - Olayinka Ayobami Aiyegoro
- Unit for Environmental Sciences and Management, Northwest University, Potchefstroom, Northwest 2520, South Africa
| | - Matthew Adekunle Adeleke
- Discipline of Genetics, School of Life Sciences, College of Agriculture, Engineering and Sciences, University of KwaZulu-Natal, 4090 Durban, South Africa
| |
Collapse
|
20
|
Gregory AL, Bussan HE, Topf MA, Hryckowian AJ. Impacts of perR on oxygen sensitivity, gene expression, and murine infection in Clostridioides difficile 630Δ erm. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.30.621113. [PMID: 39554039 PMCID: PMC11565800 DOI: 10.1101/2024.10.30.621113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/19/2024]
Abstract
Clostridioides difficile infection (CDI), characterized by colitis and diarrhea, afflicts approximately half a million people in the United States every year, burdening both individuals and the healthcare system. C. difficile 630Δerm is an erythromycin-sensitive variant of the clinical isolate C. difficile 630 and is commonly used in the C. difficile research community due to its genetic tractability. 630Δerm possesses a point mutation in perR, an autoregulated transcriptional repressor that regulates oxidative stress resistance genes. This point mutation results in a constitutively de-repressed PerR operon in 630Δerm. To address the impacts of perR on phenotypes relevant for oxygen tolerance and relevant to a murine model of CDI, we corrected the point mutant to restore PerR function in 630Δerm (herein, 630Δerm perR WT). We demonstrate that there is no difference in growth between 630Δerm and a 630Δerm perR WT under anaerobic conditions or when exposed to concentrations of O2 that mimic those found near the surface of the colonic epithelium. However, 630Δerm perR WT is more sensitive to ambient oxygen than 630Δerm, which coincides with alterations in expression of a variety of perR-dependent and perR-independent genes. Finally, we show that 630Δerm and 630Δerm perR WT do not differ in their ability to infect and cause disease in a well-established murine model of CDI. Together, these data support the hypothesis that the perR mutation in 630Δerm arose as a result of exposure to ambient oxygen and that the perR mutation in 630Δerm is unlikely to impact CDI-relevant phenotypes in laboratory studies.
Collapse
Affiliation(s)
- Anna L. Gregory
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
- Department of Medical Microbiology & Immunology, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
- Microbiology Doctoral Training Program, University of Wisconsin-Madison, Madison, WI, USA
| | - Hailey E. Bussan
- Microbiology Doctoral Training Program, University of Wisconsin-Madison, Madison, WI, USA
- Department of Bacteriology, University of Wisconsin-Madison, Madison, WI, USA
| | - Madeline A. Topf
- Department of Medical Microbiology & Immunology, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
- Microbiology Doctoral Training Program, University of Wisconsin-Madison, Madison, WI, USA
| | - Andrew J. Hryckowian
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
- Department of Medical Microbiology & Immunology, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| |
Collapse
|
21
|
McMillan AS, Zhang G, Dougherty MK, McGill SK, Gulati AS, Baker ES, Theriot CM. Metagenomic, metabolomic, and lipidomic shifts associated with fecal microbiota transplantation for recurrent Clostridioides difficile infection. mSphere 2024; 9:e0070624. [PMID: 39377587 PMCID: PMC11520286 DOI: 10.1128/msphere.00706-24] [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: 08/26/2024] [Accepted: 09/08/2024] [Indexed: 10/09/2024] Open
Abstract
Recurrent C. difficile infection (rCDI) is an urgent public health threat, for which the last resort and lifesaving treatment is a fecal microbiota transplant (FMT). However, the exact mechanisms that mediate a successful FMT are not well-understood. Here, we use longitudinal stool samples collected from patients undergoing FMT to evaluate intra-individual changes in the microbiome, metabolome, and lipidome after successful FMTs relative to their baselines pre-FMT. We show changes in the abundance of many lipids, specifically a decrease in acylcarnitines post-FMT, and a shift from conjugated bile acids pre-FMT to deconjugated secondary bile acids post-FMT. These changes correlate with a decrease in Enterobacteriaceae, which encode carnitine metabolism genes, and an increase in Lachnospiraceae, which encode bile acid altering genes such as bile salt hydrolases (BSHs) and the bile acid-inducible (bai) operon, post-FMT. We also show changes in gut microbe-encoded amino acid biosynthesis genes, of which Enterobacteriaceae was the primary contributor to amino acids C. difficile is auxotrophic for. Liquid chromatography, ion mobility spectrometry, and mass spectrometry (LC-IMS-MS) revealed a shift from microbial conjugation of primary bile acids pre-FMT to secondary bile acids post-FMT. Here, we define the structural and functional changes associated with a successful FMT and generate hypotheses that require further experimental validation. This information is meant to help guide the development of new microbiota-focused therapeutics to treat rCDI.IMPORTANCERecurrent C. difficile infection is an urgent public health threat, for which the last resort and lifesaving treatment is a fecal microbiota transplant. However, the exact mechanisms that mediate a successful FMT are not well-understood. Here, we show changes in the abundance of many lipids, specifically acylcarnitines and bile acids, in response to FMT. These changes correlate with Enterobacteriaceae pre-FMT, which encodes carnitine metabolism genes, and Lachnospiraceae post-FMT, which encodes bile salt hydrolases and baiA genes. There was also a shift from microbial conjugation of primary bile acids pre-FMT to secondary bile acids post-FMT. Here, we define the structural and functional changes associated with a successful FMT, which we hope will help aid in the development of new microbiota-focused therapeutics to treat rCDI.
Collapse
Affiliation(s)
- Arthur S. McMillan
- Genetics Program, Department of Biological Sciences, College of Science, North Carolina State University, Raleigh, North Carolina, USA
- Department of Population Health and Pathobiology, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina, USA
| | - Guozhi Zhang
- Department of Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Michael K. Dougherty
- Department of Medicine, Division of Gastroenterology and Hepatology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Rex Digestive Healthcare, Raleigh, North Carolina, USA
| | - Sarah K. McGill
- Department of Medicine, Division of Gastroenterology and Hepatology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Ajay S. Gulati
- Department of Pediatrics, Division of Gastroenterology and Hepatology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Erin S. Baker
- Department of Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Casey M. Theriot
- Department of Population Health and Pathobiology, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina, USA
| |
Collapse
|
22
|
Lee A, Yoo JS, Yoon EJ. Gut Microbiota and New Microbiome-Targeted Drugs for Clostridioides difficile Infections. Antibiotics (Basel) 2024; 13:995. [PMID: 39452261 PMCID: PMC11505460 DOI: 10.3390/antibiotics13100995] [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: 08/21/2024] [Revised: 10/16/2024] [Accepted: 10/18/2024] [Indexed: 10/26/2024] Open
Abstract
Clostridioides difficile is a major causative pathogen for antibiotic-associated diarrhea and C. difficile infections (CDIs) may lead to life-threatening diseases in clinical settings. Most of the risk factors for the incidence of CDIs, i.e., antibiotic use, treatment by proton pump inhibitors, old age, and hospitalization, are associated with dysbiosis of gut microbiota and associated metabolites and, consequently, treatment options for CDIs include normalizing the composition of the intestinal microbiome. In this review, with an introduction to the CDI and its global epidemiology, CDI-associated traits of the gut microbiome and its metabolites were reviewed, and microbiome-targeting treatment options were introduced, which was approved recently as a new drug by the United States Food and Drug Administration (U.S. FDA), rather than a medical practice.
Collapse
Affiliation(s)
| | | | - Eun-Jeong Yoon
- Division of Antimicrobial Resistance Research, Korea National Institute of Health, Korea Disease Control and Prevention Agency, Cheongju-si 28159, Republic of Korea
| |
Collapse
|
23
|
Ke F, Dong ZH, Bu F, Li CN, He QT, Liu ZC, Lu J, Yu K, Wang DG, Xu HN, Ye CT. Clostridium difficile infection following colon subtotal resection in a patient with gallstones: A case report and review of literature. World J Gastrointest Surg 2024; 16:3048-3056. [PMID: 39351567 PMCID: PMC11438826 DOI: 10.4240/wjgs.v16.i9.3048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 07/02/2024] [Accepted: 07/10/2024] [Indexed: 09/18/2024] Open
Abstract
BACKGROUND Clostridium difficile (C. difficile) infection (CDI) is a rare clinical disease caused by changes in the intestinal microenvironment, which has a variety of causes and a poor prognosis, and for which there is no standardized clinical treatment. CASE SUMMARY A patient experienced recurrent difficulty in bowel movements over the past decade. Recently, symptoms worsened within the last ten days, leading to a clinic visit due to constipation. The patient was subsequently referred to our department. Preoperatively, the patient was diagnosed with obstructed colon accompanied by gallstones. Empirical antibiotics were administered both before and after surgery to prevent infection. On the fourth day post-surgery, symptoms of CDI emerged. Stool cultures confirmed the presence of C. difficile DNA. Treatment involved a combination of vancomycin and linezolid, resulting in the patient's successful recovery upon discharge. However, the patient failed to adhere to the prescribed medication after discharge and was discovered deceased during a follow-up two months later. CONCLUSION CDI is the leading cause of nosocomial post-operative care, with limited clinical cases and poor patient prognosis, and comprehensive clinical treatment guidelines are still lacking. This infection can be triggered by a variety of factors, including intestinal hypoxia, inappropriate antibiotic use, and bile acid circulation disorders. In patients with chronic bowel disease and related etiologies, prompt preoperative attention to possible CDI and preoperative bowel preparation is critical. Adequate and prolonged medication should be maintained in the treatment of CDI to prevent recurrence of the disease.
Collapse
Affiliation(s)
- Feng Ke
- Department of General Surgery, Affiliated Hospital of Changchun University of Chinese Medicine, Changchun 130000, Jilin Province, China
| | - Zhen-Hua Dong
- Department of Gastrointestinal Surgery, The First Hospital of Jilin University, Changchun 130061, Jilin Province, China
| | - Fan Bu
- Department of Plastic and Aesthetic Surgery, The First Hospital of Jilin University, Changchun 130061, Jilin Province, China
| | - Cheng-Nan Li
- Department of Encephalopathy Rehabilitation, Chaoyi Hospital, Yanbian Korean Autonomous Prefecture, Yanji 133000, Jilin Province, China
| | - Qi-Tong He
- Department of Gastrointestinal Surgery, The First Hospital of Jilin University, Changchun 130061, Jilin Province, China
| | - Zhi-Cheng Liu
- Department of Gastrointestinal Surgery, The First Hospital of Jilin University, Changchun 130061, Jilin Province, China
| | - Ji Lu
- Department of Urology, The First Hospital of Jilin University, Changchun 130061, Jilin Province, China
| | - Kai Yu
- Department of Urology, The First Hospital of Jilin University, Changchun 130061, Jilin Province, China
| | - Da-Guang Wang
- Department of Gastrointestinal Surgery, The First Hospital of Jilin University, Changchun 130061, Jilin Province, China
| | - He-Nan Xu
- Department of Gastrointestinal Surgery, The First Hospital of Jilin University, Changchun 130061, Jilin Province, China
| | - Chang-Tao Ye
- Department of Urology, The First Hospital of Jilin University, Changchun 130061, Jilin Province, China
| |
Collapse
|
24
|
Dekker Nitert M, Ovadia C. Microbial metabolites as a way to provide crosstalk between gut and liver. Obstet Med 2024; 17:168-174. [PMID: 39262911 PMCID: PMC11384814 DOI: 10.1177/1753495x241258383] [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: 01/28/2024] [Accepted: 05/04/2024] [Indexed: 09/13/2024] Open
Abstract
Bile acid metabolism is partially regulated through the activity of the gut microbiota. Primary bile acids can be deconjugated and modified by bacteria expressing bile salt hydrolase and other enzymes, changing bile acid recycling by changing the interactions between enterocytes and hepatocytes. The modified bile acids can also activate signalling in cells regulating metabolism including colonic L-cells, skeletal muscle cells and brown adipocytes. In pregnancy, both bile acid metabolism and gut microbiota composition are altered. In women with intrahepatic cholestasis of pregnancy, the changes in bile acid metabolism are exacerbated and there is some evidence that the gut microbiota composition is also altered. Here we review the crosstalk between the liver and the gut especially in women with intrahepatic cholestasis of pregnancy, with a focus on the role of the gut microbiota in this crosstalk.
Collapse
Affiliation(s)
- Marloes Dekker Nitert
- School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, QLD, Australia
| | - Caroline Ovadia
- Department of Women and Children's Health, King's College London, London, UK
| |
Collapse
|
25
|
Zaatry R, Herren R, Gefen T, Geva-Zatorsky N. Microbiome and infectious disease: diagnostics to therapeutics. Microbes Infect 2024; 26:105345. [PMID: 38670215 DOI: 10.1016/j.micinf.2024.105345] [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: 07/13/2023] [Revised: 04/22/2024] [Accepted: 04/22/2024] [Indexed: 04/28/2024]
Abstract
Over 300 years of research on the microbial world has revealed their importance in human health and disease. This review explores the impact and potential of microbial-based detection methods and therapeutic interventions, integrating research of early microbiologists, current findings, and future perspectives.
Collapse
Affiliation(s)
- Rawan Zaatry
- Rappaport Faculty of Medicine, Rappaport Technion Integrated Cancer Center, Technion, Haifa, Israel
| | - Rachel Herren
- Rappaport Faculty of Medicine, Rappaport Technion Integrated Cancer Center, Technion, Haifa, Israel
| | - Tal Gefen
- Rappaport Faculty of Medicine, Rappaport Technion Integrated Cancer Center, Technion, Haifa, Israel
| | - Naama Geva-Zatorsky
- Rappaport Faculty of Medicine, Rappaport Technion Integrated Cancer Center, Technion, Haifa, Israel; CIFAR, Humans & the Microbiome, Toronto, Canada.
| |
Collapse
|
26
|
Sulaiman JE, Thompson J, Qian Y, Vivas EI, Diener C, Gibbons SM, Safdar N, Venturelli OS. Elucidating human gut microbiota interactions that robustly inhibit diverse Clostridioides difficile strains across different nutrient landscapes. Nat Commun 2024; 15:7416. [PMID: 39198411 PMCID: PMC11358386 DOI: 10.1038/s41467-024-51062-w] [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: 04/12/2024] [Accepted: 07/25/2024] [Indexed: 09/01/2024] Open
Abstract
The human gut pathogen Clostridioides difficile displays substantial inter-strain genetic variability and confronts a changeable nutrient landscape in the gut. We examined how human gut microbiota inter-species interactions influence the growth and toxin production of various C. difficile strains across different nutrient environments. Negative interactions influencing C. difficile growth are prevalent in an environment containing a single highly accessible resource and sparse in an environment containing C. difficile-preferred carbohydrates. C. difficile toxin production displays significant community-context dependent variation and does not trend with growth-mediated inter-species interactions. C. difficile strains exhibit differences in interactions with Clostridium scindens and the ability to compete for proline. Further, C. difficile shows substantial differences in transcriptional profiles in co-culture with C. scindens or Clostridium hiranonis. C. difficile exhibits massive alterations in metabolism and other cellular processes in co-culture with C. hiranonis, reflecting their similar metabolic niches. C. hiranonis uniquely inhibits the growth and toxin production of diverse C. difficile strains across different nutrient environments and robustly ameliorates disease severity in mice. In sum, understanding the impact of C. difficile strain variability and nutrient environments on inter-species interactions could help improve the effectiveness of anti-C. difficile strategies.
Collapse
Affiliation(s)
- Jordy Evan Sulaiman
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI, USA
| | - Jaron Thompson
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI, USA
- Department of Chemical & Biological Engineering, University of Wisconsin-Madison, Madison, WI, USA
| | - Yili Qian
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI, USA
| | - Eugenio I Vivas
- Gnotobiotic Animal Core Facility, University of Wisconsin-Madison, Madison, WI, USA
- Department of Bacteriology, University of Wisconsin-Madison, Madison, WI, USA
| | - Christian Diener
- Institute for Systems Biology, Seattle, WA, USA
- Diagnostic and Research Institute of Hygiene, Microbiology and Environmental Medicine, Medical University of Graz, Graz, Austria
| | - Sean M Gibbons
- Institute for Systems Biology, Seattle, WA, USA
- Department of Bioengineering, University of Washington, Seattle, WA, USA
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
- eScience Institute, University of Washington, Seattle, WA, USA
| | - Nasia Safdar
- Division of Infectious Disease, Department of Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
- Department of Medicine, William S. Middleton Veterans Hospital Madison, Madison, WI, USA
| | - Ophelia S Venturelli
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI, USA.
- Department of Chemical & Biological Engineering, University of Wisconsin-Madison, Madison, WI, USA.
- Department of Bacteriology, University of Wisconsin-Madison, Madison, WI, USA.
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, USA.
| |
Collapse
|
27
|
Reitmeyer KM, Rana B, Awad D, Huang E, Park JJ, Yassin A, Mills JP, Abdul Azim A, Bhatt PJ, Narayanan N. Effectiveness of oral vancomycin as prophylaxis against Clostridioides difficile infection in hematopoietic stem cell transplant patients. ANTIMICROBIAL STEWARDSHIP & HEALTHCARE EPIDEMIOLOGY : ASHE 2024; 4:e107. [PMID: 39823126 PMCID: PMC11736458 DOI: 10.1017/ash.2024.90] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 04/20/2024] [Accepted: 04/23/2024] [Indexed: 01/19/2025]
Abstract
Objective Patients receiving hematopoietic stem cell transplants (HSCT) are at increased risk for Clostridioides difficile infection (CDI). The purpose of this study was to assess the effectiveness of oral vancomycin prophylaxis (OVP) for CDI in HSCT patients. Design Single-center, retrospective cohort. Setting Tertiary care academic medical center in New Jersey. Patients Patients ≥18 years old during admission for the HSCT were included. Patients who were admitted <72 hours or who had an active CDI prior to HSCT day 0 were excluded. Methods Medical records of patients admitted between January 2015 and August 2022 to undergo an allogeneic or autologous HSCT were reviewed. The primary end point was the incidence of in-hospital CDI. Secondary end points included the incidence of vancomycin-resistant enterococci (VRE) bloodstream infections, VRE isolated from any clinical culture, gram-negative bloodstream infections, hospital survival, and hospital length of stay. Exploratory end points, including 1-year survival, relapse, and incidence of graft-versus-host disease, were also collected. Results A total of 156 HSCT patients were included. There was 1 case of CDI (1 of 81, 1.23%) in the OVP group compared to 8 CDI cases (8 of 75, 10.67%) in the no OVP group (P = .0147). There were no significant (P > .05) between-group differences in incidence of gram-negative bloodstream infections, hospital survival, and length of stay. There were zero clinical cultures positive for VRE. Conclusions In-hospital incidence of CDI in HSCT patients was significantly decreased with OVP. Randomized controlled trials are needed in this high-risk population to assess the efficacy and risks of OVP for CDI.
Collapse
Affiliation(s)
- Kelly M. Reitmeyer
- Department of Pharmacy, Robert Wood Johnson University Hospital, New Brunswick, NJ, USA
| | - Brijesh Rana
- Rutgers University School of Public Health, Piscataway, NJ, USA
- Department of Pharmacy Practice and Administration, Rutgers University Ernest Mario School of Pharmacy, Piscataway, NJ, USA
| | - David Awad
- Department of Pharmacy, Robert Wood Johnson University Hospital, New Brunswick, NJ, USA
| | - Esther Huang
- Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, USA
| | - Jiyeon J. Park
- Department of Pharmacy, Englewood Health, Englewood, NJ, USA
| | - Arsheena Yassin
- Department of Pharmacy, Robert Wood Johnson University Hospital, New Brunswick, NJ, USA
| | - John P. Mills
- Division of Allergy, Immunology, and Infectious Diseases, Department of Medicine, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - Ahmed Abdul Azim
- Division of Allergy, Immunology, and Infectious Diseases, Department of Medicine, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - Pinki J. Bhatt
- Department of Pharmacy Practice and Administration, Rutgers University Ernest Mario School of Pharmacy, Piscataway, NJ, USA
- Division of Allergy, Immunology, and Infectious Diseases, Department of Medicine, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - Navaneeth Narayanan
- Department of Pharmacy, Robert Wood Johnson University Hospital, New Brunswick, NJ, USA
- Department of Pharmacy Practice and Administration, Rutgers University Ernest Mario School of Pharmacy, Piscataway, NJ, USA
- Division of Allergy, Immunology, and Infectious Diseases, Department of Medicine, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| |
Collapse
|
28
|
Asghari Ozma M, Mahmoodzadeh Hosseini H, Ataee MH, Mirhosseini SA. Evaluating the antibacterial, antibiofilm, and anti-toxigenic effects of postbiotics from lactic acid bacteria on Clostridium difficile. IRANIAN JOURNAL OF MICROBIOLOGY 2024; 16:497-508. [PMID: 39267941 PMCID: PMC11389761 DOI: 10.18502/ijm.v16i4.16309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 09/15/2024]
Abstract
Background and Objectives The most common cause of healthcare-associated diarrhea is Clostridium difficile infection (CDI), which causes severe and recurring symptoms. The increase of antibiotic-resistant C. difficile requires alternate treatments. Postbiotics, metabolites produced by probiotics, fight CDI owing to their antibacterial capabilities. This study aims to evaluate the antibacterial, antibiofilm, and anti-toxigenic potential of postbiotics in combating CDI. Materials and Methods GC-MS evaluated postbiotics from Bifidobacterium bifidum and Lactobacillus plantarum. Disk diffusion and broth microdilution determined C. difficile antibacterial inhibition zones and MICs. Microtiter plates assessed antibiofilm activity. MTT assay evaluated postbiotics anti-viability on HEK293. ELISA testing postbiotic detoxification of toxins A and B. Postbiotics were examined for tcdA and tcdB genes expression using real-time PCR. Results The most identified B. bifidum and L. plantarum postbiotic compounds were glycolic acid (7.2%) and butyric acid (13.57%). B. bifidum and L. plantarum displayed 13 and 10 mm inhibition zones and 2.5 and 5 mg/ml MICs against C. difficile. B. bifidum reduced biofilm at 1.25 mg/ml by 49% and L. plantarum by 31%. MTT assay showed both postbiotics had little influence on cell viability, which was over 80%. The detoxification power of postbiotics revealed that B. bifidum decreased toxin A and B production more effectively than L. plantarum, and also their related tcdA and tcdB genes expression reduction were statistically significant (p < 0.05). Conclusion Postbiotics' ability to inhibit bacterial growth, biofilm disruption, and toxin reduction makes them a promising adjunctive for CDI treatment and a good solution to pathogens' antibiotic resistance.
Collapse
Affiliation(s)
- Mahdi Asghari Ozma
- Student Research Committee, Baqiytallah University of Medical Sciences, Tehran, Iran
| | - Hamideh Mahmoodzadeh Hosseini
- Applied Microbiology Research Center, Biomedicine Technologies Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Mohammad Hossein Ataee
- Applied Microbiology Research Center, Biomedicine Technologies Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Seyed Ali Mirhosseini
- Applied Microbiology Research Center, Biomedicine Technologies Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| |
Collapse
|
29
|
Zaidi SMH, Haider R, Kazmi SAB, Husnain A, Khan S, Merchant S, Tayyab H, Wazeen FR, Chaudhary AJ. Beyond Antibiotics: Novel Approaches in the Treatment of Recurrent Clostridioides difficile Infection. ACG Case Rep J 2024; 11:e01333. [PMID: 39081300 PMCID: PMC11286250 DOI: 10.14309/crj.0000000000001333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 03/06/2024] [Indexed: 08/02/2024] Open
Affiliation(s)
| | - Ramsha Haider
- Karachi Medical and Dental College, Karachi, Pakistan
| | | | - Ali Husnain
- Department of Medicine, King Edward Medical University, Lahore, Pakistan
| | - Saniah Khan
- Department of Medicine, King Edward Medical University, Lahore, Pakistan
| | | | - Hamnah Tayyab
- Department of Medicine, King Edward Medical University, Lahore, Pakistan
| | - Fazl Rahim Wazeen
- Department of Medicine, Greater Baltimore Medical Center, Towson, MD
| | | |
Collapse
|
30
|
Spigaglia P. Clostridioides difficile and Gut Microbiota: From Colonization to Infection and Treatment. Pathogens 2024; 13:646. [PMID: 39204246 PMCID: PMC11357127 DOI: 10.3390/pathogens13080646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 07/23/2024] [Accepted: 07/29/2024] [Indexed: 09/03/2024] Open
Abstract
Clostridioides difficile is the main causative agent of antibiotic-associated diarrhea (AAD) in hospitals in the developed world. Both infected patients and asymptomatic colonized individuals represent important transmission sources of C. difficile. C. difficile infection (CDI) shows a large range of symptoms, from mild diarrhea to severe manifestations such as pseudomembranous colitis. Epidemiological changes in CDIs have been observed in the last two decades, with the emergence of highly virulent types and more numerous and severe CDI cases in the community. C. difficile interacts with the gut microbiota throughout its entire life cycle, and the C. difficile's role as colonizer or invader largely depends on alterations in the gut microbiota, which C. difficile itself can promote and maintain. The restoration of the gut microbiota to a healthy state is considered potentially effective for the prevention and treatment of CDI. Besides a fecal microbiota transplantation (FMT), many other approaches to re-establishing intestinal eubiosis are currently under investigation. This review aims to explore current data on C. difficile and gut microbiota changes in colonized individuals and infected patients with a consideration of the recent emergence of highly virulent C. difficile types, with an overview of the microbial interventions used to restore the human gut microbiota.
Collapse
Affiliation(s)
- Patrizia Spigaglia
- Department of Infectious Diseases, Istituto Superiore di Sanità, 00161 Roma, Italy
| |
Collapse
|
31
|
Martignoni MM, Raulo A, Linkovski O, Kolodny O. SIR+ models: accounting for interaction-dependent disease susceptibility in the planning of public health interventions. Sci Rep 2024; 14:12908. [PMID: 38839831 PMCID: PMC11153654 DOI: 10.1038/s41598-024-63008-9] [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: 10/10/2023] [Accepted: 05/23/2024] [Indexed: 06/07/2024] Open
Abstract
Avoiding physical contact is regarded as one of the safest and most advisable strategies to follow to reduce pathogen spread. The flip side of this approach is that a lack of social interactions may negatively affect other dimensions of health, like induction of immunosuppressive anxiety and depression or preventing interactions of importance with a diversity of microbes, which may be necessary to train our immune system or to maintain its normal levels of activity. These may in turn negatively affect a population's susceptibility to infection and the incidence of severe disease. We suggest that future pandemic modelling may benefit from relying on 'SIR+ models': epidemiological models extended to account for the benefits of social interactions that affect immune resilience. We develop an SIR+ model and discuss which specific interventions may be more effective in balancing the trade-off between minimizing pathogen spread and maximizing other interaction-dependent health benefits. Our SIR+ model reflects the idea that health is not just the mere absence of disease, but rather a state of physical, mental and social well-being that can also be dependent on the same social connections that allow pathogen spread, and the modelling of public health interventions for future pandemics should account for this multidimensionality.
Collapse
Affiliation(s)
- Maria M Martignoni
- Department of Ecology, Evolution and Behavior, Faculty of Sciences, A. Silberman Institute of Life Sciences, Hebrew University of Jerusalem, Jerusalem, Israel.
| | - Aura Raulo
- Department of Biology, University of Oxford, Oxford, UK
- Department of Computing, University of Turku, Turku, Finland
| | - Omer Linkovski
- Department of Psychology and The Gonda Multidisciplinary Brain Research Center, Bar-Ilan University, Ramat-Gan, Israel
| | - Oren Kolodny
- Department of Ecology, Evolution and Behavior, Faculty of Sciences, A. Silberman Institute of Life Sciences, Hebrew University of Jerusalem, Jerusalem, Israel
| |
Collapse
|
32
|
Pumiglia L, Wilson L, Rashidi L. Clostridioides difficile Colitis. Surg Clin North Am 2024; 104:545-556. [PMID: 38677819 DOI: 10.1016/j.suc.2023.11.005] [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: 04/29/2024]
Abstract
Clostridioides difficile colitis is an important source of hospital-acquired diarrhea associated with antibiotic use. Symptoms are profuse watery diarrhea, typically following a course of antibiotics; however, some cases of fulminant disease may manifest with shock, ileus, or megacolon. Nonfulminant colitis is treated with oral fidaxomicin. C difficile colitis has a high potential for recurrence, and recurrent episodes are also treated with fidaxomicin. Bezlotoxumab is another medication that may be used in populations at high risk for further recurrence. Fulminant disease is treated with maximal medical therapy and early surgical consultation. Antibiotic stewardship is critical to preventing C difficile colitis.
Collapse
Affiliation(s)
- Luke Pumiglia
- Department of General Surgery, Madigan Army Medical Center, 9040 Jackson Avenue, Joint Base Lewis-McChord, WA 98431, USA
| | - Lexi Wilson
- Department of Colorectal Surgery, Swedish Medical Center, 747 Broadway, Seattle, WA 98122, USA
| | - Laila Rashidi
- Department of Surgery, MultiCare Health Care System, Washington State University, 3124 19th Street Suite 220, Tacoma, WA 98405, USA.
| |
Collapse
|
33
|
Chen H, Zhang Y, Pan Y, Wu L, Wang W, Zhang H, Lou H. Antibiotic-induced microbiome depletion promotes intestinal colonization by Campylobacter jejuni in mice. BMC Microbiol 2024; 24:156. [PMID: 38724913 PMCID: PMC11080253 DOI: 10.1186/s12866-024-03313-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 04/24/2024] [Indexed: 05/13/2024] Open
Abstract
BACKGROUND To establish a method to induce Campylobacter jejuni colonization in the intestines of C57BL/6 mice through antibiotic-induced microbiome depletion. RESULTS Fifty-four female C57BL/6 mice were divided into the normal, control, and experimental groups. The experimental group was administered intragastric cefoperazone sodium and sulbactam sodium (50 mg/mL) for 2 days; then, the experimental and control mice were intragastrically administered 200 µL C. jejuni, which was repeated once more after 2 days. Animal feces were collected, and the HipO gene of C. jejuni was detected using TaqMan qPCR from day 1 to day 14 after modeling completion. Immunofluorescence was used to detect intestinal C. jejuni colonization on day 14, and pathological changes were observed using hematoxylin and eosin staining. Additionally, 16S rDNA analyses of the intestinal contents were conducted on day 14. In the experimental group, C. jejuni was detected in the feces from days 1 to 14 on TaqMan qPCR, and immunofluorescence-labeled C. jejuni were visibly discernable in the intestinal lumen. The intestinal mucosa was generally intact and showed no significant inflammatory-cell infiltration. Diversity analysis of the colonic microbiota showed significant inter-group differences. In the experimental group, the composition of the colonic microbiota differed from that in the other 2 groups at the phylum level, and was characterized by a higher proportion of Bacteroidetes and a lower proportion of Firmicutes. CONCLUSIONS Microbiome depletion induced by cefoperazone sodium and sulbactam sodium could promote long-term colonization of C. jejuni in the intestines of mice.
Collapse
Affiliation(s)
- Haohao Chen
- Medical Molecular Biology Laboratory, School of Medicine, Jinhua Polytechnic, No. 1188 Wuzhou Street, Wucheng District, Jinhua, Zhejiang Province, P.R. China.
| | - Yanfang Zhang
- Medical Molecular Biology Laboratory, School of Medicine, Jinhua Polytechnic, No. 1188 Wuzhou Street, Wucheng District, Jinhua, Zhejiang Province, P.R. China
| | - Yi Pan
- Medical Molecular Biology Laboratory, School of Medicine, Jinhua Polytechnic, No. 1188 Wuzhou Street, Wucheng District, Jinhua, Zhejiang Province, P.R. China
| | - Lin Wu
- Medical Molecular Biology Laboratory, School of Medicine, Jinhua Polytechnic, No. 1188 Wuzhou Street, Wucheng District, Jinhua, Zhejiang Province, P.R. China
| | - Wenqian Wang
- Medical Molecular Biology Laboratory, School of Medicine, Jinhua Polytechnic, No. 1188 Wuzhou Street, Wucheng District, Jinhua, Zhejiang Province, P.R. China
| | - Hui Zhang
- Animal Center, Jinhua Food and Drug Inspection and Testing Research Institute, Jinhua, Zhejiang Province, P.R. China
| | - Hongqiang Lou
- Medical Molecular Biology Laboratory, School of Medicine, Jinhua Polytechnic, No. 1188 Wuzhou Street, Wucheng District, Jinhua, Zhejiang Province, P.R. China.
| |
Collapse
|
34
|
Reveles KR, Hickmott AJ, Strey KA, Mustoe AC, Arroyo JP, Power ML, Ridenhour BJ, Amato KR, Ross CN. Developing the Common Marmoset as a Translational Geroscience Model to Study the Microbiome and Healthy Aging. Microorganisms 2024; 12:852. [PMID: 38792682 PMCID: PMC11123169 DOI: 10.3390/microorganisms12050852] [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: 03/04/2024] [Revised: 04/02/2024] [Accepted: 04/22/2024] [Indexed: 05/26/2024] Open
Abstract
Emerging data support associations between the depletion of the healthy gut microbiome and aging-related physiological decline and disease. In humans, fecal microbiota transplantation (FMT) has been used successfully to restore gut microbiome structure and function and to treat C. difficile infections, but its application to healthy aging has been scarcely investigated. The marmoset is an excellent model for evaluating microbiome-mediated changes with age and interventional treatments due to their relatively shorter lifespan and many social, behavioral, and physiological functions that mimic human aging. Prior work indicates that FMT is safe in marmosets and may successfully mediate gut microbiome function and host health. This narrative review (1) provides an overview of the rationale for FMT to support healthy aging using the marmoset as a translational geroscience model, (2) summarizes the prior use of FMT in marmosets, (3) outlines a protocol synthesized from prior literature for studying FMT in aging marmosets, and (4) describes limitations, knowledge gaps, and future research needs in this field.
Collapse
Affiliation(s)
- Kelly R. Reveles
- College of Pharmacy, The University of Texas at Austin, Austin, TX 78712, USA;
- Graduate School of Biomedical Sciences, University of Texas Health San Antonio, San Antonio, TX 78229, USA; (A.J.H.); (A.C.M.); (J.P.A.); (C.N.R.)
| | - Alexana J. Hickmott
- Graduate School of Biomedical Sciences, University of Texas Health San Antonio, San Antonio, TX 78229, USA; (A.J.H.); (A.C.M.); (J.P.A.); (C.N.R.)
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX 78227, USA
| | - Kelsey A. Strey
- College of Pharmacy, The University of Texas at Austin, Austin, TX 78712, USA;
- Graduate School of Biomedical Sciences, University of Texas Health San Antonio, San Antonio, TX 78229, USA; (A.J.H.); (A.C.M.); (J.P.A.); (C.N.R.)
| | - Aaryn C. Mustoe
- Graduate School of Biomedical Sciences, University of Texas Health San Antonio, San Antonio, TX 78229, USA; (A.J.H.); (A.C.M.); (J.P.A.); (C.N.R.)
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX 78227, USA
| | - Juan Pablo Arroyo
- Graduate School of Biomedical Sciences, University of Texas Health San Antonio, San Antonio, TX 78229, USA; (A.J.H.); (A.C.M.); (J.P.A.); (C.N.R.)
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX 78227, USA
| | - Michael L. Power
- Center for Species Survival, Smithsonian’s National Zoo and Conservation Biology Institute, Washington, DC 20008, USA;
| | - Benjamin J. Ridenhour
- Department of Mathematics and Statistical Science, University of Idaho, Moscow, ID 83844, USA;
| | - Katherine R. Amato
- Department of Anthropology, Northwestern University, Evanston, IL 60208, USA;
| | - Corinna N. Ross
- Graduate School of Biomedical Sciences, University of Texas Health San Antonio, San Antonio, TX 78229, USA; (A.J.H.); (A.C.M.); (J.P.A.); (C.N.R.)
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX 78227, USA
| |
Collapse
|
35
|
Sulaiman JE, Thompson J, Qian Y, Vivas EI, Diener C, Gibbons SM, Safdar N, Venturelli OS. Elucidating human gut microbiota interactions that robustly inhibit diverse Clostridioides difficile strains across different nutrient landscapes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.13.589383. [PMID: 38659900 PMCID: PMC11042340 DOI: 10.1101/2024.04.13.589383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/26/2024]
Abstract
The human gut pathogen Clostridioides difficile displays extreme genetic variability and confronts a changeable nutrient landscape in the gut. We mapped gut microbiota inter-species interactions impacting the growth and toxin production of diverse C. difficile strains in different nutrient environments. Although negative interactions impacting C. difficile are prevalent in environments promoting resource competition, they are sparse in an environment containing C. difficile-preferred carbohydrates. C. difficile strains display differences in interactions with Clostridium scindens and the ability to compete for proline. C. difficile toxin production displays substantial community-context dependent variation and does not trend with growth-mediated inter-species interactions. C. difficile shows substantial differences in transcriptional profiles in the presence of the closely related species C. hiranonis or C. scindens. In co-culture with C. hiranonis, C. difficile exhibits massive alterations in metabolism and other cellular processes, consistent with their high metabolic overlap. Further, Clostridium hiranonis inhibits the growth and toxin production of diverse C. difficile strains across different nutrient environments and ameliorates the disease severity of a C. difficile challenge in a murine model. In sum, strain-level variability and nutrient environments are major variables shaping gut microbiota interactions with C. difficile.
Collapse
Affiliation(s)
- Jordy Evan Sulaiman
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI, USA
| | - Jaron Thompson
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI, USA
- Department of Chemical & Biological Engineering, University of Wisconsin-Madison, Madison, WI, USA
| | - Yili Qian
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI, USA
| | - Eugenio I. Vivas
- Gnotobiotic Animal Core Facility, University of Wisconsin-Madison, Madison, WI, USA
- Department of Bacteriology, University of Wisconsin-Madison, Madison, WI, USA
| | | | - Sean M. Gibbons
- Institute for Systems Biology, Seattle, WA, USA
- Department of Bioengineering, University of Washington, Seattle, WA, USA
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
- eScience Institute, University of Washington, Seattle, WA, USA
| | - Nasia Safdar
- Division of Infectious Disease, Department of Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
- Department of Medicine, William S. Middleton Veterans Hospital Madison, Madison, WI, USA
| | - Ophelia S. Venturelli
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI, USA
- Department of Chemical & Biological Engineering, University of Wisconsin-Madison, Madison, WI, USA
- Department of Bacteriology, University of Wisconsin-Madison, Madison, WI, USA
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, USA
| |
Collapse
|
36
|
Schmidt N, Van Den Ham K, Bower L, Li S, Lorenzi H, Doumbo S, Doumtabe D, Kayentao K, Ongoiba A, Traore B, Crompton P. Susceptibility to febrile malaria is associated with an inflammatory gut microbiome. RESEARCH SQUARE 2024:rs.3.rs-3974068. [PMID: 38645126 PMCID: PMC11030534 DOI: 10.21203/rs.3.rs-3974068/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/23/2024]
Abstract
Malaria is a major public health problem, but many of the factors underlying the pathogenesis of this disease are not well understood. Here, we demonstrate in Malian children that susceptibility to febrile malaria following infection with Plasmodium falciparum is associated with the composition of the gut microbiome prior to the malaria season. Gnotobiotic mice colonized with the fecal samples of malaria-susceptible children had a significantly higher parasite burden following Plasmodium infection compared to gnotobiotic mice colonized with the fecal samples of malaria-resistant children. The fecal microbiome of the susceptible children was enriched for bacteria associated with inflammation, mucin degradation, gut permeability and inflammatory bowel disorders (e.g., Ruminococcus gauvreauii, Ruminococcus torques, Dorea formicigenerans, Dorea longicatena, Lachnoclostridium phocaeense and Lachnoclostridium sp. YL32). However, the susceptible children also had a greater abundance of bacteria known to produce anti-inflammatory short-chain fatty acids and those associated with favorable prognosis and remission following dysbiotic intestinal events (e.g., Anaerobutyricum hallii, Blautia producta and Sellimonas intestinalis). Metabolomics analysis of the human fecal samples corroborated the existence of inflammatory and recovery-associated features within the gut microbiome of the susceptible children. There was an enrichment of nitric oxide-derived DNA adducts (deoxyinosine and deoxyuridine) and long-chain fatty acids, the absorption of which has been shown to be inhibited by inflamed intestinal epithelial cells, and a decrease in the abundance of mucus phospholipids. Nevertheless, there were also increased levels of pseudouridine and hypoxanthine, which have been shown to be regulated in response to cellular stress and to promote recovery following injury or hypoxia. Overall, these results indicate that the gut microbiome may contribute malaria pathogenesis and suggest that therapies targeting intestinal inflammation could decrease malaria susceptibility.
Collapse
|
37
|
Yang H, Wu X, Li X, Zang W, Zhou Z, Zhou Y, Cui W, Kou Y, Wang L, Hu A, Wu L, Yin Z, Chen Q, Chen Y, Huang Z, Wang Y, Gu B. A commensal protozoan attenuates Clostridioides difficile pathogenesis in mice via arginine-ornithine metabolism and host intestinal immune response. Nat Commun 2024; 15:2842. [PMID: 38565558 PMCID: PMC10987486 DOI: 10.1038/s41467-024-47075-0] [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: 04/10/2023] [Accepted: 03/20/2024] [Indexed: 04/04/2024] Open
Abstract
Antibiotic-induced dysbiosis is a major risk factor for Clostridioides difficile infection (CDI), and fecal microbiota transplantation (FMT) is recommended for treating CDI. However, the underlying mechanisms remain unclear. Here, we show that Tritrichomonas musculis (T.mu), an integral member of the mouse gut commensal microbiota, reduces CDI-induced intestinal damage by inhibiting neutrophil recruitment and IL-1β secretion, while promoting Th1 cell differentiation and IFN-γ secretion, which in turn enhances goblet cell production and mucin secretion to protect the intestinal mucosa. T.mu can actively metabolize arginine, not only influencing the host's arginine-ornithine metabolic pathway, but also shaping the metabolic environment for the microbial community in the host's intestinal lumen. This leads to a relatively low ornithine state in the intestinal lumen in C. difficile-infected mice. These changes modulate C. difficile's virulence and the host intestinal immune response, and thus collectively alleviating CDI. These findings strongly suggest interactions between an intestinal commensal eukaryote, a pathogenic bacterium, and the host immune system via inter-related arginine-ornithine metabolism in the regulation of pathogenesis and provide further insights for treating CDI.
Collapse
Affiliation(s)
- Huan Yang
- Xuzhou Key Laboratory of Laboratory Diagnostics, School of Medical Technology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Xiaoxiao Wu
- Xuzhou Key Laboratory of Laboratory Diagnostics, School of Medical Technology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Xiao Li
- Xuzhou Key Laboratory of Laboratory Diagnostics, School of Medical Technology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Wanqing Zang
- Xuzhou Key Laboratory of Laboratory Diagnostics, School of Medical Technology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Zhou Zhou
- Xuzhou Key Laboratory of Laboratory Diagnostics, School of Medical Technology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Yuan Zhou
- Xuzhou Key Laboratory of Laboratory Diagnostics, School of Medical Technology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Wenwen Cui
- Xuzhou Center for Disease Control and Prevention, Xuzhou, Jiangsu, China
| | - Yanbo Kou
- Laboratory of Infection and Immunity, Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Liang Wang
- Department of Clinical Laboratory Medicine, Guangdong Provincial People's Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Ankang Hu
- Center of Animal Laboratory, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Lianlian Wu
- Center of Animal Laboratory, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Zhinan Yin
- The Biomedical Translational Research Institute, Health Science Center (School of Medicine), Jinan University, Guangzhou, Guangdong, China
| | - Quangang Chen
- Center of Animal Laboratory, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Ying Chen
- Xuzhou Key Laboratory of Laboratory Diagnostics, School of Medical Technology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Zhutao Huang
- Center of Animal Laboratory, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Yugang Wang
- Laboratory of Infection and Immunity, Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu, China.
| | - Bing Gu
- Xuzhou Key Laboratory of Laboratory Diagnostics, School of Medical Technology, Xuzhou Medical University, Xuzhou, Jiangsu, China.
- Department of Clinical Laboratory Medicine, Guangdong Provincial People's Hospital, Southern Medical University, Guangzhou, Guangdong, China.
| |
Collapse
|
38
|
McMillan AS, Zhang G, Dougherty MK, McGill SK, Gulati AS, Baker ES, Theriot CM. Metagenomic, metabolomic, and lipidomic shifts associated with fecal microbiota transplantation for recurrent Clostridioides difficile infection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.07.579219. [PMID: 38370838 PMCID: PMC10871284 DOI: 10.1101/2024.02.07.579219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/20/2024]
Abstract
Recurrent C. difficile infection (rCDI) is an urgent public health threat for which the last resort and lifesaving treatment is a fecal microbiota transplant (FMT). However, the exact mechanisms which mediate a successful FMT are not well understood. Here we use longitudinal stool samples collected from patients undergoing FMT to evaluate changes in the microbiome, metabolome, and lipidome after successful FMTs. We show changes in the abundance of many lipids, specifically acylcarnitines and bile acids, in response to FMT. These changes correlate with Enterobacteriaceae, which encode carnitine metabolism genes, and Lachnospiraceae, which encode bile salt hydrolases and baiA genes. LC-IMS-MS revealed a shift from microbial conjugation of primary bile acids pre-FMT to secondary bile acids post-FMT. Here we define the structural and functional changes in successful FMTs. This information will help guide targeted Live Biotherapeutic Product development for the treatment of rCDI and other intestinal diseases.
Collapse
|
39
|
Wang M, Chen X, Fang Y, Zheng X, Huang T, Nie Y, Wu XL. The trade-off between individual metabolic specialization and versatility determines the metabolic efficiency of microbial communities. Cell Syst 2024; 15:63-74.e5. [PMID: 38237552 DOI: 10.1016/j.cels.2023.12.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 09/17/2023] [Accepted: 12/12/2023] [Indexed: 01/23/2024]
Abstract
In microbial systems, a metabolic pathway can be either completed by one autonomous population or distributed among a consortium performing metabolic division of labor (MDOL). MDOL facilitates the system's function by reducing the metabolic burden; however, it may hinder the function by reducing the exchange efficiency of metabolic intermediates among individuals. As a result, the function of a community is influenced by the trade-offs between the metabolic specialization and versatility of individuals. To experimentally test this hypothesis, we deconstructed the naphthalene degradation pathway into four steps and introduced them individually or combinatorically into different strains with varying levels of metabolic specialization. Using these strains, we engineered 1,456 synthetic consortia and found that 74 consortia exhibited higher degradation function than both the autonomous population and rigorous MDOL consortium. Quantitative modeling provides general strategies for identifying the most effective MDOL configuration. Our study provides critical insights into the engineering of high-performance microbial systems.
Collapse
Affiliation(s)
- Miaoxiao Wang
- College of Engineering, Peking University, Beijing 100871, China; Department of Environmental Systems Science, ETH Zürich, Zürich 8092, Switzerland; Department of Environmental Microbiology, Eawag, Dübendorf 8600, Switzerland
| | - Xiaoli Chen
- College of Engineering, Peking University, Beijing 100871, China; Institute of Ocean Research, Peking University, Beijing 100871, China
| | - Yuan Fang
- School of Resource and Environmental Engineering, Hefei University of Technology, Hefei 230000, China
| | - Xin Zheng
- School of Resource and Environmental Engineering, Hefei University of Technology, Hefei 230000, China
| | - Ting Huang
- School of Resource and Environmental Engineering, Hefei University of Technology, Hefei 230000, China
| | - Yong Nie
- College of Engineering, Peking University, Beijing 100871, China.
| | - Xiao-Lei Wu
- College of Engineering, Peking University, Beijing 100871, China; Institute of Ocean Research, Peking University, Beijing 100871, China; Institute of Ecology, Peking University, Beijing 100871, China.
| |
Collapse
|
40
|
Dooley KD, Henry LP, Bergelson J. Impact of timing on the invasion of synthetic bacterial communities. THE ISME JOURNAL 2024; 18:wrae220. [PMID: 39498487 PMCID: PMC11605641 DOI: 10.1093/ismejo/wrae220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Revised: 08/12/2024] [Accepted: 11/03/2024] [Indexed: 11/30/2024]
Abstract
Microbial communities regularly experience ecological invasions that can lead to changes in composition and function. Factors thought to impact susceptibility to invasions, such as diversity and resource use, vary over the course of community assembly. We used synthetic bacterial communities to evaluate the success and impact of invasions occurring at different times during the community assembly process. Fifteen distinct communities were subjected to each of three bacterial invaders at the initial assembly of the community ("initial invasion"), 24 h into community assembly ("early invasion"), when the community was still undergoing transient dynamics, and 7 days into community assembly ("late invasion"), once the community had settled into its final composition. Communities were passaged daily and characterized through sequencing after reaching a stable composition. Invasions often failed to persist over time, particularly in higher richness communities. However, invasions had their largest effect on composition when they occurred before a community had settled into a stable composition. We found instances where an invader was ultimately excluded yet had profound and long-lasting effects on invaded communities. Invasion outcome was positively associated with lower community richness and resource use efficiency by the community, which varied throughout assembly. Our results demonstrate that microbial communities experiencing transient community dynamics are more affected by, and in some instances prone to, invasion, a finding relevant to efforts to modify the composition of microbial communities.
Collapse
Affiliation(s)
- Keven D Dooley
- Committee on Microbiology, University of Chicago, 924 East 57th Street, Chicago, IL 60637, United States
| | - Lucas P Henry
- Center for Genomics and System Biology, Department of Biology, New York University, 12 Waverly Place, New York, NY 10003, United States
| | - Joy Bergelson
- Center for Genomics and System Biology, Department of Biology, New York University, 12 Waverly Place, New York, NY 10003, United States
- Department of Ecology and Evolution, University of Chicago, 1101 East 57th Street, Chicago, IL 60637, United States
| |
Collapse
|
41
|
Van Den Ham KM, Little MR, Bednarski OJ, Fusco EM, Mandal RK, Mitra R, Li S, Doumbo S, Doumtabe D, Kayentao K, Ongoiba A, Traore B, Crompton PD, Schmidt NW. Creation of a non-Western humanized gnotobiotic mouse model through the transplantation of rural African fecal microbiota. Microbiol Spectr 2023; 11:e0155423. [PMID: 37819130 PMCID: PMC10714993 DOI: 10.1128/spectrum.01554-23] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 09/05/2023] [Indexed: 10/13/2023] Open
Abstract
IMPORTANCE There is increasing evidence that microbes residing within the intestines (gut microbiota) play important roles in the well-being of humans. Yet, there are considerable challenges in determining the specific role of gut microbiota in human diseases owing to the complexity of diverse internal and environmental factors that can contribute to diseases. Mice devoid of all microorganisms (germ-free mice) can be colonized with human stool samples to examine the specific contribution of the gut microbiota to a disease. These approaches have been primarily focused on stool samples obtained from individuals in Western countries. Thus, there is limited understanding as to whether the same methods used to colonize germ-free mice with stool from Western individuals would apply to the colonization of germ-free mice with stool from non-Western individuals. Here, we report the results from colonizing germ-free mice with stool samples of Malian children.
Collapse
Affiliation(s)
- Kristin M. Van Den Ham
- Department of Pediatrics, Ryan White Center for Pediatric Infectious Diseases and Global Health, Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Morgan R. Little
- Department of Pediatrics, Ryan White Center for Pediatric Infectious Diseases and Global Health, Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Olivia J. Bednarski
- Department of Pediatrics, Ryan White Center for Pediatric Infectious Diseases and Global Health, Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Elizabeth M. Fusco
- Department of Pediatrics, Ryan White Center for Pediatric Infectious Diseases and Global Health, Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Rabindra K. Mandal
- Department of Pediatrics, Ryan White Center for Pediatric Infectious Diseases and Global Health, Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Riten Mitra
- Department of Bioinformatics and Biostatistics, University of Louisville, Louisville, Kentucky, USA
| | - Shanping Li
- Malaria Infection Biology and Immunity Section, Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, USA
| | - Safiatou Doumbo
- Mali International Center of Excellence in Research, Malaria Research and Training Center, University of Sciences, Techniques and Technologies of Bamako, Bamako, Mali
| | - Didier Doumtabe
- Mali International Center of Excellence in Research, Malaria Research and Training Center, University of Sciences, Techniques and Technologies of Bamako, Bamako, Mali
| | - Kassoum Kayentao
- Mali International Center of Excellence in Research, Malaria Research and Training Center, University of Sciences, Techniques and Technologies of Bamako, Bamako, Mali
| | - Aissata Ongoiba
- Mali International Center of Excellence in Research, Malaria Research and Training Center, University of Sciences, Techniques and Technologies of Bamako, Bamako, Mali
| | - Boubacar Traore
- Mali International Center of Excellence in Research, Malaria Research and Training Center, University of Sciences, Techniques and Technologies of Bamako, Bamako, Mali
| | - Peter D. Crompton
- Malaria Infection Biology and Immunity Section, Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, USA
| | - Nathan W. Schmidt
- Department of Pediatrics, Ryan White Center for Pediatric Infectious Diseases and Global Health, Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana, USA
| |
Collapse
|
42
|
Sidner B, Lerma A, Biswas B, Do TVT, Yu Y, Ronish LA, McCullough H, Auchtung JM, Piepenbrink KH. Flagellin is essential for initial attachment to mucosal surfaces by Clostridioides difficile. Microbiol Spectr 2023; 11:e0212023. [PMID: 37823657 PMCID: PMC10714722 DOI: 10.1128/spectrum.02120-23] [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] [Received: 05/21/2023] [Accepted: 09/04/2023] [Indexed: 10/13/2023] Open
Abstract
IMPORTANCE Clostridioides difficile is one of the leading causes of hospital-acquired infections worldwide and presents challenges in treatment due to recurrent gastrointestinal disease after treatment with antimicrobials. The mechanisms by which C. difficile colonizes the gut represent a key gap in knowledge, including its association with host cells and mucosa. Our results show the importance of flagellin for specific adhesion to mucosal hydrogels and can help to explain prior observations of adhesive defects in flagellin and pilin mutants.
Collapse
Affiliation(s)
- Ben Sidner
- Department of Food Science and Technology, University of Nebraska-Lincoln, Lincoln, Nebraska, USA
| | - Armando Lerma
- Department of Food Science and Technology, University of Nebraska-Lincoln, Lincoln, Nebraska, USA
| | - Baishakhi Biswas
- Department of Food Science and Technology, University of Nebraska-Lincoln, Lincoln, Nebraska, USA
| | - Thi Van Thanh Do
- Department of Food Science and Technology, University of Nebraska-Lincoln, Lincoln, Nebraska, USA
| | - Yafan Yu
- Department of Biochemistry, University of Nebraska-Lincoln, Lincoln, Nebraska, USA
| | - Leslie A. Ronish
- Department of Biochemistry, University of Nebraska-Lincoln, Lincoln, Nebraska, USA
| | - Hugh McCullough
- Department of Food Science and Technology, University of Nebraska-Lincoln, Lincoln, Nebraska, USA
- Nebraska Food for Health Center, University of Nebraska-Lincoln, Lincoln, Nebraska, USA
| | - Jennifer M. Auchtung
- Department of Food Science and Technology, University of Nebraska-Lincoln, Lincoln, Nebraska, USA
- Nebraska Food for Health Center, University of Nebraska-Lincoln, Lincoln, Nebraska, USA
| | - Kurt H. Piepenbrink
- Department of Food Science and Technology, University of Nebraska-Lincoln, Lincoln, Nebraska, USA
- Department of Biochemistry, University of Nebraska-Lincoln, Lincoln, Nebraska, USA
- Nebraska Food for Health Center, University of Nebraska-Lincoln, Lincoln, Nebraska, USA
- Department of Chemistry, University of Nebraska-Lincoln, Lincoln, Nebraska, USA
- Center for Integrated Biomolecular Communication, University of Nebraska-Lincoln, Lincoln, Nebraska, USA
| |
Collapse
|
43
|
Puerta-Alcalde P, Garcia-Vidal C, Soriano A. Prevention and treatment of C. difficile in cancer patients. Curr Opin Infect Dis 2023; 36:473-480. [PMID: 37527003 DOI: 10.1097/qco.0000000000000954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/03/2023]
Abstract
PURPOSE OF REVIEW We provide an update on the recent literature on Clostridioides difficile infection (CDI) in cancer patients. RECENT FINDINGS Distinguishing between colonization and infection remains challenging in cancer patients. Many patients with negative toxin analysis are still treated for CDI, and some meet criteria for severe cases. The incidence of CDI is high in cancer patients, especially those with haematological malignancies. Disruption of the gut microbiome due to antibiotic consumption, chemotherapy and radiotherapy is the primary factor contributing to CDI development. The severity of CDI in cancer patients is often unclear due to the absence of well-defined severity criteria. Certain microbiome species predominance and specific ribotypes have been associated with worse outcomes. Whole genome sequencing could be helpful for differentiating recurrence from reinfection and exploring potential nosocomial transmission. While certain new drugs such as fidaxomicin or bezlotoxumab show promise, the optimal treatment and prevention strategies for CDI in cancer patients remain uncertain. Faecal microbiota transplantation (FMT) holds potential for reducing CDI recurrence rates. SUMMARY Further studies are needed to provide robust recommendations for diagnosis, grading severity, and therapeutic management of CDI in cancer patients. Recurrences are particularly concerning due to subsequent exposition to CDI risk factors.
Collapse
|
44
|
Shah SS, Noman O, Jaiswal N. Unveiling the Gut Microbiome: How Junk Food Impacts the Gut. Cureus 2023; 15:e49179. [PMID: 38130525 PMCID: PMC10734656 DOI: 10.7759/cureus.49179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Accepted: 11/21/2023] [Indexed: 12/23/2023] Open
Abstract
The human gut microbiome, a complex community of microorganisms, profoundly influences human health and disease. Bacteroidetes and Firmicutes make up the majority of the normal human gut microbiota. These microorganisms wield considerable influence over our physiological functions, impacting both our well-being and our susceptibility to disease. The surge of interest in the gut microbiome over the past decade has been remarkable. Once overlooked, the gastrointestinal tract's microbiota has gained recognition for its significance in maintaining optimal health. The food industry has capitalized on this, flooding the market with "probiotic" and "fermented" products. This article aims to provide a critical review of the current literature on the gut microbiome and its significance in human health, with a particular focus on the impact of dietary choices, especially junk food, on the composition and function of the gut microbiota. Microbes possess the remarkable ability to unlock nutrients from otherwise indigestible substances. The gut microbiome of individuals who consume healthy foods and those who prefer junk food varies significantly. Healthy diets promote a diverse and beneficial gut microbiome, while junk food consumption often leads to a less diverse microbiome with negative consequences for health.
Collapse
Affiliation(s)
- Sania S Shah
- Microbiology, Datta Meghe Medical College, Datta Meghe Institute of Higher Education and Research (DU), Wardha, IND
| | - Obaid Noman
- Pathology, Datta Meghe Medical College, Datta Meghe Institute of Higher Education and Research (DU), Wardha, IND
| | - Neha Jaiswal
- Pathology, Datta Meghe Medical College, Datta Meghe Institute of Higher Education and Research (DU), Wardha, IND
| |
Collapse
|
45
|
Worldwide Antimicrobial Resistance National/International Network Group (WARNING) Collaborators, Sartelli M, Barie PS, Coccolini F, Abbas M, Abbo LM, Abdukhalilova GK, Abraham Y, Abubakar S, Abu-Zidan FM, Adebisi YA, Adamou H, Afandiyeva G, Agastra E, Alfouzan WA, Al-Hasan MN, Ali S, Ali SM, Allaw F, Allwell-Brown G, Amir A, Amponsah OKO, Al Omari A, Ansaloni L, Ansari S, Arauz AB, Augustin G, Awazi B, Azfar M, Bah MSB, Bala M, Banagala ASK, Baral S, Bassetti M, Bavestrello L, Beilman G, Bekele K, Benboubker M, Beović B, Bergamasco MD, Bertagnolio S, Biffl WL, Blot S, Boermeester MA, Bonomo RA, Brink A, Brusaferro S, Butemba J, Caínzos MA, Camacho-Ortiz A, Canton R, Cascio A, Cassini A, Cástro-Sanchez E, Catarci M, Catena R, Chamani-Tabriz L, Chandy SJ, Charani E, Cheadle WG, Chebet D, Chikowe I, Chiara F, Cheng VCC, Chioti A, Cocuz ME, Coimbra R, Cortese F, Cui Y, Czepiel J, Dasic M, de Francisco Serpa N, de Jonge SW, Delibegovic S, Dellinger EP, Demetrashvili Z, De Palma A, De Silva D, De Simone B, De Waele J, Dhingra S, Diaz JJ, Dima C, Dirani N, Dodoo CC, Dorj G, Duane TM, Eckmann C, Egyir B, Elmangory MM, Enani MA, Ergonul O, Escalera-Antezana JP, Escandon K, Ettu AWOO, Fadare JO, Fantoni M, Farahbakhsh M, Faro MP, Ferreres A, et alWorldwide Antimicrobial Resistance National/International Network Group (WARNING) Collaborators, Sartelli M, Barie PS, Coccolini F, Abbas M, Abbo LM, Abdukhalilova GK, Abraham Y, Abubakar S, Abu-Zidan FM, Adebisi YA, Adamou H, Afandiyeva G, Agastra E, Alfouzan WA, Al-Hasan MN, Ali S, Ali SM, Allaw F, Allwell-Brown G, Amir A, Amponsah OKO, Al Omari A, Ansaloni L, Ansari S, Arauz AB, Augustin G, Awazi B, Azfar M, Bah MSB, Bala M, Banagala ASK, Baral S, Bassetti M, Bavestrello L, Beilman G, Bekele K, Benboubker M, Beović B, Bergamasco MD, Bertagnolio S, Biffl WL, Blot S, Boermeester MA, Bonomo RA, Brink A, Brusaferro S, Butemba J, Caínzos MA, Camacho-Ortiz A, Canton R, Cascio A, Cassini A, Cástro-Sanchez E, Catarci M, Catena R, Chamani-Tabriz L, Chandy SJ, Charani E, Cheadle WG, Chebet D, Chikowe I, Chiara F, Cheng VCC, Chioti A, Cocuz ME, Coimbra R, Cortese F, Cui Y, Czepiel J, Dasic M, de Francisco Serpa N, de Jonge SW, Delibegovic S, Dellinger EP, Demetrashvili Z, De Palma A, De Silva D, De Simone B, De Waele J, Dhingra S, Diaz JJ, Dima C, Dirani N, Dodoo CC, Dorj G, Duane TM, Eckmann C, Egyir B, Elmangory MM, Enani MA, Ergonul O, Escalera-Antezana JP, Escandon K, Ettu AWOO, Fadare JO, Fantoni M, Farahbakhsh M, Faro MP, Ferreres A, Flocco G, Foianini E, Fry DE, Garcia AF, Gerardi C, Ghannam W, Giamarellou H, Glushkova N, Gkiokas G, Goff DA, Gomi H, Gottfredsson M, Griffiths EA, Guerra Gronerth RI, Guirao X, Gupta YK, Halle-Ekane G, Hansen S, Haque M, Hardcastle TC, Hayman DTS, Hecker A, Hell M, Ho VP, Hodonou AM, Isik A, Islam S, Itani KMF, Jaidane N, Jammer I, Jenkins DR, Kamara IF, Kanj SS, Jumbam D, Keikha M, Khanna AK, Khanna S, Kapoor G, Kapoor G, Kariuki S, Khamis F, Khokha V, Kiggundu R, Kiguba R, Kim HB, Kim PK, Kirkpatrick AW, Kluger Y, Ko WC, Kok KYY, Kotecha V, Kouma I, Kovacevic B, Krasniqi J, Krutova M, Kryvoruchko I, Kullar R, Labi KA, Labricciosa FM, Lakoh S, Lakatos B, Lansang MAD, Laxminarayan R, Lee YR, Leone M, Leppaniemi A, Hara GL, Litvin A, Lohsiriwat V, Machain GM, Mahomoodally F, Maier RV, Majumder MAA, Malama S, Manasa J, Manchanda V, Manzano-Nunez R, Martínez-Martínez L, Martin-Loeches I, Marwah S, Maseda E, Mathewos M, Maves RC, McNamara D, Memish Z, Mertz D, Mishra SK, Montravers P, Moro ML, Mossialos E, Motta F, Mudenda S, Mugabi P, Mugisha MJM, Mylonakis E, Napolitano LM, Nathwani D, Nkamba L, Nsutebu EF, O’Connor DB, Ogunsola S, Jensen PØ, Ordoñez JM, Ordoñez CA, Ottolino P, Ouedraogo AS, Paiva JA, Palmieri M, Pan A, Pant N, Panyko A, Paolillo C, Patel J, Pea F, Petrone P, Petrosillo N, Pintar T, Plaudis H, Podda M, Ponce-de-Leon A, Powell SL, Puello-Guerrero A, Pulcini C, Rasa K, Regimbeau JM, Rello J, Retamozo-Palacios MR, Reynolds-Campbell G, Ribeiro J, Rickard J, Rocha-Pereira N, Rosenthal VD, Rossolini GM, Rwegerera GM, Rwigamba M, Sabbatucci M, Saladžinskas Ž, Salama RE, Sali T, Salile SS, Sall I, Kafil HS, Sakakushev BE, Sawyer RG, Scatizzi M, Seni J, Septimus EJ, Sganga G, Shabanzadeh DM, Shelat VG, Shibabaw A, Somville F, Souf S, Stefani S, Tacconelli E, Tan BK, Tattevin P, Rodriguez-Taveras C, Telles JP, Téllez-Almenares O, Tessier J, Thang NT, Timmermann C, Timsit JF, Tochie JN, Tolonen M, Trueba G, Tsioutis C, Tumietto F, Tuon FF, Ulrych J, Uranues S, van Dongen M, van Goor H, Velmahos GC, Vereczkei A, Viaggi B, Viale P, Vila J, Voss A, Vraneš J, Watkins RR, Wanjiru-Korir N, Waworuntu O, Wechsler-Fördös A, Yadgarova K, Yahaya M, Yahya AI, Xiao Y, Zakaria AD, Zakrison TL, Zamora Mesia V, Siquini W, Darzi A, Pagani L, Catena F. Ten golden rules for optimal antibiotic use in hospital settings: the WARNING call to action. World J Emerg Surg 2023; 18:50. [PMID: 37845673 PMCID: PMC10580644 DOI: 10.1186/s13017-023-00518-3] [Show More Authors] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Accepted: 09/23/2023] [Indexed: 10/18/2023] Open
Abstract
Antibiotics are recognized widely for their benefits when used appropriately. However, they are often used inappropriately despite the importance of responsible use within good clinical practice. Effective antibiotic treatment is an essential component of universal healthcare, and it is a global responsibility to ensure appropriate use. Currently, pharmaceutical companies have little incentive to develop new antibiotics due to scientific, regulatory, and financial barriers, further emphasizing the importance of appropriate antibiotic use. To address this issue, the Global Alliance for Infections in Surgery established an international multidisciplinary task force of 295 experts from 115 countries with different backgrounds. The task force developed a position statement called WARNING (Worldwide Antimicrobial Resistance National/International Network Group) aimed at raising awareness of antimicrobial resistance and improving antibiotic prescribing practices worldwide. The statement outlined is 10 axioms, or "golden rules," for the appropriate use of antibiotics that all healthcare workers should consistently adhere in clinical practice.
Collapse
|
46
|
Chesdachai S, Yetmar ZA, Mendoza MA, Ranganath N, Schettle SD, Boilson BA, Shah AS, Razonable RR. Clinical Characteristics and Outcomes of Clostridioides difficile Infection in Patients With Left Ventricular Assist Device. ASAIO J 2023; 69:950-955. [PMID: 37367716 DOI: 10.1097/mat.0000000000002008] [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] [Indexed: 06/28/2023] Open
Abstract
The literature regarding Clostridioides difficile infection (CDI) in left ventricular assist devices (LVADs) patients is limited. Therefore, we aimed to characterize the clinical course, risk factors, management, and outcomes of LVAD patients who developed CDI. Adult patients who underwent LVAD placement during 2010-2022 and developed CDI were included. To determine risk factors and outcomes, we matched CDI patients with LVAD patients who did not develop CDI. Each CDI case was matched with up to two control subjects by age, sex, and time from LVAD implantation. Forty-seven of 393 LVAD patients (12.0%) developed CDI. The median time from LVAD implantation to CDI was 147 days (interquartile range 22.5-647.0). The most common CDI treatment was oral vancomycin (n = 26, 55.3%). Thirteen patients (27.7%) required treatment extension because of a lack of clinical response. Three patients (6.4%) developed recurrent CDI. When 42 cases were matched to 79 control subjects, antibiotic exposure within 90 days was significantly associated with CDI (adjusted odds ratio 5.77; 95% confidence interval, 1.87-17.74; p = 0.002). Moreover, CDI was associated with 1 year mortality (adjusted hazard ratio 2.62; 95% confidence interval, 1.18-5.82; p = 0.018). This infection occurs most often within the first year after LVAD implantation and was associated with 1 year mortality. Antibiotic exposure is an important risk for CDI.
Collapse
Affiliation(s)
- Supavit Chesdachai
- From the Division of Public Health, Infectious Diseases and Occupational Medicine, Department of Medicine, Mayo Clinic, Rochester, MN
| | - Zachary A Yetmar
- From the Division of Public Health, Infectious Diseases and Occupational Medicine, Department of Medicine, Mayo Clinic, Rochester, MN
| | - Maria A Mendoza
- From the Division of Public Health, Infectious Diseases and Occupational Medicine, Department of Medicine, Mayo Clinic, Rochester, MN
| | - Nischal Ranganath
- From the Division of Public Health, Infectious Diseases and Occupational Medicine, Department of Medicine, Mayo Clinic, Rochester, MN
| | - Sarah D Schettle
- Department of Cardiovascular Surgery, Mayo Clinic, Rochester, MN
| | - Barry A Boilson
- Department of Advanced Heart Failure, Transplant, and LVAD, Mayo Clinic, Rochester, MN
| | - Aditya S Shah
- From the Division of Public Health, Infectious Diseases and Occupational Medicine, Department of Medicine, Mayo Clinic, Rochester, MN
| | - Raymund R Razonable
- From the Division of Public Health, Infectious Diseases and Occupational Medicine, Department of Medicine, Mayo Clinic, Rochester, MN
| |
Collapse
|
47
|
Sriboonvorakul N, Chotivanich K, Silachamroon U, Phumratanaprapin W, Adams JH, Dondorp AM, Leopold SJ. Intestinal injury and the gut microbiota in patients with Plasmodium falciparum malaria. PLoS Pathog 2023; 19:e1011661. [PMID: 37856470 PMCID: PMC10586672 DOI: 10.1371/journal.ppat.1011661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2023] Open
Abstract
The pathophysiology of severe falciparum malaria involves a complex interaction between the host, parasite, and gut microbes. In this review, we focus on understanding parasite-induced intestinal injury and changes in the human intestinal microbiota composition in patients with Plasmodium falciparum malaria. During the blood stage of P. falciparum infection, infected red blood cells adhere to the vascular endothelium, leading to widespread microcirculatory obstruction in critical tissues, including the splanchnic vasculature. This process may cause intestinal injury and gut leakage. Epidemiological studies indicate higher rates of concurrent bacteraemia in severe malaria cases. Furthermore, severe malaria patients exhibit alterations in the composition and diversity of the intestinal microbiota, although the exact contribution to pathophysiology remains unclear. Mouse studies have demonstrated that the gut microbiota composition can impact susceptibility to Plasmodium infections. In patients with severe malaria, the microbiota shows an enrichment of pathobionts, including pathogens that are known to cause concomitant bloodstream infections. Microbial metabolites have also been detected in the plasma of severe malaria patients, potentially contributing to metabolic acidosis and other clinical complications. However, establishing causal relationships requires intervention studies targeting the gut microbiota.
Collapse
Affiliation(s)
- Natthida Sriboonvorakul
- Department of Clinical Tropical Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Kesinee Chotivanich
- Department of Clinical Tropical Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Udomsak Silachamroon
- Department of Clinical Tropical Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Weerapong Phumratanaprapin
- Department of Clinical Tropical Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - John H. Adams
- Center for Global Health and Infectious Diseases Research, College of Public Health, University of South Florida, Tampa, Florida, United States of America
| | - Arjen M. Dondorp
- Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Centre for Tropical Medicine, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, United Kingdom
| | - Stije J. Leopold
- Department of Internal Medicine, Division of Infectious Diseases, Amsterdam University Medical Center, location AMC, the Netherlands
| |
Collapse
|
48
|
Iancu MA, Profir M, Roşu OA, Ionescu RF, Cretoiu SM, Gaspar BS. Revisiting the Intestinal Microbiome and Its Role in Diarrhea and Constipation. Microorganisms 2023; 11:2177. [PMID: 37764021 PMCID: PMC10538221 DOI: 10.3390/microorganisms11092177] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 08/24/2023] [Accepted: 08/25/2023] [Indexed: 09/29/2023] Open
Abstract
The gut microbiota represents a community of microorganisms (bacteria, fungi, archaea, viruses, and protozoa) that colonize the gut and are responsible for gut mucosal structural integrity and immune and metabolic homeostasis. The relationship between the gut microbiome and human health has been intensively researched in the past years. It is now widely recognized that gut microbial composition is highly responsible for the general health of the host. Among the diseases that have been linked to an altered gut microbial population are diarrheal illnesses and functional constipation. The capacity of probiotics to modulate the gut microbiome population, strengthen the intestinal barrier, and modulate the immune system together with their antioxidant properties have encouraged the research of probiotic therapy in many gastrointestinal afflictions. Dietary and lifestyle changes and the use of probiotics seem to play an important role in easing constipation and effectively alleviating diarrhea by suppressing the germs involved. This review aims to describe how probiotic bacteria and the use of specific strains could interfere and bring benefits as an associated treatment for diarrhea and constipation.
Collapse
Affiliation(s)
- Mihaela Adela Iancu
- Department of Family Medicine, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania;
| | - Monica Profir
- Department of Oncology, Elias University Emergency Hospital, 011461 Bucharest, Romania; (M.P.); (O.A.R.)
| | - Oana Alexandra Roşu
- Department of Oncology, Elias University Emergency Hospital, 011461 Bucharest, Romania; (M.P.); (O.A.R.)
| | - Ruxandra Florentina Ionescu
- Department of Morphological Sciences, Cell and Molecular Biology and Histology, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania;
- Department of Cardiology I, “Dr. Carol Davila” Central Military Emergency Hospital, 010825 Bucharest, Romania
| | - Sanda Maria Cretoiu
- Department of Morphological Sciences, Cell and Molecular Biology and Histology, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania;
| | - Bogdan Severus Gaspar
- Surgery Clinic, Emergency Clinical Hospital, 014461 Bucharest, Romania;
- Department of Surgery, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania
| |
Collapse
|
49
|
Wang S, Xiang L, li F, Deng W, lv P, Chen Y. Butyrate Protects against Clostridium difficile Infection by Regulating Bile Acid Metabolism. Microbiol Spectr 2023; 11:e0447922. [PMID: 37350595 PMCID: PMC10434071 DOI: 10.1128/spectrum.04479-22] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Accepted: 06/03/2023] [Indexed: 06/24/2023] Open
Abstract
Clostridium difficile infection (CDI) is caused by a prevalent nosocomial enteric pathogen, leading to high morbidity and mortality. CDI recurrence after antibiotic treatment is high; therefore, it is necessary to develop novel therapeutics against this enteric pathogen. Butyrate is used to treat many diseases because it provides energy, has anti-inflammatory properties, and maintains intestinal barrier function. An anti-CDI effect for butyrate has been reported; however, the specific mechanism remains elusive. This study aimed to explore the potential role and mechanism of butyrate in the treatment of CDI. Using a CDI mouse model, we found that butyrate significantly inhibited CDI development by regulating bile acid metabolism. Dysregulation of fecal bile acid was significantly higher, and levels of short-chain fatty acids were significantly lower in patients with CDI than those in controls. In CDI mice, butyrate exhibited a protective role by enhancing barrier protection, exerting anti-inflammatory effects, and regulating bile acid metabolism. Butyrate treatment also regulated the production of bile salt hydrolase (BSH) flora and activated farnesoid X receptor (FXR), and its therapeutic effects were reduced in CDI mice treated with BSH or FXR inhibitors. Thus, butyrate treatment may serve as a novel therapeutic approach for patients with CDI. IMPORTANCE Here, we show that levels of fecal short-chain fatty acids (SCFAs), particularly butyrate, are reduced, and normal colon structure is damaged in patients with CDI compared with those in healthy individuals. Bile acid (BA) metabolic disorder in patients with CDI is characterized by increased primary BA levels and decreased secondary BAs. In mice, butyrate alters BA metabolism in CDI and may play a vital role in CDI treatment by promoting secondary BA metabolism. Lastly, butyrate-mediated therapeutic effects in CDI require FXR. Our findings demonstrate that butyrate treatment significantly decreases the severity of CDI-induced colitis in mice and affects BA metabolism and FXR activation, which provides a potential alternative treatment for CDI.
Collapse
Affiliation(s)
- Siqi Wang
- Department of Gastroenterology, The First Affiliated of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China
- Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Leyang Xiang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Fang li
- Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Department of Gastroenterology, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Wenlin Deng
- Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Pinjing lv
- Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Ye Chen
- Department of Gastroenterology, The First Affiliated of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China
- Department of Gastroenterology, Integrative Clinical Microecology Center, Shenzhen Hospital, Southern Medical University, Shenzhen, China
| |
Collapse
|
50
|
Li Z, Yuan H, Chu H, Yang L. The Crosstalk between Gut Microbiota and Bile Acids Promotes the Development of Non-Alcoholic Fatty Liver Disease. Microorganisms 2023; 11:2059. [PMID: 37630619 PMCID: PMC10459427 DOI: 10.3390/microorganisms11082059] [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: 06/21/2023] [Revised: 07/27/2023] [Accepted: 08/01/2023] [Indexed: 08/27/2023] Open
Abstract
Recently the roles of gut microbiota are highly regarded in the pathogenesis of nonalcoholic fatty liver disease (NAFLD). The intestinal bacteria regulate the metabolism of bile acids depending on bile salt hydrolase (BSH), 7-dehydroxylation, hydroxysteroid dehydrogenase (HSDH), or amide conjugation reaction, thus exerting effects on NAFLD development through bile acid receptors such as farnesoid X receptor (FXR), Takeda G-protein-coupled bile acid protein 5 (TGR5), and vitamin D receptor (VDR), which modulate nutrient metabolism and insulin sensitivity via interacting with downstream molecules. Reversely, the composition of gut microbiota is also affected by the level of bile acids in turn. We summarize the mutual regulation between the specific bacteria and bile acids in NAFLD and the latest clinical research based on microbiota and bile acids, which facilitate the development of novel treatment modalities in NAFLD.
Collapse
Affiliation(s)
| | | | | | - Ling Yang
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China; (Z.L.); (H.Y.); (H.C.)
| |
Collapse
|