51
|
Diseases of the Small and Large Bowel. Fam Med 2021. [DOI: 10.1007/978-1-4939-0779-3_181-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
52
|
Soto Chervin C, Gajewski T. Microbiome-based interventions: therapeutic strategies in cancer immunotherapy. IMMUNO-ONCOLOGY TECHNOLOGY 2020; 8:12-20. [PMID: 35757563 PMCID: PMC9216398 DOI: 10.1016/j.iotech.2020.11.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The composition of the commensal microbiota has recently emerged as a key element influencing the efficacy of cancer treatments. It has become apparent that the interplay between the microbiome and immune system within the host influences the response to immunotherapy, particularly immune checkpoint inhibitor therapy. Identifying the key components of the gut microbiota that influence this response is paramount for designing therapeutic interventions to enhance the response to cancer therapy. This review will discuss strategies being considered to modulate the gut microbiota, including fecal microbiota transplantation, administration of defined bacterial isolates as well as bacterial consortia, supplementation with probiotics, and lifestyle modifications such as dietary changes. Understanding the influence of the complex variables of the human microbiota on the effectiveness of cancer therapy will help drive the clinical design of microbial-based interventions in the field of oncology.
Collapse
Affiliation(s)
- C. Soto Chervin
- Department of Pathology and Department of Medicine, Section of Hematology/Oncology, University of Chicago, Chicago, USA
| | - T.F. Gajewski
- Department of Pathology and Department of Medicine, Section of Hematology/Oncology, University of Chicago, Chicago, USA
| |
Collapse
|
53
|
Lee KA, Shaw HM, Bataille V, Nathan P, Spector TD. Role of the gut microbiome for cancer patients receiving immunotherapy: Dietary and treatment implications. Eur J Cancer 2020; 138:149-155. [PMID: 32889369 DOI: 10.1016/j.ejca.2020.07.026] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2020] [Revised: 07/01/2020] [Accepted: 07/22/2020] [Indexed: 01/01/2023]
Abstract
Immune-checkpoint inhibitors (ICIs) have revolutionised the therapeutic landscape for multiple malignancies and the health of the gut microbiome (GM) is strongly linked with therapeutic responses to ICI. This review explores the implications of diet and medication on the GM for patients receiving ICI. Clinical trials are underway to explore the impact of factors such as faecal microbiota transfer, probiotics, prebiotics, bacteria consortia and a number of dietary interventions on patients receiving ICI. Randomised controlled trials are lacking, and inferences are currently based on short-term clinical and observational studies. Antibiotics should be avoided before ICI initiation, and depending on prospective data, future consideration may be given to temporary delay of initiation of non-urgent ICI if patient has had broad spectrum antibiotics within 1 month of planned treatment initiation. Proton pump inhibitor use should be discontinued when not clearly indicated and potential switch to a histamine H2-receptor antagonist considered. Patients should be advised to minimise animal meat intake and maximise plants, aiming to consume ≥30 plant types weekly. A high fibre intake (>30 g/day) has been seen to be beneficial in increasing the chance of ICI response. Fermented foods may have a beneficial effect on the GM and should be introduced where possible. Ideally, all patients should be referred to a nutritionist or dietician with knowledge of GM before commencing ICI.
Collapse
Affiliation(s)
- Karla A Lee
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK; Department of Medical Oncology, Mount Vernon Hospital, Northwood, UK; Department of Medical Oncology, The Royal Marsden, London, UK.
| | - Heather M Shaw
- Department of Medical Oncology, Mount Vernon Hospital, Northwood, UK; Early Phase Trial Unit, Department of Medical Oncology, University College London Hospital, London, UK
| | - Veronique Bataille
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK; Department of Dermatology, Mount Vernon Hospital, Northwood, UK
| | - Paul Nathan
- Department of Medical Oncology, Mount Vernon Hospital, Northwood, UK
| | - Tim D Spector
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK
| |
Collapse
|
54
|
Bender AC, Faulkner JA, Tulimieri K, Boise TH, Elkins KM. High Resolution Melt Assays to Detect and Identify Vibrio parahaemolyticus, Bacillus cereus, Escherichia coli, and Clostridioides difficile Bacteria. Microorganisms 2020; 8:microorganisms8040561. [PMID: 32295121 PMCID: PMC7232521 DOI: 10.3390/microorganisms8040561] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 04/02/2020] [Accepted: 04/10/2020] [Indexed: 01/05/2023] Open
Abstract
Over one hundred bacterial species have been determined to comprise the human microbiota in a healthy individual. Bacteria including Escherichia coli, Bacillus cereus, Clostridioides difficile, and Vibrio parahaemolyticus are found inside of the human body and B. cereus and E. coli are also found on the skin. These bacteria can act as human pathogens upon ingestion of contaminated food or water, if they enter an open wound, or antibiotics, and environment or stress can alter the microbiome. In this study, we present new polymerase chain reaction (PCR) high-resolution melt (HRM) assays to detect and identify the above microorganisms. Amplified DNA from C. difficile, E. coli, B. cereus, and V. parahaemolyticus melted at 80.37 ± 0.45 °C, 82.15 ± 0.37 °C, 84.43 ± 0.50 °C, and 86.74 ± 0.65 °C, respectively. A triplex PCR assay was developed to simultaneously detect and identify E. coli, B. cereus, and V. parahaemolyticus, and cultured microorganisms were successfully amplified, detected, and identified. The assays demonstrated sensitivity, specificity, reproducibility, and robustness in testing.
Collapse
|
55
|
Jiménez-Avalos JA, Arrevillaga-Boni G, González-López L, García-Carvajal ZY, González-Avila M. Classical methods and perspectives for manipulating the human gut microbial ecosystem. Crit Rev Food Sci Nutr 2020; 61:234-258. [PMID: 32114770 DOI: 10.1080/10408398.2020.1724075] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
A healthy Human Gut Microbial Ecosystem (HGME) is a necessary condition for maintaining the orderly function of the whole body. Major alterations in the normal gut microbial composition, activity and functionality (dysbiosis) by an environmental or host-related disruptive event, can compromise metabolic, inflammatory, and neurological processes, causing disorders such as obesity, inflammatory bowel disease, colorectal cancer, and depressive episodes. The restore or the maintaining of the homeostatic balance of Gut Microbiota (GM) populations (eubiosis) is possible through diet, the use of probiotics, prebiotics, antibiotics, and even Fecal Microbiota Transplantation (FMT). Although these "classic methods" represent an effective and accepted way to modulate GM, the complexity of HGME requires new approaches to control it in a more appropriate way. Among the most promising emergent strategies for modulating GM are the use of engineered nanomaterials (metallic nanoparticles (NP), polymeric-NP, quantum dots, micelles, dendrimers, and liposomes); phagotherapy (i.e., phages linked with the CRISPR/Cas9 system), and the use of antimicrobial peptides, non-antibiotic drugs, vaccines, and immunoglobulins. Here we review the current state of development, implications, advantages, disadvantages, and perspectives of the different approaches for manipulating HGME.
Collapse
Affiliation(s)
- Jorge Armando Jiménez-Avalos
- Medical and Pharmaceutical Biotechnology Department, Centro de Investigación y Asistencia en Tecnología y Diseño del Estado de Jalisco (CIATEJ), Guadalajara, Jalisco, Mexico
| | - Gerardo Arrevillaga-Boni
- Medical and Pharmaceutical Biotechnology Department, Centro de Investigación y Asistencia en Tecnología y Diseño del Estado de Jalisco (CIATEJ), Guadalajara, Jalisco, Mexico
| | | | - Zaira Yunuen García-Carvajal
- Medical and Pharmaceutical Biotechnology Department, Centro de Investigación y Asistencia en Tecnología y Diseño del Estado de Jalisco (CIATEJ), Guadalajara, Jalisco, Mexico
| | - Marisela González-Avila
- Medical and Pharmaceutical Biotechnology Department, Centro de Investigación y Asistencia en Tecnología y Diseño del Estado de Jalisco (CIATEJ), Guadalajara, Jalisco, Mexico
| |
Collapse
|
56
|
Xavier JB, Young VB, Skufca J, Ginty F, Testerman T, Pearson AT, Macklin P, Mitchell A, Shmulevich I, Xie L, Caporaso JG, Crandall KA, Simone NL, Godoy-Vitorino F, Griffin TJ, Whiteson KL, Gustafson HH, Slade DJ, Schmidt TM, Walther-Antonio MRS, Korem T, Webb-Robertson BJM, Styczynski MP, Johnson WE, Jobin C, Ridlon JM, Koh AY, Yu M, Kelly L, Wargo JA. The Cancer Microbiome: Distinguishing Direct and Indirect Effects Requires a Systemic View. Trends Cancer 2020; 6:192-204. [PMID: 32101723 PMCID: PMC7098063 DOI: 10.1016/j.trecan.2020.01.004] [Citation(s) in RCA: 155] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 12/29/2019] [Accepted: 01/06/2020] [Indexed: 02/06/2023]
Abstract
The collection of microbes that live in and on the human body - the human microbiome - can impact on cancer initiation, progression, and response to therapy, including cancer immunotherapy. The mechanisms by which microbiomes impact on cancers can yield new diagnostics and treatments, but much remains unknown. The interactions between microbes, diet, host factors, drugs, and cell-cell interactions within the cancer itself likely involve intricate feedbacks, and no single component can explain all the behavior of the system. Understanding the role of host-associated microbial communities in cancer systems will require a multidisciplinary approach combining microbial ecology, immunology, cancer cell biology, and computational biology - a systems biology approach.
Collapse
Affiliation(s)
- Joao B Xavier
- Program for Computational and Systems Biology, Memorial Sloan-Kettering Cancer Center, New York, NY, USA.
| | - Vincent B Young
- Department of Internal Medicine, Division of Infectious Diseases, The University of Michigan Medical School, Ann Arbor, MI, USA
| | - Joseph Skufca
- Department of Mathematics, Clarkson University, Potsdam, NY, USA
| | | | - Traci Testerman
- Department of Pathology, Microbiology, and Immunology, University of South Carolina School of Medicine, Columbia, SC, USA
| | - Alexander T Pearson
- Section of Hematology/Oncology, Department of Medicine, Comprehensive Cancer Center, University of Chicago, Chicago, Illinois, IL, USA
| | - Paul Macklin
- Intelligent Systems Engineering, Indiana University, Bloomington, IN, USA
| | - Amir Mitchell
- Program in Systems Biology, University of Massachusetts Medical School, Worcester, MA, USA
| | | | - Lei Xie
- Hunter College, Department of Computer Science, New York, NY, USA
| | - J Gregory Caporaso
- Center for Applied Microbiome Science, Pathogen and Microbiome Institute, Northern Arizona University, Flagstaff, AZ, USA
| | - Keith A Crandall
- Computational Biology Institute, Milken Institute School of Public Health, George Washington University, Washington, DC, USA
| | - Nicole L Simone
- Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - Filipa Godoy-Vitorino
- Department of Microbiology and Medical Zoology, School of Medicine, University of Puerto Rico, San Juan, Puerto Rico
| | - Timothy J Griffin
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, MN, USA
| | - Katrine L Whiteson
- Department of Molecular Biology and Biochemistry, University of California Irvine, Irvine, CA, USA
| | - Heather H Gustafson
- Seattle Children's Research Institute, Ben Towne Center for Childhood Cancer Research, Seattle, WA, USA
| | - Daniel J Slade
- Department of Biochemistry, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA
| | | | - Marina R S Walther-Antonio
- Department of Surgery, Department of Obstetrics and Gynecology, and Microbiome Program, Center for Individualized Medicine, Mayo Clinic, Rochester, MN, USA
| | - Tal Korem
- Department of Systems Biology, Columbia University, New York, NY, USA
| | | | - Mark P Styczynski
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - W Evan Johnson
- Division of Computational Biomedicine, Boston University School of Medicine, Boston, MA, USA
| | - Christian Jobin
- Departments of Medicine, Anatomy, and Cell Biology, and of Infectious Diseases and Immunology, University of Florida, Gainesville, FL, USA
| | - Jason M Ridlon
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Andrew Y Koh
- University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Michael Yu
- Toyota Technological Institute at Chicago, Chicago, IL, USA
| | | | - Jennifer A Wargo
- The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
57
|
Li X, Chu Q, Huang Y, Xiao Y, Song L, Zhu S, Kang Y, Lu S, Xu J, Ren Z. Consortium of Probiotics Attenuates Colonization of Clostridioides difficile. Front Microbiol 2019; 10:2871. [PMID: 31921049 PMCID: PMC6920126 DOI: 10.3389/fmicb.2019.02871] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Accepted: 11/28/2019] [Indexed: 12/11/2022] Open
Abstract
Clostridioides difficile infection (CDI) is increasing morbidity and mortality rates globally. Fecal microbiota transplantation (FMT), an effective therapy for eliminating Clostridioides difficile (C. difficile), cannot be used extensive due to a range of challenges. Probiotics thus constitutes a promising alternative therapy. In our study, we evaluated the effect of consortium of probiotics including five Lactobacilli strains and two Bifidobacterium strains on the colonization of toxigenic BI/NAP1/027 C. difficile in a mouse model. The results of 16S rRNA sequencing and targeted metabolomics showed the consortium of probiotics effectively decreased the colonization of C. difficile, changed the α- and β-diversity of the gut microbiota, decreased the primary bile acids, and increased the secondary bile acids. Spearman’s correlation showed that some of the OTUs such as Akkermansia, Bacteroides, Blautia et al. were positively correlated with C. difficile numbers and the primary bile acids, and negatively correlated with the secondary bile acids. However, some of the OTUs, such as Butyricicoccus, Ruminococcus, and Rikenellaceae, were negatively correlated with C. difficile copies and the primary bile acids, and positively correlated with the secondary bile acids. In summary, the consortium of probiotics effectively decreases the colonization of C. difficile, probably via alteration of gut microbiota and bile acids. Our probiotics mixture thus offers a promising FMT alternative.
Collapse
Affiliation(s)
- Xianping Li
- State Key Laboratory for Infectious Disease Prevention and Control, National Institute for Communicable Disease Control and Prevention, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Chinese Center for Disease Control and Prevention, Changping, China.,Research Units of Discovery of Unknown Bacteria and Function (2018 RU010), Chinese Academy of Medical Sciences, Beijing, China
| | - Qiongfang Chu
- State Key Laboratory for Infectious Disease Prevention and Control, National Institute for Communicable Disease Control and Prevention, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Chinese Center for Disease Control and Prevention, Changping, China.,Research Units of Discovery of Unknown Bacteria and Function (2018 RU010), Chinese Academy of Medical Sciences, Beijing, China.,Beijing Dongcheng District Longtan Community Health Center, Beijing, China
| | - Yuanming Huang
- State Key Laboratory for Infectious Disease Prevention and Control, National Institute for Communicable Disease Control and Prevention, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Chinese Center for Disease Control and Prevention, Changping, China.,Research Units of Discovery of Unknown Bacteria and Function (2018 RU010), Chinese Academy of Medical Sciences, Beijing, China
| | - Yuchun Xiao
- State Key Laboratory for Infectious Disease Prevention and Control, National Institute for Communicable Disease Control and Prevention, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Chinese Center for Disease Control and Prevention, Changping, China.,Research Units of Discovery of Unknown Bacteria and Function (2018 RU010), Chinese Academy of Medical Sciences, Beijing, China
| | - Liqiong Song
- State Key Laboratory for Infectious Disease Prevention and Control, National Institute for Communicable Disease Control and Prevention, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Chinese Center for Disease Control and Prevention, Changping, China.,Research Units of Discovery of Unknown Bacteria and Function (2018 RU010), Chinese Academy of Medical Sciences, Beijing, China
| | - Siyi Zhu
- State Key Laboratory for Infectious Disease Prevention and Control, National Institute for Communicable Disease Control and Prevention, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Chinese Center for Disease Control and Prevention, Changping, China.,Research Units of Discovery of Unknown Bacteria and Function (2018 RU010), Chinese Academy of Medical Sciences, Beijing, China
| | - Ying Kang
- State Key Laboratory for Infectious Disease Prevention and Control, National Institute for Communicable Disease Control and Prevention, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Chinese Center for Disease Control and Prevention, Changping, China.,Research Units of Discovery of Unknown Bacteria and Function (2018 RU010), Chinese Academy of Medical Sciences, Beijing, China
| | - Shan Lu
- State Key Laboratory for Infectious Disease Prevention and Control, National Institute for Communicable Disease Control and Prevention, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Chinese Center for Disease Control and Prevention, Changping, China.,Research Units of Discovery of Unknown Bacteria and Function (2018 RU010), Chinese Academy of Medical Sciences, Beijing, China
| | - Jianguo Xu
- State Key Laboratory for Infectious Disease Prevention and Control, National Institute for Communicable Disease Control and Prevention, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Chinese Center for Disease Control and Prevention, Changping, China.,Research Units of Discovery of Unknown Bacteria and Function (2018 RU010), Chinese Academy of Medical Sciences, Beijing, China
| | - Zhihong Ren
- State Key Laboratory for Infectious Disease Prevention and Control, National Institute for Communicable Disease Control and Prevention, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Chinese Center for Disease Control and Prevention, Changping, China.,Research Units of Discovery of Unknown Bacteria and Function (2018 RU010), Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
58
|
Garmaeva S, Sinha T, Kurilshikov A, Fu J, Wijmenga C, Zhernakova A. Studying the gut virome in the metagenomic era: challenges and perspectives. BMC Biol 2019; 17:84. [PMID: 31660953 PMCID: PMC6819614 DOI: 10.1186/s12915-019-0704-y] [Citation(s) in RCA: 91] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Accepted: 09/22/2019] [Indexed: 12/12/2022] Open
Abstract
The human gut harbors a complex ecosystem of microorganisms, including bacteria and viruses. With the rise of next-generation sequencing technologies, we have seen a quantum leap in the study of human-gut-inhabiting bacteria, yet the viruses that infect these bacteria, known as bacteriophages, remain underexplored. In this review, we focus on what is known about the role of bacteriophages in human health and the technical challenges involved in studying the gut virome, of which they are a major component. Lastly, we discuss what can be learned from studies of bacteriophages in other ecosystems.
Collapse
Affiliation(s)
- Sanzhima Garmaeva
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Trishla Sinha
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Alexander Kurilshikov
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Jingyuan Fu
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands.,Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Cisca Wijmenga
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Alexandra Zhernakova
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands.
| |
Collapse
|
59
|
Lin TC, Hung YP, Ko WC, Ruan JW. Fecal microbiota transplantation for Clostridium difficile infection in Taiwan: Establishment and implementation. JOURNAL OF MICROBIOLOGY, IMMUNOLOGY, AND INFECTION = WEI MIAN YU GAN RAN ZA ZHI 2019; 52:841-850. [PMID: 31607571 DOI: 10.1016/j.jmii.2019.08.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Revised: 08/22/2019] [Accepted: 08/25/2019] [Indexed: 12/17/2022]
Abstract
Clostridium difficile infection (CDI) remains a major public health issue, and fecal microbiota transplantation (FMT) has become one of the standard therapies for recurrent or refractory CDI. When compared to medical therapies, such as metronidazole or vancomycin, FMT has a high rate of treatment response with acceptable safety and efficiency. Following promulgation of the amendments in September 2018 in Taiwan, FMT has been indicated for recurrent or refractory CDI. The Taiwan Microbiota Consortium contributed to the Taiwan FMT Expert Consensus, which established basic norms and stipulated essential principles, including the indications for transplantation, eligible locations and personnel, donor screening policies, fecal sample handling, and post-FMT follow-up. However, establishing an eligible FMT team in a qualified hospital remains a clinical challenge, and the requirement for facilities and well-screened donors impedes the implementation of FMT. In this review, we aim to provide domestic FMT teams with explicit instructions to facilitate realization and increase the practice of FMT. Based on the Taiwan FMT Expert Consensus and current regulations, we performed a literature review and integrated the experiences of Taiwanese multidisciplinary experts into this article. The content intends to offer clinicians up-to-date evidence and highlight the essential points of FMT.
Collapse
Affiliation(s)
- Tien-Ching Lin
- Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan; Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yuan-Pin Hung
- Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan; Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan; Department of Internal Medicine, Tainan Hospital, Ministry of Health and Welfare, Executive Yuan, Tainan, Taiwan
| | - Wen-Chien Ko
- Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan; Department of Medicine, National Cheng Kung University, Tainan, Taiwan.
| | - Jhen-Wei Ruan
- Department of Medical Laboratory Science and Biotechnology, National Cheng Kung University, Tainan, Taiwan.
| |
Collapse
|
60
|
Revisiting Bacterial Interference in the Age of Methicillin-resistant Staphylococcus aureus: Insights Into Staphylococcus aureus Carriage, Pathogenicity and Potential Control. Pediatr Infect Dis J 2019; 38:958-966. [PMID: 31274832 PMCID: PMC6692185 DOI: 10.1097/inf.0000000000002411] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Bacteria compete with each other for local supremacy in biologic and environmental niches. In humans, who host an array of commensal bacteria, the presence of one species or strain can sometimes prevent colonization by another, a phenomenon known as "bacterial interference." We describe how, in the 1960s, infants (and later adults) were actively inoculated with a relatively benign strain of Staphylococcus aureus, 502A, to prevent colonization with an epidemic S. aureus strain, 80/81. This introduced bacterial interference as a clinical approach to disease prevention, but little was known about the mechanisms of interference at that time. Since then, much has been learned about how bacteria interact with each other and the host to establish carriage, compete for niches and shift from harmless commensal to invasive pathogen. We provide an overview of these findings and summarize recent studies in which the genome and function of 502A were compared with those of the current epidemic strain, USA300, providing insight into differences in their invasiveness and immunogenicity. Although staphylococcal vaccines have been developed, none has yet been approved for clinical use. Further studies of staphylococcal strains and the molecular characteristics that lead to exclusion of specific bacteria from some niches may provide an alternative path to disease prevention.
Collapse
|
61
|
Transplantation of fecal microbiota rich in short chain fatty acids and butyric acid treat cerebral ischemic stroke by regulating gut microbiota. Pharmacol Res 2019; 148:104403. [PMID: 31425750 DOI: 10.1016/j.phrs.2019.104403] [Citation(s) in RCA: 237] [Impact Index Per Article: 39.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 08/07/2019] [Accepted: 08/15/2019] [Indexed: 01/07/2023]
Abstract
The gut microbiota and its short chain fatty acid (SCFA) metabolites have been established to play an important protective role against neurodegenerative diseases. Our previous study demonstrated that cerebral ischemic stroke triggers dysfunctional gut microbiota and increased intestinal permeability. In this study, we aimed to clarify the mechanism by which gut microbiota and SCFAs can treat cerebral ischemic stroke in rat middle cerebral artery occlusion models and use the information to develop new therapies. Our results show that oral administration of non-absorbable antibiotics reduced neurological impairment and the cerebral infarct volume, relieved cerebral edemas, and decreased blood lipid levels by altering the gut microbiota. We also found that ischemic stroke decreased intestinal levels of SCFAs. And that transplanting fecal microbiota rich in these metabolites was an effective means of treating the condition. Compared with other SCFAs, butyric acid showed the highest negative correlation with ischemic stroke. Supplementation with butyric acid treated models of ischemic stroke effectively by remodeling the gut microbiota, enriching the beneficial Lactobacillus, and repairing the leaky gut. In conclusion, interfering with the gut microbiota by transplanting fecal bacteria rich in SCFAs and supplementing with butyric acid were found to be effective treatments for cerebral ischemic stroke.
Collapse
|
62
|
Canibe N, O’Dea M, Abraham S. Potential relevance of pig gut content transplantation for production and research. J Anim Sci Biotechnol 2019; 10:55. [PMID: 31304012 PMCID: PMC6604143 DOI: 10.1186/s40104-019-0363-4] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Accepted: 05/10/2019] [Indexed: 02/06/2023] Open
Abstract
It is becoming increasingly evident that the gastrointestinal microbiota has a significant impact on the overall health and production of the pig. This has led to intensified research on the composition of the gastrointestinal microbiota, factors affecting it, and the impact of the microbiota on health, growth performance, and more recently, behavior of the host. Swine production research has been heavily focused on assessing the effects of feed additives and dietary modifications to alter or take advantage of select characteristics of gastrointestinal microbes to improve health and feed conversion efficiency. Research on faecal microbiota transplantation (FMT) as a possible tool to improve outcomes in pigs through manipulation of the gastrointestinal microbiome is very recent and limited data is available. Results on FMT in humans demonstrating the transfer of phenotypic traits from donors to recipients and the high efficacy of FMT to treat Clostridium difficile infections in humans, together with data from pigs relating GI-tract microbiota composition with growth performance has likely played an important role in the interest towards this strategy in pig production. However, several factors can influence the impact of FMT on the recipient, and these need to be identified and optimized before this tool can be applied to pig production. There are obvious inherent biosecurity and regulatory issues in this strategy, since the donor's microbiome can never be completely screened for all possible non-desirable microorganisms. However, considering the success observed in humans, it seems worth investigating this strategy for certain applications in pig production. Further, FMT research may lead to the identification of specific bacterial group(s) essential for a particular outcome, resulting in the development of banks of clones which can be used as targeted therapeutics, rather than the broader approach applied in FMT. This review examines the factors associated with the use of FMT, and its potential application to swine production, and includes research on using the pig as model for human medical purposes.
Collapse
Affiliation(s)
- Nuria Canibe
- Department of Animal Science, Aarhus University, AU-FOULUM, PO BOX 50, 8830 Tjele, Denmark
| | - Mark O’Dea
- Antimicrobial Resistance and Infectious Disease laboratory, College of Science, Health, Engineering and Education, Murdoch University, Western Australia, Australia
| | - Sam Abraham
- Antimicrobial Resistance and Infectious Disease laboratory, College of Science, Health, Engineering and Education, Murdoch University, Western Australia, Australia
| |
Collapse
|
63
|
Jayakumar S, Loomba R. Review article: emerging role of the gut microbiome in the progression of nonalcoholic fatty liver disease and potential therapeutic implications. Aliment Pharmacol Ther 2019; 50:144-158. [PMID: 31149745 PMCID: PMC6771496 DOI: 10.1111/apt.15314] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2018] [Revised: 10/24/2018] [Accepted: 04/30/2019] [Indexed: 02/06/2023]
Abstract
BACKGROUND Nonalcoholic fatty liver disease (NAFLD) is a prevalent disorder associated with obesity and diabetes. Few treatment options are effective for patients with NAFLD, but connections between the gut microbiome and NAFLD and NAFLD-associated conditions suggest that modulation of the gut microbiota could be a novel therapeutic option. AIM To examine the effect of the gut microbiota on pathophysiologic causes of NAFLD and assess the potential of microbiota-targeting therapies for NAFLD. METHODS A PubMed search of the literature was performed; relevant articles were included. RESULTS The composition of bacteria in the gastrointestinal tract can enhance fat deposition, modulate energy metabolism and alter inflammatory processes. Emerging evidence suggests a role for the gut microbiome in obesity and metabolic syndrome. NAFLD is often considered the hepatic manifestation of metabolic syndrome, and there has been tremendous progress in understanding the association of gut microbiome composition with NAFLD disease severity. We discuss the role of the gut microbiome in NAFLD pathophysiology and whether the microbiome composition can differentiate the two categories of NAFLD: nonalcoholic fatty liver (NAFL, the non-progressive form) vs nonalcoholic steatohepatitis (NASH, the progressive form). The association between gut microbiome and fibrosis progression in NAFLD is also discussed. Finally, we review whether modulation of the gut microbiome plays a role in improving treatment outcomes for patients with NAFLD. CONCLUSIONS Multiple pathophysiologic pathways connect the gut microbiome with the pathophysiology of NAFLD. Therefore, therapeutics that effectively target the gut microbiome may be beneficial for the treatment of patients with NAFLD.
Collapse
Affiliation(s)
- Saumya Jayakumar
- Division of Gastroenterology and Hepatology, Department of MedicineNAFLD Research Center, University of California at San DiegoLa JollaCalifornia
| | - Rohit Loomba
- Division of Gastroenterology and Hepatology, Department of MedicineNAFLD Research Center, University of California at San DiegoLa JollaCalifornia,Division of Epidemiology, Department of Family Medicine and Public HealthUniversity of California at San DiegoLa JollaCalifornia
| |
Collapse
|
64
|
Dutta SK, Verma S, Jain V, Surapaneni BK, Vinayek R, Phillips L, Nair PP. Parkinson's Disease: The Emerging Role of Gut Dysbiosis, Antibiotics, Probiotics, and Fecal Microbiota Transplantation. J Neurogastroenterol Motil 2019; 25:363-376. [PMID: 31327219 PMCID: PMC6657920 DOI: 10.5056/jnm19044] [Citation(s) in RCA: 93] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 05/17/2019] [Accepted: 06/24/2019] [Indexed: 12/19/2022] Open
Abstract
The role of the microbiome in health and human disease has emerged at the forefront of medicine in the 21st century. Over the last 2 decades evidence has emerged to suggest that inflammation-derived oxidative damage and cytokine induced toxicity may play a significant role in the neuronal damage associated with Parkinson’s disease (PD). Presence of pro-inflammatory cytokines and T cell infiltration has been observed in the brain parenchyma of patients with PD. Furthermore, evidence for inflammatory changes has been reported in the enteric nervous system, the vagus nerve branches and glial cells. The presence of α-synuclein deposits in the post-mortem brain biopsy in patients with PD has further substantiated the role of inflammation in PD. It has been suggested that the α-synuclein misfolding might begin in the gut and spread “prion like” via the vagus nerve into lower brainstem and ultimately to the midbrain; this is known as the Braak hypothesis. It is noteworthy that the presence of gastrointestinal symptoms (constipation, dysphagia, and hypersalivation), altered gut microbiota and leaky gut have been observed in PD patients several years prior to the clinical onset of the disease. These clinical observations have been supported by in vitro studies in mice as well, demonstrating the role of genetic (α-synuclein overexpression) and environmental (gut dysbiosis) factors in the pathogenesis of PD. The restoration of the gut microbiome in patients with PD may alter the clinical progression of PD and this alteration can be accomplished by carefully designed studies using customized probiotics and fecal microbiota transplantation.
Collapse
Affiliation(s)
- Sudhir K Dutta
- Sinai Hospital, Baltimore, MD, USA.,University of Maryland School of Medicine, Baltimore, MD, USA
| | | | | | | | | | | | - Padmanabhan P Nair
- Sinai Hospital, Baltimore, MD, USA.,Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA.,NonInvasive Technologies LLC, Elkridge, MD, USA
| |
Collapse
|
65
|
Koontz JM, Dancy BCR, Horton CL, Stallings JD, DiVito VT, Lewis JA. The Role of the Human Microbiome in Chemical Toxicity. Int J Toxicol 2019; 38:251-264. [PMID: 31220972 DOI: 10.1177/1091581819849833] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
There is overwhelming evidence that the microbiome must be considered when evaluating the toxicity of chemicals. Disruption of the normal microbial flora is a known effect of toxic exposure, and these disruptions may lead to human health effects. In addition, the biotransformation of numerous compounds has been shown to be dependent on microbial enzymes, with the potential for different host health outcomes resulting from variations in the microbiome. Evidence suggests that such metabolism of environmental chemicals by enzymes from the host's microbiota can affect the toxicity of that chemical to the host. Chemical-microbial interactions can be categorized into two classes: Microbiome Modulation of Toxicity (MMT) and Toxicant Modulation of the Microbiome (TMM). MMT refers to transformation of a chemical by microbial enzymes or metabolites to modify the chemical in a way that makes it more or less toxic. TMM is a change in the microbiota that results from a chemical exposure. These changes span a large magnitude of effects and may vary from microbial gene regulation, to inhibition of a specific enzyme, to the death of the microbes. Certain microbiomes or microbiota may become associated with different health outcomes, such as resistance or susceptibility to exposure to certain toxic chemicals, the ability to recover following a chemical-induced injury, the presence of disease-associated phenotypes, and the effectiveness of immune responses. Future work in toxicology will require an understanding of how the microbiome interacts with toxicants to fully elucidate how a compound will affect a diverse, real-world population.
Collapse
Affiliation(s)
- Jason M Koontz
- 1 US Army Center for Environmental Health Research, Fort Detrick, MD, USA
| | - Blair C R Dancy
- 1 US Army Center for Environmental Health Research, Fort Detrick, MD, USA
| | | | | | - Valerie T DiVito
- 1 US Army Center for Environmental Health Research, Fort Detrick, MD, USA
| | - John A Lewis
- 1 US Army Center for Environmental Health Research, Fort Detrick, MD, USA
| |
Collapse
|
66
|
Vigvári S, Vincze Á, Solt J, Sipos D, Feiszt Z, Kovács B, Kappéter Á, Péterfi Z. Experiences with fecal microbiota transplantation in Clostridium difficile infections via upper gastrointestinal tract. Acta Microbiol Immunol Hung 2019; 66:179-188. [PMID: 30585500 DOI: 10.1556/030.65.2018.051] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Dramatic changes in the epidemiology of Clostridium difficile infections have been reported from the western world in the past decade. The proportion of severe cases is significantly elevating and clinicians now have to contend with the problem of additional and more frequent episodes of recurrences including an upward trend in the mortality rate. This situation led us to investigate the possibility of the fecal microbiota transplantation (FMT). An amount of 100 ml of fecal microbiota solution was instilled into a nasojejunal (NJ) tube in 16 cases and into a nasogastric (NG) tube in 44 cases. In all of the cases, where the solution was instilled via nasojejunal tubes, the symptoms resolved within 24 h. We did not note any recurrences in this group. When the material was flushed in through nasogastric tubes, the symptoms resolved in 39 (88.64%) cases within 24 h. In this group, we have experienced a recurrent episode of C. difficile infection in five (11.36%) cases. Three of them were cured with a second transplantation. We have found that in our practice the upper gastrointestinal tract methods had the primary cure rate of 91.67%, whereas the secondary cure rate is 96.67%. When we compared the NJ and NG methods, we have found that the differences in the outcomes are not significant statistically (p = 0.3113 using Fisher's exact probability test). In conclusion, FMT proved to be very effective, particularly in recurrent infections and in cases where conventional treatment had failed.
Collapse
Affiliation(s)
- Szabolcs Vigvári
- 1 Department of Infectology, 1st Department of Internal Medicine, University of Pécs, Pécs, Hungary
| | - Áron Vincze
- 2 Department of Gastroenterology, 1st Department of Internal Medicine, University of Pécs, Pécs, Hungary
| | - Jenő Solt
- 2 Department of Gastroenterology, 1st Department of Internal Medicine, University of Pécs, Pécs, Hungary
| | - Dávid Sipos
- 1 Department of Infectology, 1st Department of Internal Medicine, University of Pécs, Pécs, Hungary
| | - Zsófia Feiszt
- 1 Department of Infectology, 1st Department of Internal Medicine, University of Pécs, Pécs, Hungary
| | - Beáta Kovács
- 1 Department of Infectology, 1st Department of Internal Medicine, University of Pécs, Pécs, Hungary
| | - Ágnes Kappéter
- 1 Department of Infectology, 1st Department of Internal Medicine, University of Pécs, Pécs, Hungary
| | - Zoltán Péterfi
- 1 Department of Infectology, 1st Department of Internal Medicine, University of Pécs, Pécs, Hungary
| |
Collapse
|
67
|
Fessler J, Matson V, Gajewski TF. Exploring the emerging role of the microbiome in cancer immunotherapy. J Immunother Cancer 2019; 7:108. [PMID: 30995949 PMCID: PMC6471869 DOI: 10.1186/s40425-019-0574-4] [Citation(s) in RCA: 197] [Impact Index Per Article: 32.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2018] [Accepted: 03/22/2019] [Indexed: 12/24/2022] Open
Abstract
The activity of the commensal microbiota significantly impacts human health and has been linked to the development of many diseases, including cancer. Gnotobiotic animal models have shown that the microbiota has many effects on host physiology, including on the development and regulation of immune responses. More recently, evidence has indicated that the microbiota can more specifically influence the outcome of cancer immunotherapy. Therapeutic interventions to optimize microbiota composition to improve immunotherapy outcomes have shown promise in mouse studies. Ongoing endeavors are translating these pre-clinical findings to early stage clinical testing. In this review we summarize 1) basic methodologies and considerations for studies of host-microbiota interactions; 2) experimental evidence towards a causal link between gut microbiota composition and immunotherapeutic efficacy; 3) possible mechanisms governing the microbiota-mediated impact on immunotherapy efficacy. Moving forward, there is need for a deeper understanding of the underlying biological mechanisms that link specific bacterial strains to host immunity. Integrating microbiome effects with other tumor and host factors regulating immunotherapy responsiveness versus resistance could facilitate optimization of therapeutic outcomes.
Collapse
Affiliation(s)
- Jessica Fessler
- Department of Pathology, The University of Chicago, Chicago, IL, USA
| | - Vyara Matson
- Department of Pathology, The University of Chicago, Chicago, IL, USA
| | - Thomas F Gajewski
- Department of Pathology, The University of Chicago, Chicago, IL, USA.
- Department of Medicine, Section of Hematology/Oncology, The University of Chicago, 5841 S. Maryland Ave., MC2115, Chicago, IL, 60637, USA.
| |
Collapse
|
68
|
Dehlholm-Lambertsen E, Hall BK, Jørgensen SMD, Jørgensen CW, Jensen ME, Larsen S, Jensen JS, Ehlers L, Dahlerup JF, Hvas CL. Cost savings following faecal microbiota transplantation for recurrent Clostridium difficile infection. Therap Adv Gastroenterol 2019; 12:1756284819843002. [PMID: 31007720 PMCID: PMC6460887 DOI: 10.1177/1756284819843002] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Accepted: 02/26/2019] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND Recurrent Clostridium difficile infection (rCDI) is becoming increasingly common. Faecal microbiota transplantation (FMT) is effective for rCDI, but the costs of an FMT and hospital cost savings related to FMT are unknown. The aim of this study was to calculate the cost of an FMT and the total hospital costs before and after FMT. METHODS This was an observational single-centre study, carried out in a public teaching hospital. We included all patients referred for rCDI from January 2014 through December 2015 and documented costs related to donor screening, laboratory processing, and clinical FMT application. We calculated patient-related hospital costs 1 year before FMT (pre-FMT) and 1 year after FMT (post-FMT). Sensitivity analyses were applied to assess the robustness of the results. RESULTS We included 50 consecutive adult patients who had a verified diagnosis of rCDI and were referred for FMT. The average cost of an outpatient FMT procedure if donor faeces were applied by colonoscopy was €3,326 per patient and €2,864 if donor faeces were applied using a nasojejunal tube. The total annual pre-FMT hospital costs per patient were €56,415 (95% confidence interval (CI) 41,133-71,697), and these costs dropped by 42% to €32,816 (22,618-42,014) post-FMT (p = 0.004). The main cost driver was hospital admissions. Sensitivity analyses demonstrated cost reductions in all scenarios. CONCLUSIONS In a public hospital with an implemented FMT service, the average cost of FMT applied by either colonoscopy or nasojejunal tube was €3,095. Total hospital costs dropped by 42% the first year after FMT. The reduction was mainly caused by reductions in the number of hospital admissions and in length of stay.
Collapse
Affiliation(s)
| | | | - Simon M. D. Jørgensen
- Department of Hepatology and Gastroenterology, Aarhus University Hospital, Aarhus, Denmark
| | | | - Mia E. Jensen
- School of Medicine and Health (SMH), Aalborg University, Denmark
| | - Sara Larsen
- School of Medicine and Health (SMH), Aalborg University, Denmark
| | | | - Lars Ehlers
- Department of Business and Management, Danish Center for Healthcare Improvements, Aalborg University, Aalborg, Denmark
| | - Jens F. Dahlerup
- Department of Hepatology and Gastroenterology, Aarhus University Hospital, Aarhus, Denmark
| | | |
Collapse
|
69
|
Zhou X, Devescovi V, Liu Y, Dent JE, Nardini C. Host-Microbiome Synergistic Control on Sphingolipid Metabolism by Mechanotransduction in Model Arthritis. Biomolecules 2019; 9:biom9040144. [PMID: 30970641 PMCID: PMC6523851 DOI: 10.3390/biom9040144] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Revised: 04/01/2019] [Accepted: 04/03/2019] [Indexed: 12/27/2022] Open
Abstract
Chronic inflammatory autoimmune disorders are systemic diseases with increasing incidence and still lack a cure. More recently, attention has been placed in understanding gastrointestinal (GI) dysbiosis and, although important progress has been made in this area, it is currently unclear to what extent microbiome manipulation can be used in the treatment of autoimmune disorders. Via the use of appropriate models, rheumatoid arthritis (RA), a well-known exemplar of such pathologies, can be exploited to shed light on the currently overlooked effects of existing therapies on the GI microbiome. In this direction, we here explore the crosstalk between the GI microbiome and the host immunity in model arthritis (collagen induced arthritis, CIA). By exploiting omics from samples of limited invasiveness (blood and stools), we assess the host-microbiome responses to standard therapy (methotrexate, MTX) combined with mechanical subcutaneous stimulation (MS) and to mechanical stimulation alone. When MS is involved, results reveal the sphingolipid metabolism as the trait d’union among known hallmarks of (model) RA, namely: Imbalance in the S1P-S1PR1 axis, expansion of Prevotella sp., and invariant Natural Killer T (iNKT)-penia, thus offering the base of a rationale to mechanically modulate this pathway as a therapeutic target in RA.
Collapse
Affiliation(s)
- Xiaoyuan Zhou
- Department of Neurology, University of California, San Francisco, CA 94158, USA.
- CAS-MPG Partner Institute for Computational Biology, Shanghai Institutes for Biological Sciences, Shanghai 200031, China.
| | - Valentina Devescovi
- CAS-MPG Partner Institute for Computational Biology, Shanghai Institutes for Biological Sciences, Shanghai 200031, China.
| | - Yuanhua Liu
- CAS-MPG Partner Institute for Computational Biology, Shanghai Institutes for Biological Sciences, Shanghai 200031, China.
- Bioinformatics Platform, Institut Pasteur of Shanghai, CAS, Shanghai 200031, China.
| | - Jennifer E Dent
- NORSAS Consultancy Ltd., Hoveton, Norwich, Norfolk, NR128QP, UK.
| | - Christine Nardini
- CAS-MPG Partner Institute for Computational Biology, Shanghai Institutes for Biological Sciences, Shanghai 200031, China.
- Department of Laboratory Medicine, Division of Clinical Chemistry Karolinska Institute, 17177 Stockholm, Sweden.
- Scientific and Medical Direction, SOL Group S.r.l, 20900 Monza, Italy.
- CNR IAC "Mauro Picone", 00185 Roma, Italy.
| |
Collapse
|
70
|
Nobs SP, Tuganbaev T, Elinav E. Microbiome diurnal rhythmicity and its impact on host physiology and disease risk. EMBO Rep 2019; 20:embr.201847129. [PMID: 30877136 DOI: 10.15252/embr.201847129] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Revised: 11/29/2018] [Accepted: 02/22/2019] [Indexed: 12/29/2022] Open
Abstract
Host-microbiome interactions constitute key determinants of host physiology, while their dysregulation is implicated in a wide range of human diseases. The microbiome undergoes diurnal variation in composition and function, and this in turn drives oscillations in host gene expression and functions. In this review, we discuss the newest developments in understanding circadian host-microbiome interplays, and how they may be relevant in health and disease contexts. We summarize the molecular mechanisms by which the microbiome influences host function in a diurnal manner, and inversely describe how the host orchestrates circadian rhythmicity of the microbiome. Furthermore, we highlight the future perspectives and challenges in studying this new and exciting facet of host-microbiome interactions. Finally, we illustrate how the elucidation of the microbiome chronobiology may pave the way for novel therapeutic approaches.
Collapse
Affiliation(s)
| | - Timur Tuganbaev
- Immunology Department, Weizmann Institute of Science, Rehovot, Israel
| | - Eran Elinav
- Immunology Department, Weizmann Institute of Science, Rehovot, Israel .,Cancer-Microbiome Division, Deutsches Krebsforschungszentrum (DKFZ), Heidelberg, Germany
| |
Collapse
|
71
|
Vigvári S, Sipos D, Solt J, Vincze Á, Kocsis B, Nemes Z, Kappéter Á, Feiszt Z, Kovács B, Péterfi Z. Faecal microbiota transplantation for Clostridium difficile infection using a lyophilized inoculum from non-related donors: A case series involving 19 patients. Acta Microbiol Immunol Hung 2019; 66:69-78. [PMID: 29239198 DOI: 10.1556/030.64.2017.042] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Faecal microbiota transplantation (FMT) has been reported to be effective in treating relapsing of refractory Clostridium difficile infections, although some practical barriers are limiting its widespread use. In this study, our objective was to evaluate the rate of resolution of diarrhea following administration of lyophilized and resolved FMT via a nasogastric (NG) tube. We recruited 19 patients suffered from laboratory-confirmed C. difficile infection. Each of them was treated by lyophilized and resolved inoculum through a NG tube. One participant succumbed following the procedure due to unrelated diseases. Out of 18 cases, 15 patients reportedly experienced a resolution of the symptoms. One patient was treated with another course of antibiotics, and two of the non-responders were successfully retreated with another course of FMT utilizing a lyophilized inoculum. Notably, no significant adverse activities were observed. In accordance to our clinical experiences, a patient will likely benefit from FMT treatment including lyophilized inoculum.
Collapse
Affiliation(s)
- Szabolcs Vigvári
- 1 Department of Infectious Diseases, 1st Department of Internal Medicine, University of Pécs, Pécs, Hungary
| | - Dávid Sipos
- 1 Department of Infectious Diseases, 1st Department of Internal Medicine, University of Pécs, Pécs, Hungary
| | - Jenő Solt
- 2 Department of Gastroenterology, 1st Department of Internal Medicine, University of Pécs, Pécs, Hungary
| | - Áron Vincze
- 2 Department of Gastroenterology, 1st Department of Internal Medicine, University of Pécs, Pécs, Hungary
| | - Béla Kocsis
- 3 Department of Medical Microbiology and Immunology, University of Pécs, Pécs, Hungary
| | - Zsuzsanna Nemes
- 1 Department of Infectious Diseases, 1st Department of Internal Medicine, University of Pécs, Pécs, Hungary
| | - Ágnes Kappéter
- 1 Department of Infectious Diseases, 1st Department of Internal Medicine, University of Pécs, Pécs, Hungary
| | - Zsófia Feiszt
- 1 Department of Infectious Diseases, 1st Department of Internal Medicine, University of Pécs, Pécs, Hungary
| | - Beáta Kovács
- 1 Department of Infectious Diseases, 1st Department of Internal Medicine, University of Pécs, Pécs, Hungary
| | - Zoltán Péterfi
- 1 Department of Infectious Diseases, 1st Department of Internal Medicine, University of Pécs, Pécs, Hungary
| |
Collapse
|
72
|
Li Y, Zou Z, Bian X, Huang Y, Wang Y, Yang C, Zhao J, Xie L. Fecal microbiota transplantation research output from 2004 to 2017: a bibliometric analysis. PeerJ 2019; 7:e6411. [PMID: 30809438 PMCID: PMC6387576 DOI: 10.7717/peerj.6411] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Accepted: 01/01/2019] [Indexed: 12/15/2022] Open
Abstract
Background Fecal microbiota transplantation (FMT) is an emerging therapy against Clostridium difficile infection (CDI) and inflammatory bowel disease (IBD). Although the therapy has gained prominence, there has been no bibliometric analysis of FMT. Methods Studies published from 2004 to 2017 were extracted from the Science Citation Index Expanded. Bibliometric analysis was used to evaluate the number or cooperation network of publications, countries, citations, references, journals, authors, institutions and keywords. Results A total of 796 items were included, showing an increasing trend annually. Publications mainly came from 10 countries, led by the US (n = 363). In the top 100 articles ranked by the number of citations (range 47–1,158), American Journal of Gastroenterology (2017 IF = 10.231) took the top spot. The co-citation network had 7 co-citation clusters headed by ‘recurrent Clostridium difficile infection’. The top 7 keywords with the strongest citation bursts had three parts, ‘microbiota’, ‘ diarrhea ’, and ‘case series’. All keywords were divided into four domains, ‘disease’, ‘nosogenesis’, ‘trial’, and ‘therapy’. Conclusions This study shows the research performance of FMT from 2004 to 2017 and helps investigators master the trend of FMT, which is also an ongoing hotspot of research.
Collapse
Affiliation(s)
- Yan Li
- Department of General Surgery, Zhujiang Hospital of Southern Medical University, Guangzhou, Guangdong, China.,The Second School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Ziyuan Zou
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Xiaohui Bian
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Yushan Huang
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Yanru Wang
- Department of Radiation Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangzhou, China
| | - Chen Yang
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Jian Zhao
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Lang Xie
- Department of General Surgery, Zhujiang Hospital of Southern Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
73
|
Tsai YL, Lin TL, Chang CJ, Wu TR, Lai WF, Lu CC, Lai HC. Probiotics, prebiotics and amelioration of diseases. J Biomed Sci 2019; 26:3. [PMID: 30609922 PMCID: PMC6320572 DOI: 10.1186/s12929-018-0493-6] [Citation(s) in RCA: 215] [Impact Index Per Article: 35.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Accepted: 12/13/2018] [Indexed: 02/06/2023] Open
Abstract
Dysbiosis of gut microbiota is closely related to occurrence of many important chronic inflammations-related diseases. So far the traditionally prescribed prebiotics and probiotics do not show significant impact on amelioration of these diseases in general. Thus the development of next generation prebiotics and probiotics designed to target specific diseases is urgently needed. In this review, we first make a brief introduction on current understandings of normal gut microbiota, microbiome, and their roles in homeostasis of mucosal immunity and gut integrity. Then, under the situation of microbiota dysbiosis, development of chronic inflammations in the intestine occurs, leading to leaky gut situation and systematic chronic inflammation in the host. These subsequently resulted in development of many important diseases such as obesity, type 2 diabetes mellitus, liver inflammations, and other diseases such as colorectal cancer (CRC), obesity-induced chronic kidney disease (CKD), the compromised lung immunity, and some on brain/neuro disorders. The strategy used to optimally implant the effective prebiotics, probiotics and the derived postbiotics for amelioration of the diseases is presented. While the effectiveness of these agents seems promising, additional studies are needed to establish recommendations for most clinical settings.
Collapse
Affiliation(s)
- Yu-Ling Tsai
- Microbiota Research Center and Emerging Viral Infections Research Center, Chang Gung University, Gueishan, Taoyuan, 33302, Taiwan
| | - Tzu-Lung Lin
- Microbiota Research Center and Emerging Viral Infections Research Center, Chang Gung University, Gueishan, Taoyuan, 33302, Taiwan
| | - Chih-Jung Chang
- Microbiota Research Center and Emerging Viral Infections Research Center, Chang Gung University, Gueishan, Taoyuan, 33302, Taiwan.,Chang Gung Immunology Consortium, Linkou Chang Gung Memorial Hospital, Gueishan, Taoyuan, 33305, Taiwan
| | - Tsung-Ru Wu
- Institute of Biomedical Sciences, Academia Sinica, Taipei, 115, Taiwan
| | - Wei-Fan Lai
- Department of Medicine, College of Medicine, Chang Gung University, Gueishan, Taoyuan, 33302, Taiwan
| | - Chia-Chen Lu
- Department of Respiratory Therapy, Fu Jen Catholic University, Xinzhuang, New Taipei City, 24205, Taiwan.
| | - Hsin-Chih Lai
- Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Gueishan, Taoyuan, 33302, Taiwan. .,Microbiota Research Center and Emerging Viral Infections Research Center, Chang Gung University, Gueishan, Taoyuan, 33302, Taiwan. .,Chang Gung Immunology Consortium, Linkou Chang Gung Memorial Hospital, Gueishan, Taoyuan, 33305, Taiwan. .,Department of Laboratory Medicine, Linkou Chang Gung Memorial Hospital, Gueishan, Taoyuan, 33305, Taiwan. .,Research Center for Chinese Herbal Medicine, College of Human Ecology, Chang Gung University of Science and Technology, Gueishan, Taoyuan, 33303, Taiwan. .,Research Center for Food and Cosmetic Safety, College of Human Ecology, Chang Gung University of Science and Technology, Gueishan, Taoyuan, 33303, Taiwan.
| |
Collapse
|
74
|
Kim P, Gadani A, Abdul-Baki H, Mitre R, Mitre M. Fecal microbiota transplantation in recurrent Clostridium difficile infection: A retrospective single-center chart review. JGH OPEN 2018; 3:4-9. [PMID: 30834334 PMCID: PMC6386733 DOI: 10.1002/jgh3.12093] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Revised: 08/25/2018] [Accepted: 08/30/2018] [Indexed: 12/13/2022]
Abstract
Background and Aim Fecal microbiota transplantation (FMT) has been proposed as a treatment option for patients with recurrent Clostridium difficile (C. difficile) infection but remains a novel option. We examined if FMT is an effective means of treating recurrent C. difficile infection. Methods A retrospective review of 35 patients who underwent FMT was completed. Demographics and other variables, including the use of proton pump inhibitor therapy and history of inflammatory bowel disease, were collected. Results Twenty-five patients (71.4%) belonged to a high-risk population (working in a hospital setting, rehabilitation center, or nursing facility) and a total of 74.3% of patients (n = 26 patients) had no history of proton pump inhibitor use. Twenty-five patients (71.4%) had used metronidazole prior to transplantation, 35 patients (100%) had used vancomycin, and 7 patients (20%) had used fidaxomicin for prior infection. Four patients (11.4%) had used all three antibiotics during prior treatment. Of the eight patients who had a history of inflammatory bowel disease, six (75%) experienced resolution of symptoms after transplantation. A total of 30 patients (85.7%) had resolution of their symptoms 6-8 weeks' posttransplant, while 5 patients (14.3%) continued to have symptoms. Conclusions Our retrospective chart review supports that patients benefit from FMT in the setting of recurrent C. difficile infection.
Collapse
Affiliation(s)
- Pamela Kim
- Department of Internal Medicine and Division of Gastroenterology and Hepatology Allegheny General Hospital Pittsburgh Pennsylvania United States of America
| | - Akash Gadani
- Department of Internal Medicine and Division of Gastroenterology and Hepatology Allegheny General Hospital Pittsburgh Pennsylvania United States of America
| | - Heitham Abdul-Baki
- Department of Internal Medicine and Division of Gastroenterology and Hepatology Allegheny General Hospital Pittsburgh Pennsylvania United States of America
| | - Ricardo Mitre
- Department of Internal Medicine and Division of Gastroenterology and Hepatology Allegheny General Hospital Pittsburgh Pennsylvania United States of America
| | - Marcia Mitre
- Department of Internal Medicine and Division of Gastroenterology and Hepatology Allegheny General Hospital Pittsburgh Pennsylvania United States of America
| |
Collapse
|
75
|
Martinez C, Edwards J, Hassoun A. Commercialized fecal microbiota transplantation provides efficacious treatment of Clostridium difficile infection. Infect Dis (Lond) 2018; 50:864-867. [PMID: 30265170 DOI: 10.1080/23744235.2018.1500709] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Affiliation(s)
- Chad Martinez
- a Alabama College of Osteopathic Medicine , Dothan , United States
| | - Jonathan Edwards
- b Department of Pharmacy , Huntsville Hospital , Huntsville , United States
| | - Ali Hassoun
- c Department of Medicine , University of Alabama School of Medicine , Huntsville , United States
| |
Collapse
|
76
|
Speck PG, Mitchell JG. Faecal microbiota transplantation donor stools need screening for poliovirus. Gut 2018; 67:1559-1560. [PMID: 29030396 DOI: 10.1136/gutjnl-2017-314356] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Revised: 09/18/2017] [Accepted: 10/02/2017] [Indexed: 02/04/2023]
Affiliation(s)
- Peter Gerald Speck
- College of Science and Engineering, Flinders University, Bedford Park, South Australia, Australia
| | - James G Mitchell
- College of Science and Engineering, Flinders University, Bedford Park, South Australia, Australia
| |
Collapse
|
77
|
Enright EF, Griffin BT, Gahan CG, Joyce SA. Microbiome-mediated bile acid modification: Role in intestinal drug absorption and metabolism. Pharmacol Res 2018; 133:170-186. [DOI: 10.1016/j.phrs.2018.04.009] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Revised: 04/07/2018] [Accepted: 04/12/2018] [Indexed: 01/03/2023]
|
78
|
Bhutiani N, Schucht JE, Miller KR, McClave SA. Technical Aspects of Fecal Microbial Transplantation (FMT). Curr Gastroenterol Rep 2018; 20:30. [PMID: 29886561 DOI: 10.1007/s11894-018-0636-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
PURPOSE OF REVIEW Fecal microbial transplantation (FMT) has become established as an effective therapeutic modality in the treatment of antibiotic-refractory recurrent Clostridium difficile colitis. A number of formulations and methods of delivery of FMT are currently available, each with distinct advantages. This review aims to review donor and patient selection for FMT as well as procedural aspects of FMT to help guide clinical practice. RECENT FINDINGS FMT can be obtained in fresh, frozen, lyophilized, and capsule-based formulations for delivery by oral ingestion, nasoenteric tube, colonoscopy, or enema (depending on the formulation used). Choosing the optimal method relies heavily on patient-related factors, including underlying pathology and severity of illness. As potential applications for FMT expand, careful donor screening and patient selection are critical to minimizing risk to patients and physicians. FMT represents an excellent therapeutic option for treatment of recurrent Clostridium difficile colitis and holds promise as a possible treatment modality in a variety of other conditions. The wide array of delivery methods allows for its application in various disease states in both the inpatient and outpatient setting.
Collapse
Affiliation(s)
- N Bhutiani
- Department of Surgery, Division of Trauma and Critical Care, University of Louisville, Louisville, KY, USA
| | - J E Schucht
- Department of Surgery, Division of Trauma and Critical Care, University of Louisville, Louisville, KY, USA
| | - K R Miller
- Department of Surgery, Division of Trauma and Critical Care, University of Louisville, Louisville, KY, USA
| | - Stephen A McClave
- Department of Medicine, Division of Gastroenterology, University of Louisville, 550 S. Jackson St., Ambulatory Care Building 3nd Floor, Louisville, KY, 40202, USA.
| |
Collapse
|
79
|
Chen YE, Fischbach MA, Belkaid Y. Skin microbiota-host interactions. Nature 2018; 553:427-436. [PMID: 29364286 DOI: 10.1038/nature25177] [Citation(s) in RCA: 428] [Impact Index Per Article: 61.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Accepted: 11/28/2017] [Indexed: 12/23/2022]
Abstract
The skin is a complex and dynamic ecosystem that is inhabited by bacteria, archaea, fungi and viruses. These microbes-collectively referred to as the skin microbiota-are fundamental to skin physiology and immunity. Interactions between skin microbes and the host can fall anywhere along the continuum between mutualism and pathogenicity. In this Review, we highlight how host-microbe interactions depend heavily on context, including the state of immune activation, host genetic predisposition, barrier status, microbe localization, and microbe-microbe interactions. We focus on how context shapes the complex dialogue between skin microbes and the host, and the consequences of this dialogue for health and disease.
Collapse
Affiliation(s)
- Y Erin Chen
- Department of Dermatology, University of California San Francisco, San Francisco, California, USA.,Department of Bioengineering and ChEM-H, Stanford University, Stanford, California, USA
| | - Michael A Fischbach
- Department of Bioengineering and ChEM-H, Stanford University, Stanford, California, USA
| | - Yasmine Belkaid
- NIAID Microbiome Program, National Institute of Allergy and Infectious Disease, NIH, Bethesda, Maryland, USA.,Mucosal Immunology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Disease, NIH, Bethesda, Maryland, USA
| |
Collapse
|
80
|
Vargason AM, Anselmo AC. Clinical translation of microbe-based therapies: Current clinical landscape and preclinical outlook. Bioeng Transl Med 2018; 3:124-137. [PMID: 30065967 PMCID: PMC6063871 DOI: 10.1002/btm2.10093] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Revised: 04/23/2018] [Accepted: 04/26/2018] [Indexed: 12/11/2022] Open
Abstract
Next generation microbe-based therapeutics, inspired by the success of fecal microbiota transplants, are being actively investigated in clinical trials to displace or eliminate pathogenic microbes to treat various diseases in the gastrointestinal tract, skin, and vagina. Genetically engineered microbes are also being investigated in the clinic as drug producing factories for biologic delivery, which can provide a constant local source of drugs. In either case, microbe-therapeutics have the opportunity to address unmet clinical needs and open new areas of research by reducing clinical side effects associated with current treatment modalities or by facilitating the delivery of biologics. This review will discuss examples of past and current clinical trials that are investigating microbe-therapeutics, both microbiome-modulating and drug-producing, for the treatment of a range of diseases. We then offer a perspective on how preclinical approaches, both those focused on developing advanced delivery systems and those that use in vitro microbiome model systems to inform formulation design, will lead to the realization of next-generation microbe-therapeutics.
Collapse
Affiliation(s)
- Ava M. Vargason
- Div. of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of PharmacyUniversity of North Carolina at Chapel HillChapel HillNC 27599
| | - Aaron C. Anselmo
- Div. of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of PharmacyUniversity of North Carolina at Chapel HillChapel HillNC 27599
| |
Collapse
|
81
|
Abstract
PURPOSE OF REVIEW Emerging research on the pediatric microbiome implicates the importance of the microbiome on the development of the immune system, nervous system, and growth. Changes to the microbiome during infancy are associated with the development of chronic illnesses such as asthma and inflammatory bowel disease. Additionally, the microbiome provides protection against certain pathogens, affects vaccine responses, and alters drug metabolism. This review highlights what is known about the microbiome, the establishment of a healthy microbiome and the significance that changes to the microbiome composition have on growth and health of children and adolescents. RECENT FINDINGS Vaginal delivery, breastfeeding, maternal health, and nutrition help shape a healthy microbiome. Caesarian delivery, formula feeding, and antibiotic use perturb the microbiome and are associated with the development of type II diabetes, asthma, allergic diseases, and obesity later in life. Specific interventions using pre and probiotics in multiple settings are under investigation with limited success. SUMMARY A better understanding of the microbiome and the interaction with the immune system may help guide interventions to alter the microbiome toward a state of lifelong health.
Collapse
|
82
|
Chen B, Avinashi V, Dobson S. Fecal microbiota transplantation for recurrent clostridium difficile infection in children. J Infect 2018. [PMID: 28646951 DOI: 10.1016/s0163-4453(17)30202-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Fecal microbiota transplantation (FMT) is a relatively simple, promising treatment for recurrent Clostridium difficile infection. While there are a wide variety of approaches including mode of delivery, the results are nonetheless encouraging, even amongst younger children. Experience with FMT in the pediatric population is increasing, showing similar success compared to adults. This article will provide an overview of C. difficile infection along with review of the rationale, methods and complications of FMT including the current experience of FMT in children.
Collapse
Affiliation(s)
- Becky Chen
- Division of Gastroenterology, Hepatology and Nutrition, British Columbia Children's Hospital, Vancouver, BC, Canada
| | - Vishal Avinashi
- Division of Gastroenterology, Hepatology and Nutrition, British Columbia Children's Hospital, Vancouver, BC, Canada.
| | - Simon Dobson
- Division of Infectious Diseases, British Columbia Children's Hospital, Vancouver, BC, Canada
| |
Collapse
|
83
|
Daliri EBM, Wei S, Oh DH, Lee BH. The human microbiome and metabolomics: Current concepts and applications. Crit Rev Food Sci Nutr 2018; 57:3565-3576. [PMID: 27767329 DOI: 10.1080/10408398.2016.1220913] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The mammalian gastrointestinal tract has co-developed with a large number of microbes in a symbiotic relationship over millions of years. Recent studies indicate that indigenous bacteria are intimate with the intestine and play essential roles in health and disease. In the quest to maintain a stable niche, these prokaryotes influence multiple host metabolic pathways, resulting from an interactive host-microbiota metabolic signaling and impacting strongly on the metabolic phenotypes of the host. Since dysbiosis of the gut bacteria result in alteration in the levels of certain microbial and host co-metabolites, identifying these markers could enhance early detection of diseases. Also, identification of these metabolic fingerprints could give us clues as to how to manipulate the microbiome to promote health or treat diseases. This review provides an overview of our current knowledge of the microbiome and metablomics, applications and the future perspectives.
Collapse
Affiliation(s)
- Eric Banan-Mwine Daliri
- a Department of Food Science and Biotechnology , Kangwon National University , Chuncheon , South Korea
| | - Shuai Wei
- a Department of Food Science and Biotechnology , Kangwon National University , Chuncheon , South Korea
| | - Deog H Oh
- a Department of Food Science and Biotechnology , Kangwon National University , Chuncheon , South Korea
| | - Byong H Lee
- a Department of Food Science and Biotechnology , Kangwon National University , Chuncheon , South Korea.,b Department of Food Science/Agricultural Chemistry , McGill University , Ste.-Anne-de-Bellevue , Quebec , Canada
| |
Collapse
|
84
|
Friedman-Korn T, Livovsky DM, Maharshak N, Aviv Cohen N, Paz K, Bar-Gil Shitrit A, Goldin E, Koslowsky B. Fecal Transplantation for Treatment of Clostridium Difficile Infection in Elderly and Debilitated Patients. Dig Dis Sci 2018; 63:198-203. [PMID: 29134299 DOI: 10.1007/s10620-017-4833-2] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Accepted: 11/01/2017] [Indexed: 02/08/2023]
Abstract
BACKGROUND Fecal microbiota transplantation (FMT) is a new technique recently introduced to treat recurrent Clostridium difficile infection (CDI). Little is known about the efficacy and risks of FMT in elderly and ill patients. AIM To investigate FMT efficacy in ill and elderly patients compared to conventional treatment. METHODS The study comprised two groups of patients between 2012 and 2016 with recurrent CDI at two medical centers in Israel. The study group received FMT and the controls conventional therapy. The primary end points were CDI recurrence, length of hospitalization, and short-term survival. RESULTS Thirty-four patients altogether, (21 females, mean age 82 years) participated, 11 received FMT and 23 controls. Demographics and clinical characteristics were similar between the two groups. Comorbidity indexes, i.e., Charlson index was high in both groups. In the FMT group, 10/11 (90%) patients showed clinical improvement 3 days after initiating treatment compared to 9/23 (39%) in the control group, p = 0.02. Survival at 2 months did not differ between the groups (FMT 54%, Control 50%, p = 0.816), but mean survival in the FMT group was higher than in the control (12 vs. 4 months, respectively, p = 0.015). Two significant adverse events from the FMT group included suspected aspirations, both occurring during gastroscopy route of administration. CONCLUSIONS FMT is effective for elderly and very ill patients. Safety is a concern, but is rare even in patients with much comorbidity. Colonoscopy may be the preferred route of FMT infusion.
Collapse
Affiliation(s)
- Tali Friedman-Korn
- Digestive Diseases Institute, Shaare Zedek Medical Center, Jerusalem, Israel
| | - Dan Meir Livovsky
- Digestive Diseases Institute, Shaare Zedek Medical Center, Jerusalem, Israel
| | - Nitsan Maharshak
- Bacteriotherapy Clinic, Department of Gastroenterology and Liver Diseases, Tel Aviv Medical Center, Tel Aviv, Israel.,The Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Nathaniel Aviv Cohen
- Bacteriotherapy Clinic, Department of Gastroenterology and Liver Diseases, Tel Aviv Medical Center, Tel Aviv, Israel.,The Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Kalman Paz
- Digestive Diseases Institute, Shaare Zedek Medical Center, Jerusalem, Israel
| | | | - Eran Goldin
- Digestive Diseases Institute, Shaare Zedek Medical Center, Jerusalem, Israel
| | - Benjamin Koslowsky
- Digestive Diseases Institute, Shaare Zedek Medical Center, Jerusalem, Israel.
| |
Collapse
|
85
|
Zamudio-Tiburcio Á, Bermúdez-Ruiz H, Lezama-Guzmán HR, Guevara-Ortigoza MDP, Islas-Solares E, Sosa-López FA. Rompiendo paradigmas. Trasplante de microbiota intestinal: reporte preliminar. CIR CIR 2017; 85 Suppl 1:6-12. [DOI: 10.1016/j.circir.2016.11.017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2016] [Accepted: 11/26/2016] [Indexed: 02/07/2023]
|
86
|
Yoon S, Yu J, McDowell A, Kim SH, You HJ, Ko G. Bile salt hydrolase-mediated inhibitory effect of Bacteroides ovatus on growth of Clostridium difficile. J Microbiol 2017; 55:892-899. [PMID: 29076071 DOI: 10.1007/s12275-017-7340-4] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Revised: 09/11/2017] [Accepted: 09/27/2017] [Indexed: 02/08/2023]
Abstract
Clostridium difficile infection (CDI) is one of the most common nosocomial infections. Dysbiosis of the gut microbiota due to consumption of antibiotics is a major contributor to CDI. Recently, fecal microbiota transplantation (FMT) has been applied to treat CDI. However, FMT has important limitations including uncontrolled exposure to pathogens and standardization issues. Therefore, it is necessary to evaluate alternative treatment methods, such as bacteriotherapy, as well as the mechanism through which beneficial bacteria inhibit the growth of C. difficile. Here, we report bile acid-mediated inhibition of C. difficile by Bacteroides strains which can produce bile salt hydrolase (BSH). Bacteroides strains are not commonly used to treat CDI; however, as they comprise a large proportion of the intestinal microbiota, they can contribute to bile acid-mediated inhibition of C. difficile. The inhibitory effect on C. difficile growth increased with increasing bile acid concentration in the presence of Bacteroides ovatus SNUG 40239. Furthermore, this inhibitory effect on C. difficile growth was significantly attenuated when bile acid availability was reduced by cholestyramine, a bile acid sequestrant. The findings of this study are important due to the discovery of a new bacterial strain that in the presence of available bile acids inhibits growth of C. difficile. These results will facilitate development of novel bacteriotherapy strategies to control CDI.
Collapse
Affiliation(s)
- Soobin Yoon
- Department of Environmental Health Sciences, Graduate School of Public Health, Seoul National University, Seoul, 08826, Republic of Korea
| | - Junsun Yu
- Department of Environmental Health Sciences, Graduate School of Public Health, Seoul National University, Seoul, 08826, Republic of Korea
| | - Andrea McDowell
- Department of Environmental Health Sciences, Graduate School of Public Health, Seoul National University, Seoul, 08826, Republic of Korea
| | - Sung Ho Kim
- KoBioLabs, Inc., Seoul, 08826, Republic of Korea
| | - Hyun Ju You
- Center for Human and Environmental Microbiome, Institute of Health and Environment, Seoul National University, Seoul, 08826, Republic of Korea.
| | - GwangPyo Ko
- Department of Environmental Health Sciences, Graduate School of Public Health, Seoul National University, Seoul, 08826, Republic of Korea. .,KoBioLabs, Inc., Seoul, 08826, Republic of Korea. .,Center for Human and Environmental Microbiome, Institute of Health and Environment, Seoul National University, Seoul, 08826, Republic of Korea. .,Bio-MAX/N-Bio, Seoul National University, Seoul, 08826, Republic of Korea.
| |
Collapse
|
87
|
Wang WW, Zhang Y, Huang XB, You N, Zheng L, Li J. Fecal microbiota transplantation prevents hepatic encephalopathy in rats with carbon tetrachloride-induced acute hepatic dysfunction. World J Gastroenterol 2017; 23:6983-6994. [PMID: 29097871 PMCID: PMC5658316 DOI: 10.3748/wjg.v23.i38.6983] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Revised: 08/17/2017] [Accepted: 09/05/2017] [Indexed: 02/06/2023] Open
Abstract
AIM To investigate whether fecal microbiota transplantation (FMT) prevents hepatic encephalopathy (HE) in rats with carbon tetrachloride (CCl4)-induced acute hepatic dysfunction.
METHODS A rat model of HE was established with CCl4. Rat behaviors and spatial learning capability were observed, and hepatic necrosis, intestinal mucosal barrier, serum ammonia levels and intestinal permeability were determined in HE rats receiving FMT treatment. Furthermore, the expression of tight junction proteins (Claudin-1, Claudin-6 and Occludin), Toll-like receptor (TLR) 4/TLR9, interleukin (IL)-1β, IL-6 and tumor necrosis factor (TNF)-α was examined.
RESULTS FMT improved rat behaviors, HE grade and spatial learning capability. Moreover, FMT prevented hepatic necrosis and intestinal mucosal barrier damage, leading to hepatic clearance of serum ammonia levels and reduced intestinal permeability. The expression of TLR4 and TLR9, two potent mediators of inflammatory response, was significantly downregulated in the liver of rats treated with FMT. Consistently, circulating pro-inflammatory factors such as interleukin (IL)-1β, IL-6 and tumor necrosis factor-α were remarkably decreased, indicating that FMT is able to limit systemic inflammation by decreasing the expression of TLR4 and TLR9. Importantly, HE-induced loss of tight junction proteins (Claudin-1, Claudin-6 and Occludin) was restored in intestinal tissues of rats receiving FMT treatment.
CONCLUSION FMT enables protective effects in HE rats, and it improves the cognitive function and reduces the liver function indexes. FMT may cure HE by altering the intestinal permeability and improving the TLR response of the liver.
Collapse
Affiliation(s)
- Wei-Wei Wang
- Department of Hepatobiliary Surgery, the Second Affiliated Hospital of Third Military Medical University, Chongqing 400037, China
| | - Yu Zhang
- Department of Hepatobiliary Surgery, the Second Affiliated Hospital of Third Military Medical University, Chongqing 400037, China
| | - Xiao-Bing Huang
- Department of Hepatobiliary Surgery, the Second Affiliated Hospital of Third Military Medical University, Chongqing 400037, China
| | - Nan You
- Department of Hepatobiliary Surgery, the Second Affiliated Hospital of Third Military Medical University, Chongqing 400037, China
| | - Lu Zheng
- Department of Hepatobiliary Surgery, the Second Affiliated Hospital of Third Military Medical University, Chongqing 400037, China
| | - Jing Li
- Department of Hepatobiliary Surgery, the Second Affiliated Hospital of Third Military Medical University, Chongqing 400037, China
| |
Collapse
|
88
|
Hafza N, Challita C, Dandachi I, Bousaab M, Dahdouh E, Daoud Z. Competition assays between ESBL-producing E. coli and K. pneumoniae isolates collected from Lebanese elderly: An additional cost on fitness. J Infect Public Health 2017; 11:393-397. [PMID: 28988774 DOI: 10.1016/j.jiph.2017.09.010] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Revised: 08/13/2017] [Accepted: 09/09/2017] [Indexed: 11/15/2022] Open
Abstract
The dissemination of Multi Drug Resistant Organisms (MDROs) is one of the major public health problems addressed nowadays. High fecal carriage rates of MDR Enterobacteriaceae were reported from Lebanese nursing homes. Studies have shown that the acquisition of resistance genes by bacteria might confer a fitness cost detected as a decrease in the frequency of these bacteria as compared to sensitive isolates. In this study, the competitive growth of MDR Enterobacteriaceae isolated from elderly is assessed. Sensitive and ESBL-producing Escherichia coli and Klebsiella pneumoniae isolates were identified. Inter-species in-vitro competition assays were conducted in different combinations. ESBL-producing K. pneumoniae presented a fitness cost when competing against sensitive E. coli. On the other hand, resistant E. coli only showed a fitness cost when growing in presence of two sensitive K. pneumoniae isolates. These results suggest that ESBL-production genes in E. coli and K. pneumoniae may confer a fitness cost that leads to the decrease in frequency of these bacteria in interspecies competitions. Culturing bacteria in a medium with more diverse isolates can provide better insights into bacterial competition and resistance dynamics, which can be exploited in the search for alternative therapeutic approaches towards the colonization of resistant bacteria.
Collapse
Affiliation(s)
| | - Caren Challita
- Faculty of Medicine, Clinical Microbiology Lab, University of Balamand, Lebanon
| | - Iman Dandachi
- Faculty of Medicine, Clinical Microbiology Lab, University of Balamand, Lebanon
| | - Mounir Bousaab
- Faculty of Medicine, Clinical Microbiology Lab, University of Balamand, Lebanon
| | - Elias Dahdouh
- Faculty of Veterinary, Department of Animal Health, Universidad Complutense de Madrid, Madrid, Spain
| | - Ziad Daoud
- Faculty of Medicine, Clinical Microbiology Lab, University of Balamand, Lebanon.
| |
Collapse
|
89
|
Aggarwala V, Liang G, Bushman FD. Viral communities of the human gut: metagenomic analysis of composition and dynamics. Mob DNA 2017; 8:12. [PMID: 29026445 PMCID: PMC5627405 DOI: 10.1186/s13100-017-0095-y] [Citation(s) in RCA: 98] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Accepted: 09/24/2017] [Indexed: 01/17/2023] Open
Abstract
Background The numerically most abundant biological entities on Earth are viruses. Vast populations prey on the cellular microbiota in all habitats, including the human gut. Main body Here we review approaches for studying the human virome, and some recent results on movement of viral sequences between bacterial cells and eukaryotic hosts. We first overview biochemical and bioinformatic methods, emphasizing that specific choices in the methods used can have strong effects on the results obtained. We then review studies characterizing the virome of the healthy human gut, which reveal that most of the viruses detected are typically uncharacterized phage - the viral dark matter - and that viruses that infect human cells are encountered only rarely. We then review movement of phage between bacterial cells during antibiotic treatment. Here a radical proposal for extensive movement of antibiotic genes on phage has been challenged by a careful reanalysis of the metagenomic annotation methods used. We then review two recent studies of movement of whole phage communities between human individuals during fecal microbial transplantation, which emphasize the possible role of lysogeny in dispersal. Short conclusion Methods for studying the human gut virome are improving, yielding interesting data on movement of phage genes between cells and mammalian host organisms. However, viral populations are vast, and studies of their composition and function are just beginning.
Collapse
Affiliation(s)
- Varun Aggarwala
- Department of Microbiology, University of Pennsylvania School of Medicine, 3610 Hamilton Walk, Philadelphia, PA 19104-6076 USA
| | - Guanxiang Liang
- Department of Microbiology, University of Pennsylvania School of Medicine, 3610 Hamilton Walk, Philadelphia, PA 19104-6076 USA.,Division of Gastroenterology, Hepatology, and Nutrition, Children's Hospital of Philadelphia, Philadelphia, PA 19104-4319 USA
| | - Frederic D Bushman
- Department of Microbiology, University of Pennsylvania School of Medicine, 3610 Hamilton Walk, Philadelphia, PA 19104-6076 USA
| |
Collapse
|
90
|
Kelly BJ, Tebas P. Clinical Practice and Infrastructure Review of Fecal Microbiota Transplantation for Clostridium difficile Infection. Chest 2017; 153:266-277. [PMID: 28923757 DOI: 10.1016/j.chest.2017.09.002] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Revised: 08/31/2017] [Accepted: 09/05/2017] [Indexed: 12/14/2022] Open
Abstract
A substantial proportion of Clostridium difficile infection (CDI) cases recur after completion of antibiotic therapy, and antibiotic cure rates diminish with each recurrence of CDI. Fecal microbiota transplantation (FMT) is an effective therapy for recurrent FMT, which otherwise requires prolonged or indefinite antibiotic treatment. FMT is performed by introducing the fecal microbial community obtained from a healthy donor or pool of donors into the stomach, small intestine, or colon of a patient with CDI. Multiple clinical trials support the usefulness of FMT in treating recurrent CDI, and CDI treatment guidelines now include consideration of FMT at the third CDI recurrence. However, there remain challenges to incorporating FMT into clinical practice. First, methods of fecal bacterial community processing vary, as do methods of FMT administration. Second, the optimal dosing strategy and expected benefit of FMT for refractory CDI, particularly for severe and severe complicated cases, are uncertain. Third, the US Food and Drug Administration (FDA) considers FMT an investigational treatment. Fourth, insurance reimbursement for FMT usually falls short of FMT administration costs. In the setting of rising C difficile incidence and growing evidence for FMT efficacy, the demand for FMT has increased. However, uncertainty surrounding optimal FMT preparation and administration methods, FDA oversight, and insurance reimbursement presently limits the clinical practice of FMT.
Collapse
Affiliation(s)
- Brendan J Kelly
- Division of Infectious Diseases and Fecal Microbiota Transplantation Program, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA.
| | - Pablo Tebas
- Division of Infectious Diseases and Fecal Microbiota Transplantation Program, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
91
|
Kufel WD, Devanathan AS, Marx AH, Weber DJ, Daniels LM. Bezlotoxumab: A Novel Agent for the Prevention of Recurrent Clostridium difficile Infection. Pharmacotherapy 2017; 37:1298-1308. [PMID: 28730660 DOI: 10.1002/phar.1990] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
During the past decade, the incidence and severity of Clostridium difficile infection (CDI) have significantly increased, leading to a rise in CDI-associated hospitalizations, health care costs, and mortality. Although treatment options exist for CDI, recurrence is frequent following treatment. Furthermore, patients with at least one CDI recurrence are at an increased risk of developing additional recurrences. A novel approach to the prevention of recurrent CDI is the use of monoclonal antibodies directed against the toxins responsible for CDI as an adjunct to antibiotic treatment. Bezlotoxumab, a human monoclonal antibody that binds and neutralizes C. difficile toxin B, is the first therapeutic agent to receive United States Food and Drug Administration approval for the prevention of CDI recurrence. Clinical studies have demonstrated superior efficacy of bezlotoxumab in adults receiving antibiotic therapy for CDI compared with antibiotic therapy alone for the prevention of CDI recurrence. Bezlotoxumab was well tolerated in clinical trials, with the most common adverse effects being nausea, vomiting, fatigue, pyrexia, headache, and diarrhea. The demonstrated efficacy, safety, and characteristics of bezlotoxumab present an advance in prevention of CDI recurrence.
Collapse
Affiliation(s)
- Wesley D Kufel
- Department of Pharmacy, University of North Carolina Medical Center, Chapel Hill, North Carolina.,Department of Practice Advancement and Clinical Education, University of North Carolina Eshelman School of Pharmacy, Chapel Hill, North Carolina
| | - Aaron S Devanathan
- Department of Pharmacy, University of North Carolina Medical Center, Chapel Hill, North Carolina.,Department of Practice Advancement and Clinical Education, University of North Carolina Eshelman School of Pharmacy, Chapel Hill, North Carolina
| | - Ashley H Marx
- Department of Pharmacy, University of North Carolina Medical Center, Chapel Hill, North Carolina.,Department of Practice Advancement and Clinical Education, University of North Carolina Eshelman School of Pharmacy, Chapel Hill, North Carolina
| | - David J Weber
- Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina.,School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Lindsay M Daniels
- Department of Pharmacy, University of North Carolina Medical Center, Chapel Hill, North Carolina.,Department of Practice Advancement and Clinical Education, University of North Carolina Eshelman School of Pharmacy, Chapel Hill, North Carolina
| |
Collapse
|
92
|
Chopra T, Goldstein EJC, Gorbach SL. Rethinking Strategies to Select Antibiotic Therapy in Clostridium difficile infection. Pharmacotherapy 2017; 36:1281-1289. [PMID: 27862113 DOI: 10.1002/phar.1863] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
In recent years, Clostridium difficile infection (CDI) has become a global public health threat associated with increased morbidity, mortality, and economic burden, all of which are exacerbated with disease recurrence. Current guidelines informing treatment decisions are largely based on definitions of disease severity at diagnosis, with subjective components not well delineated across treatment algorithms and clinical trials. Furthermore, there is little evidence linking severity at onset to outcome. However, reducing the risk of recurrence may offer both a better outcome for the individual and decreased downstream economic impact. The authors present data supporting the opinion that patients deemed at low risk for recurrence should receive vancomycin (or metronidazole when cost is an issue), while those at higher risk of recurrence would benefit from fidaxomicin treatment. Although further prospective studies are needed, choosing treatment with the goal of preventing recurrent CDI may offer a better guide than disease severity.
Collapse
Affiliation(s)
- Teena Chopra
- Division of Infectious Diseases, Detroit Medical Center and Wayne State University, Detroit, Michigan
| | - Ellie J C Goldstein
- David Geffen School of Medicine, Los Angeles, California.,R.M. Alden Research Laboratory, Santa Monica, California
| | - Sherwood L Gorbach
- Department of Public Health and Family Medicine, Tufts University School of Medicine, Boston, Massachusetts
| |
Collapse
|
93
|
Alrabaa S, Jariwala R, Zeitler K, Montero J. Fecal microbiota transplantation outcomes in immunocompetent and immunocompromised patients: A single-center experience. Transpl Infect Dis 2017; 19. [DOI: 10.1111/tid.12726] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Revised: 02/20/2017] [Accepted: 03/06/2017] [Indexed: 12/21/2022]
Affiliation(s)
- Sally Alrabaa
- University of South Florida Morsani College of Medicine; Tampa FL USA
| | - Ripal Jariwala
- Department of Pharmacy; Tampa General Hospital; Tampa FL USA
| | - Kristen Zeitler
- Department of Pharmacy; Tampa General Hospital; Tampa FL USA
| | - Jose Montero
- University of South Florida Morsani College of Medicine; Tampa FL USA
| |
Collapse
|
94
|
Sung J, Kim S, Cabatbat JJT, Jang S, Jin YS, Jung GY, Chia N, Kim PJ. Global metabolic interaction network of the human gut microbiota for context-specific community-scale analysis. Nat Commun 2017; 8:15393. [PMID: 28585563 PMCID: PMC5467172 DOI: 10.1038/ncomms15393] [Citation(s) in RCA: 173] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Accepted: 03/27/2017] [Indexed: 12/18/2022] Open
Abstract
A system-level framework of complex microbe–microbe and host–microbe chemical cross-talk would help elucidate the role of our gut microbiota in health and disease. Here we report a literature-curated interspecies network of the human gut microbiota, called NJS16. This is an extensive data resource composed of ∼570 microbial species and 3 human cell types metabolically interacting through >4,400 small-molecule transport and macromolecule degradation events. Based on the contents of our network, we develop a mathematical approach to elucidate representative microbial and metabolic features of the gut microbial community in a given population, such as a disease cohort. Applying this strategy to microbiome data from type 2 diabetes patients reveals a context-specific infrastructure of the gut microbial ecosystem, core microbial entities with large metabolic influence, and frequently produced metabolic compounds that might indicate relevant community metabolic processes. Our network presents a foundation towards integrative investigations of community-scale microbial activities within the human gut. The metabolic interactions between gut microbes and host cells play roles in human health. Here, Sung et al. present a literature-curated metabolic network of the human gut microbiota and three human cell types, together with a mathematical approach to identify distinct microbial and metabolic features in gut microbiomes.
Collapse
Affiliation(s)
- Jaeyun Sung
- Asia Pacific Center for Theoretical Physics, Pohang, Gyeongbuk 37673, Republic of Korea.,Center for Computational and Integrative Biology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114, USA.,Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, USA
| | - Seunghyeon Kim
- Asia Pacific Center for Theoretical Physics, Pohang, Gyeongbuk 37673, Republic of Korea.,Department of Physics, Pohang University of Science and Technology, Pohang, Gyeongbuk 37673, Republic of Korea.,The Abdus Salam International Centre for Theoretical Physics, 34151 Trieste, Italy
| | | | - Sungho Jang
- Department of Chemical Engineering, Pohang University of Science and Technology, Pohang, Gyeongbuk 37673, Republic of Korea
| | - Yong-Su Jin
- Department of Food Science and Human Nutrition, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA.,Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA
| | - Gyoo Yeol Jung
- Department of Chemical Engineering, Pohang University of Science and Technology, Pohang, Gyeongbuk 37673, Republic of Korea.,School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology, Pohang, Gyeongbuk 37673, Republic of Korea
| | - Nicholas Chia
- Microbiome Program, Center for Individualized Medicine, Mayo Clinic, Rochester, Minnesota 55905, USA.,Department of Surgery, Mayo Clinic, Rochester, Minnesota 55905, USA.,Department of Biomedical Engineering, Mayo College, Rochester, Minnesota 55905, USA
| | - Pan-Jun Kim
- Asia Pacific Center for Theoretical Physics, Pohang, Gyeongbuk 37673, Republic of Korea.,Department of Physics, Pohang University of Science and Technology, Pohang, Gyeongbuk 37673, Republic of Korea.,Department of Physics, Korea Advanced Institute of Science and Technology, 291 Daehak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea
| |
Collapse
|
95
|
Wolf-Meyer MJ. Normal, Regular, and Standard: Scaling the Body through Fecal Microbial Transplants. Med Anthropol Q 2017; 31:297-314. [DOI: 10.1111/maq.12328] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2015] [Revised: 05/27/2016] [Accepted: 07/12/2016] [Indexed: 12/31/2022]
|
96
|
Galla S, Chakraborty S, Mell B, Vijay-Kumar M, Joe B. Microbiotal-Host Interactions and Hypertension. Physiology (Bethesda) 2017; 32:224-233. [PMID: 28404738 DOI: 10.1152/physiol.00003.2017] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Revised: 02/24/2017] [Accepted: 02/24/2017] [Indexed: 12/12/2022] Open
Abstract
Hypertension, or elevated blood pressure (BP), has been extensively researched over decades and clearly demonstrated to be caused due to a combination of host genetic and environmental factors. Although much research remains to be conducted to pin-point the precise genetic elements on the host genome that control BP, new lines of evidence are emerging to indicate that, besides the host genome, the genomes of all indigenous commensal micro-organisms, collectively referred to as the microbial metagenome or microbiome, are important, but largely understudied, determinants of BP. Unlike the rigid host genome, the microbiome or the "second genome" can be altered by diet or microbiotal transplantation in the host. This possibility is attractive from the perspective of exploiting the microbiotal composition for clinical management of inherited hypertension. Thus, focusing on the limited current literature supporting a role for the microbiome in BP regulation, this review highlights the need to further explore the role of the co-existence of host and the microbiota as an organized biological unit called the "holobiont" in the context of BP regulation.
Collapse
Affiliation(s)
- Sarah Galla
- Physiological Genomics Laboratory, Center for Hypertension and Personalized Medicine, Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio; and
| | - Saroj Chakraborty
- Physiological Genomics Laboratory, Center for Hypertension and Personalized Medicine, Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio; and
| | - Blair Mell
- Physiological Genomics Laboratory, Center for Hypertension and Personalized Medicine, Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio; and
| | - Matam Vijay-Kumar
- Department of Nutritional Sciences and Medicine, The Pennsylvania State University, University Park, Pennsylvania
| | - Bina Joe
- Physiological Genomics Laboratory, Center for Hypertension and Personalized Medicine, Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio; and
| |
Collapse
|
97
|
Lee STM, Kahn SA, Delmont TO, Shaiber A, Esen ÖC, Hubert NA, Morrison HG, Antonopoulos DA, Rubin DT, Eren AM. Tracking microbial colonization in fecal microbiota transplantation experiments via genome-resolved metagenomics. MICROBIOME 2017; 5:50. [PMID: 28473000 PMCID: PMC5418705 DOI: 10.1186/s40168-017-0270-x] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/04/2016] [Accepted: 04/22/2017] [Indexed: 05/11/2023]
Abstract
BACKGROUND Fecal microbiota transplantation (FMT) is an effective treatment for recurrent Clostridium difficile infection and shows promise for treating other medical conditions associated with intestinal dysbioses. However, we lack a sufficient understanding of which microbial populations successfully colonize the recipient gut, and the widely used approaches to study the microbial ecology of FMT experiments fail to provide enough resolution to identify populations that are likely responsible for FMT-derived benefits. METHODS We used shotgun metagenomics together with assembly and binning strategies to reconstruct metagenome-assembled genomes (MAGs) from fecal samples of a single FMT donor. We then used metagenomic mapping to track the occurrence and distribution patterns of donor MAGs in two FMT recipients. RESULTS Our analyses revealed that 22% of the 92 highly complete bacterial MAGs that we identified from the donor successfully colonized and remained abundant in two recipients for at least 8 weeks. Most MAGs with a high colonization rate belonged to the order Bacteroidales. The vast majority of those that lacked evidence of colonization belonged to the order Clostridiales, and colonization success was negatively correlated with the number of genes related to sporulation. Our analysis of 151 publicly available gut metagenomes showed that the donor MAGs that colonized both recipients were prevalent, and the ones that colonized neither were rare across the participants of the Human Microbiome Project. Although our dataset showed a link between taxonomy and the colonization ability of a given MAG, we also identified MAGs that belong to the same taxon with different colonization properties, highlighting the importance of an appropriate level of resolution to explore the functional basis of colonization and to identify targets for cultivation, hypothesis generation, and testing in model systems. CONCLUSIONS The analytical strategy adopted in our study can provide genomic insights into bacterial populations that may be critical to the efficacy of FMT due to their success in gut colonization and metabolic properties, and guide cultivation efforts to investigate mechanistic underpinnings of this procedure beyond associations.
Collapse
Affiliation(s)
- Sonny T M Lee
- Section of Gastroenterology, Hepatology and Nutrition, Department of Medicine, University of Chicago Medicine, Chicago, IL, USA
| | - Stacy A Kahn
- Section of Gastroenterology, Hepatology and Nutrition, Department of Medicine, University of Chicago Medicine, Chicago, IL, USA
- Present address: Boston Children's Hospital, Inflammatory Bowel Disease Center, Boston, MA, USA
| | - Tom O Delmont
- Section of Gastroenterology, Hepatology and Nutrition, Department of Medicine, University of Chicago Medicine, Chicago, IL, USA
| | - Alon Shaiber
- Section of Gastroenterology, Hepatology and Nutrition, Department of Medicine, University of Chicago Medicine, Chicago, IL, USA
| | - Özcan C Esen
- Section of Gastroenterology, Hepatology and Nutrition, Department of Medicine, University of Chicago Medicine, Chicago, IL, USA
| | - Nathaniel A Hubert
- Section of Gastroenterology, Hepatology and Nutrition, Department of Medicine, University of Chicago Medicine, Chicago, IL, USA
| | - Hilary G Morrison
- Josephine Bay Paul Center for Comparative Molecular Biology and Evolution, Marine Biological Laboratory, Woods Hole, 02543, MA, USA
| | - Dionysios A Antonopoulos
- Section of Gastroenterology, Hepatology and Nutrition, Department of Medicine, University of Chicago Medicine, Chicago, IL, USA
| | - David T Rubin
- Section of Gastroenterology, Hepatology and Nutrition, Department of Medicine, University of Chicago Medicine, Chicago, IL, USA
| | - A Murat Eren
- Section of Gastroenterology, Hepatology and Nutrition, Department of Medicine, University of Chicago Medicine, Chicago, IL, USA.
- Josephine Bay Paul Center for Comparative Molecular Biology and Evolution, Marine Biological Laboratory, Woods Hole, 02543, MA, USA.
| |
Collapse
|
98
|
Rätsep M, Kõljalg S, Sepp E, Smidt I, Truusalu K, Songisepp E, Stsepetova J, Naaber P, Mikelsaar RH, Mikelsaar M. A combination of the probiotic and prebiotic product can prevent the germination of Clostridium difficile spores and infection. Anaerobe 2017; 47:94-103. [PMID: 28465256 DOI: 10.1016/j.anaerobe.2017.03.019] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Revised: 03/23/2017] [Accepted: 03/28/2017] [Indexed: 12/17/2022]
Abstract
Clostridium difficile infection (CDI) is one of the most prevalent healthcare associated infections in hospitals and nursing homes. Different approaches are used for prevention of CDI. Absence of intestinal lactobacilli and bifidobacteria has been associated with C. difficile colonization in hospitalized patients. Our aim was to test a) the susceptibility of C. difficile strains of different origin and the intestinal probiotic Lactobacillus plantarum Inducia (DSM 21379) to various antimicrobial preparations incl. metronidazole, vancomycin; b) the susceptibility of C. difficile strains to antagonistic effects of the probiotic L. plantarum Inducia, prebiotic xylitol (Xyl) and their combination as a synbiotic (Syn) product; c) the suppression of germination of C. difficile spores in vitro and in vivo in animal model of C. difficile infection with Inducia, Xyl and Syn treatment. The VPI strain 10463 (ATCC 43255), epidemic strain (M 13042) and clinical isolates (n = 12) of C. difficile from Norway and Estonia were susceptible and contrarily L. plantarum Inducia resistant to vancomycin, metronidazole and ciprofloxacin. The intact cells of Inducia, natural and neutralized cell free supernatant inhibited in vitro the growth of tested C. difficile reference strain VPI and Estonian and Norwegian clinical isolates of C. difficile after co-cultivation. This effect against C. difficile sustained in liquid media under ampicillin (0.75 μg/ml) and Xyl (5%) application. Further, incubation of Inducia in the media with 5% Xyl fully stopped germination of spores of C. difficile VPI strain after 48 h. In infection model the 48 hamsters were administered ampicillin (30 mg/kg) and 10-30 spores of C. difficile VPI strain. They also received five days before and after the challenge a pretreatment with a synbiotic (single daily dose of L. plantarum Inducia 1 ml of 1010 CFU/ml and 20% xylitol in 1 ml by orogastric gavage). The survival rate of hamsters was increased to 78% compared to 13% (p = 0.003) survival rate of hamsters who received no treatment. When administered Xyl the survival rate of hamsters reached 56% vs.13% (p = 0.06). In both Syn (6/9, p = 0.003) and Xyl (3/9, p = 0.042) groups the number of animals not colonized with C. difficile significantly increased. In conclusion, the combination of xylitol with L. plantarum Inducia suppresses the germination of spores and outgrowth into vegetative toxin producing cells of C. difficile and reduces the colonization of gut with the pathogen. Putative therapeutical approach includes usage of the synbiotic during antimicrobial therapy for prevention of CDI and its potential to reduce recurrences of CDI.
Collapse
Affiliation(s)
- M Rätsep
- Institute of Biomedicine and Translational Medicine, University of Tartu, EE 50411, Estonia; Bio-Competence Centre of Healthy Dairy Products LLC, EE 51014, Estonia
| | - S Kõljalg
- Institute of Biomedicine and Translational Medicine, University of Tartu, EE 50411, Estonia
| | - E Sepp
- Institute of Biomedicine and Translational Medicine, University of Tartu, EE 50411, Estonia
| | - I Smidt
- Institute of Biomedicine and Translational Medicine, University of Tartu, EE 50411, Estonia
| | - K Truusalu
- Institute of Biomedicine and Translational Medicine, University of Tartu, EE 50411, Estonia
| | - E Songisepp
- Bio-Competence Centre of Healthy Dairy Products LLC, EE 51014, Estonia
| | - J Stsepetova
- Institute of Biomedicine and Translational Medicine, University of Tartu, EE 50411, Estonia
| | - P Naaber
- Institute of Biomedicine and Translational Medicine, University of Tartu, EE 50411, Estonia
| | - R H Mikelsaar
- Institute of Biomedicine and Translational Medicine, University of Tartu, EE 50411, Estonia
| | - M Mikelsaar
- Institute of Biomedicine and Translational Medicine, University of Tartu, EE 50411, Estonia; Bio-Competence Centre of Healthy Dairy Products LLC, EE 51014, Estonia.
| |
Collapse
|
99
|
Faner R, Sibila O, Agustí A, Bernasconi E, Chalmers JD, Huffnagle GB, Manichanh C, Molyneaux PL, Paredes R, Pérez Brocal V, Ponomarenko J, Sethi S, Dorca J, Monsó E. The microbiome in respiratory medicine: current challenges and future perspectives. Eur Respir J 2017; 49:49/4/1602086. [PMID: 28404649 DOI: 10.1183/13993003.02086-2016] [Citation(s) in RCA: 161] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Accepted: 02/08/2017] [Indexed: 12/15/2022]
Abstract
The healthy lung has previously been considered to be a sterile organ because standard microbiological culture techniques consistently yield negative results. However, culture-independent techniques report that large numbers of microorganisms coexist in the lung. There are many unknown aspects in the field, but available reports show that the lower respiratory tract microbiota: 1) is similar in healthy subjects to the oropharyngeal microbiota and dominated by members of the Firmicutes, Bacteroidetes and Proteobacteria phyla; 2) shows changes in smokers and well-defined differences in chronic respiratory diseases, although the temporal and spatial kinetics of these changes are only partially known; and 3) shows relatively abundant non-cultivable bacteria in chronic obstructive pulmonary disease, idiopathic pulmonary fibrosis, cystic fibrosis and bronchiectasis, with specific patterns for each disease. In all of these diseases, a loss of diversity, paralleled by an over-representation of Proteobacteria (dysbiosis), has been related to disease severity and exacerbations. However, it is unknown whether dysbiosis is a cause or a consequence of the damage to bronchoalveolar surfaces.Finally, little is known about bacterial functionality and the interactions between viruses, fungi and bacteria. It is expected that future research in bacterial gene expressions, metagenomics longitudinal analysis and host-microbiome animal models will help to move towards targeted microbiome interventions in respiratory diseases.
Collapse
Affiliation(s)
- Rosa Faner
- Hospital Clinic, IDIBAPS, Universitat de Barcelona, Barcelona, Spain.,CIBER de Enfermedades Respiratorias - CIBERES, Madrid, Spain.,These co-primary authors contributed equally to this work
| | - Oriol Sibila
- Hospital Universitari de la Santa Creu i Sant Pau, Universitat Autónoma Barcelona, Barcelona, Spain.,These co-primary authors contributed equally to this work
| | - Alvar Agustí
- Hospital Clinic, IDIBAPS, Universitat de Barcelona, Barcelona, Spain.,CIBER de Enfermedades Respiratorias - CIBERES, Madrid, Spain
| | - Eric Bernasconi
- Service de Pneumologie, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | | | | | - Chaysavanh Manichanh
- Dept of Gastroenterology, Vall d'Hebron Research Institute, Barcelona, Spain.,CIBER de Enfermedades Hepáticas y Digestivas (CIBEREHD), Madrid, Spain
| | | | - Roger Paredes
- Hospital Universitari Germans Trias i Pujol, Universitat Autónoma Barcelona, Barcelona, Spain
| | - Vicente Pérez Brocal
- CIBER en Epidemiología y Salud Pública (CIBERESP), Madrid, Spain.,Joint Research Unit on Genomics and Health, Foundation for the Promotion of Health and Biomedical Research of Valencia Region (FISABIO-Public Health) and Cavanilles Institute for Biodiversity and Evolutionary Biology, University of Valencia, Valencia, Spain
| | - Julia Ponomarenko
- Centro de Regulación Genómica, Barcelona, Spain.,Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | | | - Jordi Dorca
- Hospital Universitari de Bellvitge, IDIBELL, Universitat de Barcelona, Hospitalet del Llobregat, Barcelona, Spain.,These co-senior authors contributed equally to this work
| | - Eduard Monsó
- CIBER de Enfermedades Respiratorias - CIBERES, Madrid, Spain .,Hospital Universitari Parc Taulí, Universitat Autònoma de Barcelona, Barcelona, Spain.,These co-senior authors contributed equally to this work
| |
Collapse
|
100
|
Martz SL, Guzman-Rodriguez M, He SM, Noordhof C, Hurlbut DJ, Gloor GB, Carlucci C, Weese S, Allen-Vercoe E, Sun J, Claud EC, Petrof EO. A human gut ecosystem protects against C. difficile disease by targeting TcdA. J Gastroenterol 2017; 52:452-465. [PMID: 27329502 PMCID: PMC5177537 DOI: 10.1007/s00535-016-1232-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Accepted: 06/06/2016] [Indexed: 02/07/2023]
Abstract
BACKGROUND A defined Microbial Ecosystem Therapeutic (MET-1, or "RePOOPulate") derived from the feces of a healthy volunteer can cure recurrent C. difficile infection (rCDI) in humans. The mechanisms of action whereby healthy microbiota protect against rCDI remain unclear. Since C. difficile toxins are largely responsible for the disease pathology of CDI, we hypothesized that MET-1 exerts its protective effects by inhibiting the effects of these toxins on the host. METHODS A combination of in vivo (antibiotic-associated mouse model of C. difficile colitis, mouse ileal loop model) and in vitro models (FITC-phalloidin staining, F actin Western blots and apoptosis assay in Caco2 cells, transepithelial electrical resistance measurements in T84 cells) were employed. RESULTS MET-1 decreased both local and systemic inflammation in infection and decreased both the cytotoxicity and the amount of TcdA detected in stool, without an effect on C. difficile viability. MET-1 protected against TcdA-mediated damage in a murine ileal loop model. MET-1 protected the integrity of the cytoskeleton in cells treated with purified TcdA, as indicated by FITC-phalloidin staining, F:G actin assays and preservation of transepithelial electrical resistance. Finally, co-incubation of MET-1 with purified TcdA resulted in decreased detectable TcdA by Western blot analysis. CONCLUSIONS MET-1 intestinal microbiota confers protection against C. difficile and decreases C. difficile-mediated inflammation through its protective effects against C. difficile toxins, including enhancement of host barrier function and degradation of TcdA. The effect of MET-1 on C. difficile viability seems to offer little, if any, contribution to its protective effects on the host.
Collapse
Affiliation(s)
- Sarah Lynn Martz
- Division of Infectious Diseases/GI Diseases Research Unit Wing, Department of Medicine, Kingston General Hospital, Queen's University, 76 Stuart Street, Kingston, ON, K7L 2V7, Canada
| | - Mabel Guzman-Rodriguez
- Division of Infectious Diseases/GI Diseases Research Unit Wing, Department of Medicine, Kingston General Hospital, Queen's University, 76 Stuart Street, Kingston, ON, K7L 2V7, Canada
| | - Shu-Mei He
- Division of Infectious Diseases/GI Diseases Research Unit Wing, Department of Medicine, Kingston General Hospital, Queen's University, 76 Stuart Street, Kingston, ON, K7L 2V7, Canada
| | - Curtis Noordhof
- Division of Infectious Diseases/GI Diseases Research Unit Wing, Department of Medicine, Kingston General Hospital, Queen's University, 76 Stuart Street, Kingston, ON, K7L 2V7, Canada
| | - David John Hurlbut
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, ON, K7L 2V7, Canada
| | - Gregory Brian Gloor
- Department of Biochemistry, University of Western Ontario, London, ON, N6A 5C1, Canada
| | - Christian Carlucci
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, ON, N1G 2W1, Canada
| | - Scott Weese
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, ON, N1G 2W1, Canada
| | - Emma Allen-Vercoe
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, ON, N1G 2W1, Canada
| | - Jun Sun
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Erika Chiong Claud
- Departments of Pediatrics and Medicine, University of Chicago, Chicago, IL, 60637, USA
| | - Elaine Olga Petrof
- Division of Infectious Diseases/GI Diseases Research Unit Wing, Department of Medicine, Kingston General Hospital, Queen's University, 76 Stuart Street, Kingston, ON, K7L 2V7, Canada.
| |
Collapse
|