1
|
Häsler R, Mikš MH, Bajic D, Soyyilmaz B, Bendik I, van Buul VJ, Steinert RE, Rehman A. Human milk oligosaccharides modulating inflammation in infants, adults and older individuals - from concepts to applications. Adv Nutr 2025:100433. [PMID: 40287068 DOI: 10.1016/j.advnut.2025.100433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Revised: 04/17/2025] [Accepted: 04/18/2025] [Indexed: 04/29/2025] Open
Abstract
The increasing global prevalence of inflammatory diseases such as ulcerative colitis and irritable bowel syndrome, represents a challenging task for healthcare systems. Several approaches to disease management target the intestinal microbiome, which plays a key role in health and disease. One promising approach is modulating the microbiome using human milk oligosaccharides (HMOs). Originating from human milk, HMOs are indigestible carbohydrates which act in a host-optimized prebiotic fashion by providing an energy source for health-promoting intestinal bacteria and exhibiting systemic effects. Commercial products supporting infant health and development have been the primary fields of HMO application. Advancements in the large-scale production of HMOs through bioengineering and precision fermentation have led to evaluating their potential for managing inflammatory diseases. Several in vitro studies and observations on model systems have been clinically validated in infants, resulting in a large body of evidence supporting the safety and efficacy of HMOs in inflammatory disorders. While novel approaches seek to explore interventions in adults, the primary goal for the future is to provide cost-efficient, safe, and reliable healthcare compounds across all age groups.
Collapse
Affiliation(s)
- Robert Häsler
- Department of Dermatology and Allergology, University Kiel, Rosalind-Franklin-Straße 9, 24105 Kiel, Germany
| | - Marta Hanna Mikš
- University of Warmia and Mazury in Olsztyn, Faculty of Food Science, Plac Cieszynski 1, 10-726, Olsztyn, Poland; dsm-firmenich, Kogle Allé 4, Hørsholm, DK-2970, Denmark
| | - Danica Bajic
- dsm-firmenich, Wurmisweg 576, CH-4303 Kaiseraugst, Switzerland
| | | | - Igor Bendik
- dsm-firmenich, Wurmisweg 576, CH-4303 Kaiseraugst, Switzerland
| | | | | | - Ateequr Rehman
- dsm-firmenich, Wurmisweg 576, CH-4303 Kaiseraugst, Switzerland
| |
Collapse
|
2
|
Edwards M, Brockmann L. Microbiota-dependent modulation of intestinal anti-inflammatory CD4 + T cell responses. Semin Immunopathol 2025; 47:23. [PMID: 40167791 DOI: 10.1007/s00281-025-01049-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Accepted: 03/08/2025] [Indexed: 04/02/2025]
Abstract
Barrier organs such as the gastrointestinal tract, lungs, and skin are colonized by diverse microbial strains, including bacteria, viruses, and fungi. These microorganisms, collectively known as the commensal microbiota, play critical roles in maintaining health by defending against pathogens, metabolizing nutrients, and providing essential metabolites. In the gut, commensal-derived antigens are frequently sensed by the intestinal immune system. Maintaining tolerance toward these beneficial microbial species is crucial, as failure to do so can lead to chronic inflammatory conditions like inflammatory bowel disease (IBD) and can even affect systemic immune or metabolic health. The immune system carefully regulates responses to commensals through various mechanisms, including the induction of anti-inflammatory CD4⁺ T cell responses. Foxp3⁺ regulatory T cells (Foxp3+ Tregs) and Type 1 regulatory T cells (Tr1) play a major role in promoting tolerance, as both cell types can produce the anti-inflammatory cytokine IL-10. In addition to these regulatory T cells, effector T cell subsets, such as Th17 cells, also adopt anti-inflammatory functions within the intestine in response to the microbiota. This process of anti-inflammatory CD4+ T cell induction is heavily influenced by the microbiota and their metabolites. Microbial metabolites affect intestinal epithelial cells, promoting the secretion of anti-inflammatory mediators that create a tolerogenic environment. They also modulate intestinal dendritic cells (DCs) and macrophages, inducing a tolerogenic state, and can interact directly with T cells to drive anti-inflammatory CD4⁺ T cell functionality. The disrupted balance of these signals may result in chronic inflammation, with broader implications for systemic health. In this review, we highlight the intricate interplays between commensal microorganisms and the immune system in the gut. We discuss how the microbiota influences the differentiation of commensal-specific anti-inflammatory CD4⁺ T cells, such as Foxp3⁺ Tregs, Tr1 cells, and Th17 cells, and explore the mechanisms through which microbial metabolites modulate these processes. We further discuss the innate signals that prime and commit these cells to an anti-inflammatory fate.
Collapse
Affiliation(s)
- Madeline Edwards
- Department of Microbiology and Immunology, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, 10032, USA
| | - Leonie Brockmann
- Department of Systems Biology, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, 10032, USA.
- Human Biology-Microbiome-Quantum Research Center (WPI-Bio2Q), Keio University, Tokyo, 108-8345, Japan.
| |
Collapse
|
3
|
Yu F, Zhu C, Wu W. Senile Osteoarthritis Regulated by the Gut Microbiota: From Mechanisms to Treatments. Int J Mol Sci 2025; 26:1505. [PMID: 40003971 PMCID: PMC11855920 DOI: 10.3390/ijms26041505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2024] [Revised: 01/19/2025] [Accepted: 01/26/2025] [Indexed: 02/27/2025] Open
Abstract
Osteoarthritis (OA) is a chronic, progressive degenerative joint disease that affects the entire synovial joint, leading to the progressive degeneration of articular cartilage. It seriously affects the quality of life and global disability of patients. OA is affected by a variety of factors; the most significant risk factor for OA is age. As individuals age, the risk and severity of OA increase due to the exacerbation of cartilage degeneration and wear and tear. In recent years, research has indicated that the gut microbiota may play a significant role in the aging and OA processes. It is anticipated that regulating the gut microbiota may offer novel approaches to the treatment of OA. The objective of this paper is to examine the relationship between the gut microbiota and senile OA, to investigate the potential mechanisms involved. This review also summarizes the therapeutic strategies related to gut flora in OA management, such as prebiotics and probiotics, diet, exercise, traditional Chinese medicine (TCM) modification, and fecal microbiota transplantation (FMT), highlighting the potential clinical value of gut flora and elucidating the current challenges. The foundation for future research directions is established through the summarization of current research progress.
Collapse
Affiliation(s)
- Fan Yu
- School of Exercise and Health, Shanghai University of Sports, Shanghai 200438, China; (F.Y.); (C.Z.)
| | - Chenyu Zhu
- School of Exercise and Health, Shanghai University of Sports, Shanghai 200438, China; (F.Y.); (C.Z.)
| | - Wei Wu
- School of Athletic Performance, Shanghai University of Sports, Shanghai 200438, China
| |
Collapse
|
4
|
Vashishth S, Ambasta RK, Kumar P. Deciphering the microbial map and its implications in the therapeutics of neurodegenerative disorder. Ageing Res Rev 2024; 100:102466. [PMID: 39197710 DOI: 10.1016/j.arr.2024.102466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 08/19/2024] [Accepted: 08/19/2024] [Indexed: 09/01/2024]
Abstract
Every facet of biological anthropology, including development, ageing, diseases, and even health maintenance, is influenced by gut microbiota's significant genetic and metabolic capabilities. With current advancements in sequencing technology and with new culture-independent approaches, researchers can surpass older correlative studies and develop mechanism-based studies on microbiome-host interactions. The microbiota-gut-brain axis (MGBA) regulates glial functioning, making it a possible target for the improvement of development and advancement of treatments for neurodegenerative diseases (NDDs). The gut-brain axis (GBA) is accountable for the reciprocal communication between the gastrointestinal and central nervous system, which plays an essential role in the regulation of physiological processes like controlling hunger, metabolism, and various gastrointestinal functions. Lately, studies have discovered the function of the gut microbiome for brain health-different microbiota through different pathways such as immunological, neurological and metabolic pathways. Additionally, we review the involvement of the neurotransmitters and the gut hormones related to gut microbiota. We also explore the MGBA in neurodegenerative disorders by focusing on metabolites. Further, targeting the blood-brain barrier (BBB), intestinal barrier, meninges, and peripheral immune system is investigated. Lastly, we discuss the therapeutics approach and evaluate the pre-clinical and clinical trial data regarding using prebiotics, probiotics, paraprobiotics, fecal microbiota transplantation, personalised medicine, and natural food bioactive in NDDs. A comprehensive study of the GBA will felicitate the creation of efficient therapeutic approaches for treating different NDDs.
Collapse
Affiliation(s)
- Shrutikirti Vashishth
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, Delhi Technological University (Formerly DCE), Delhi 110042, India
| | - Rashmi K Ambasta
- Department of Medicine, School of Medicine, VUMC, Vanderbilt University, TN, USA
| | - Pravir Kumar
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, Delhi Technological University (Formerly DCE), Delhi 110042, India.
| |
Collapse
|
5
|
Fanizzi F, D'Amico F, Zanotelli Bombassaro I, Zilli A, Furfaro F, Parigi TL, Cicerone C, Fiorino G, Peyrin-Biroulet L, Danese S, Allocca M. The Role of Fecal Microbiota Transplantation in IBD. Microorganisms 2024; 12:1755. [PMID: 39338430 PMCID: PMC11433743 DOI: 10.3390/microorganisms12091755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 08/19/2024] [Accepted: 08/22/2024] [Indexed: 09/30/2024] Open
Abstract
Gut microbiota dysbiosis has a critical role in the pathogenesis of inflammatory bowel diseases, prompting the exploration of novel therapeutic approaches like fecal microbiota transplantation, which involves the transfer of fecal microbiota from a healthy donor to a recipient with the aim of restoring a balanced microbial community and attenuating inflammation. Fecal microbiota transplantation may exert beneficial effects in inflammatory bowel disease through modulation of immune responses, restoration of mucosal barrier integrity, and alteration of microbial metabolites. It could alter disease course and prevent flares, although long-term durability and safety data are lacking. This review provides a summary of current evidence on fecal microbiota transplantation in inflammatory bowel disease management, focusing on its challenges, such as variability in donor selection criteria, standardization of transplant protocols, and long-term outcomes post-transplantation.
Collapse
Affiliation(s)
- Fabrizio Fanizzi
- Department of Gastroenterology and Endoscopy, IRCCS Ospedale San Raffaele, Vita-Salute San Raffaele University, 20132 Milan, Italy
| | - Ferdinando D'Amico
- Department of Gastroenterology and Endoscopy, IRCCS Ospedale San Raffaele, Vita-Salute San Raffaele University, 20132 Milan, Italy
| | - Isadora Zanotelli Bombassaro
- Department of Gastroenterology and Endoscopy, Santa Casa de Misericordia de Porto Alagre, Porto Alegre 90020-090, Brazil
| | - Alessandra Zilli
- Department of Gastroenterology and Endoscopy, IRCCS Ospedale San Raffaele, Vita-Salute San Raffaele University, 20132 Milan, Italy
| | - Federica Furfaro
- Department of Gastroenterology and Endoscopy, IRCCS Ospedale San Raffaele, Vita-Salute San Raffaele University, 20132 Milan, Italy
| | - Tommaso Lorenzo Parigi
- Department of Gastroenterology and Endoscopy, IRCCS Ospedale San Raffaele, Vita-Salute San Raffaele University, 20132 Milan, Italy
| | - Clelia Cicerone
- Department of Gastroenterology and Endoscopy, IRCCS Ospedale San Raffaele, Vita-Salute San Raffaele University, 20132 Milan, Italy
| | - Gionata Fiorino
- Department of Gastroenterology and Digestive Endoscopy, San Camillo-Forlanini Hospital, 00152 Rome, Italy
| | - Laurent Peyrin-Biroulet
- Department of Gastroenterology, Nancy University Hospital, F-54500 Vandœuvre-lès-Nancy, France
- INSERM, Nutrition-Genetics and Exposure to Environmental Risks Research Unit (NGERE), University of Lorraine, F-54000 Nancy, France
- INFINY Institute, Nancy University Hospital, F-54500 Vandœuvre-lès-Nancy, France
- Fédération Hospitalo-Universitaire CARE, Nancy University Hospital, F-54500 Vandœuvre-lès-Nancy, France
- Groupe Hospitalier Privé Ambroise Paré-Hartmann, Paris IBD Center, F-92200 Neuilly sur Seine, France
- Division of Gastroenterology and Hepatology, McGill University Health Centre, Montreal, QC H4A 3J1, Canada
| | - Silvio Danese
- Department of Gastroenterology and Endoscopy, IRCCS Ospedale San Raffaele, Vita-Salute San Raffaele University, 20132 Milan, Italy
| | - Mariangela Allocca
- Department of Gastroenterology and Endoscopy, IRCCS Ospedale San Raffaele, Vita-Salute San Raffaele University, 20132 Milan, Italy
| |
Collapse
|
6
|
Olson S, Welton L, Jahansouz C. Perioperative Considerations for the Surgical Treatment of Crohn's Disease with Discussion on Surgical Antibiotics Practices and Impact on the Gut Microbiome. Antibiotics (Basel) 2024; 13:317. [PMID: 38666993 PMCID: PMC11047551 DOI: 10.3390/antibiotics13040317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 03/22/2024] [Accepted: 03/28/2024] [Indexed: 04/29/2024] Open
Abstract
Crohn's disease, a chronic inflammatory process of the gastrointestinal tract defined by flares and periods of remission, is increasing in incidence. Despite advances in multimodal medical therapy, disease progression often necessitates multiple operations with high morbidity. The inability to treat Crohn's disease successfully is likely in part because the etiopathogenesis is not completely understood; however, recent research suggests the gut microbiome plays a critical role. How traditional perioperative management, including bowel preparation and preoperative antibiotics, further changes the microbiome and affects outcomes is not well described, especially in Crohn's patients, who are unique given their immunosuppression and baseline dysbiosis. This paper aims to outline current knowledge regarding perioperative management of Crohn's disease, the evolving role of gut dysbiosis, and how the microbiome can guide perioperative considerations with special attention to perioperative antibiotics as well as treatment of Mycobacterium avium subspecies paratuberculosis. In conclusion, dysbiosis is common in Crohn's patients and may be exacerbated by malnutrition, steroids, narcotic use, diarrhea, and perioperative antibiotics. Dysbiosis is also a major risk factor for anastomotic leak, and special consideration should be given to limiting factors that further perturb the gut microbiota in the perioperative period.
Collapse
Affiliation(s)
- Shelbi Olson
- Department of Surgery, University of Minnesota, Minneapolis, MN 55455, USA; (S.O.); (L.W.)
| | - Lindsay Welton
- Department of Surgery, University of Minnesota, Minneapolis, MN 55455, USA; (S.O.); (L.W.)
| | - Cyrus Jahansouz
- Division of Colon and Rectal Surgery, University of Minnesota, Minneapolis, MN 55455, USA
| |
Collapse
|
7
|
Porcari S, Maida M, Bibbò S, McIlroy J, Ianiro G, Cammarota G. Fecal Microbiota Transplantation as Emerging Treatment in European Countries 2.0. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1435:85-99. [PMID: 38175472 DOI: 10.1007/978-3-031-42108-2_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
Clostridioides difficile infection (CDI) is one of the most common healthcare-associated infections and one of the leading causes of morbidity and mortality in hospitalized patients in the world. Although several antibiotics effectively treat CDI, some individuals may not respond to these drugs and may be cured by transplanting stool from healthy donors. FMT has demonstrated extraordinary cure rates for the cure of CDI recurrences.Moreover, FMT has also been investigated in other disorders associated with the alteration of gut microbiota, such as inflammatory bowel disease (IBD), where the alterations of the gut microbiota ecology have been theorized to play a causative role. Although FMT is currently not recommended to cure IBD patients in clinical practice, several studies have been recently carried out with the ultimate goal to search new therapeutic options to patients.This review summarizes data on the use of FMT for the treatment of both CDI and IBD, with a special attention to highlight studies conducted in European countries.
Collapse
Affiliation(s)
- Serena Porcari
- Department of Medical and Surgical Sciences, Digestive Disease Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Marcello Maida
- Gastroenterology and Endoscopy Unit, S. Elia-Raimondi Hospital, Caltanissetta, Italy
| | - Stefano Bibbò
- Department of Medical and Surgical Sciences, Digestive Disease Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy
| | - James McIlroy
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen, UK
| | - Gianluca Ianiro
- Department of Medical and Surgical Sciences, Digestive Disease Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Giovanni Cammarota
- Department of Medical and Surgical Sciences, Digestive Disease Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy.
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy.
| |
Collapse
|
8
|
Boicean A, Bratu D, Fleaca SR, Vasile G, Shelly L, Birsan S, Bacila C, Hasegan A. Exploring the Potential of Fecal Microbiota Transplantation as a Therapy in Tuberculosis and Inflammatory Bowel Disease. Pathogens 2023; 12:1149. [PMID: 37764957 PMCID: PMC10535282 DOI: 10.3390/pathogens12091149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Revised: 09/03/2023] [Accepted: 09/07/2023] [Indexed: 09/29/2023] Open
Abstract
This review explores the potential benefits of fecal microbiota transplantation (FMT) as an adjunct treatment in tuberculosis (TB), drawing parallels from its efficacy in inflammatory bowel disease (IBD). FMT has shown promise in restoring the gut microbial balance and modulating immune responses in IBD patients. Considering the similarities in immunomodulation and dysbiosis between IBD and TB, this review hypothesizes that FMT may offer therapeutic benefits as an adjunct therapy in TB. Methods: We conducted a systematic review of the existing literature on FMT in IBD and TB, highlighting the mechanisms and potential implications of FMT in the therapeutic management of both conditions. The findings contribute to understanding FMT's potential role in TB treatment and underscore the necessity for future research in this direction to fully leverage its clinical applications. Conclusion: The integration of FMT into the comprehensive management of TB could potentially enhance treatment outcomes, reduce drug resistance, and mitigate the side effects of conventional therapies. Future research endeavors should focus on well-designed clinical trials to develop guidelines concerning the safety and short- and long-term benefits of FMT in TB patients, as well as to assess potential risks.
Collapse
Affiliation(s)
- Adrian Boicean
- Faculty of Medicine, Lucian Blaga University of Sibiu, 550169 Sibiu, Romania; (A.B.); (S.R.F.); (S.B.); (C.B.); (A.H.)
| | - Dan Bratu
- Faculty of Medicine, Lucian Blaga University of Sibiu, 550169 Sibiu, Romania; (A.B.); (S.R.F.); (S.B.); (C.B.); (A.H.)
| | - Sorin Radu Fleaca
- Faculty of Medicine, Lucian Blaga University of Sibiu, 550169 Sibiu, Romania; (A.B.); (S.R.F.); (S.B.); (C.B.); (A.H.)
| | - Gligor Vasile
- County Clinical Emergency Hospital of Sibiu, 550245 Sibiu, Romania; (G.V.); (L.S.)
| | - Leeb Shelly
- County Clinical Emergency Hospital of Sibiu, 550245 Sibiu, Romania; (G.V.); (L.S.)
| | - Sabrina Birsan
- Faculty of Medicine, Lucian Blaga University of Sibiu, 550169 Sibiu, Romania; (A.B.); (S.R.F.); (S.B.); (C.B.); (A.H.)
| | - Ciprian Bacila
- Faculty of Medicine, Lucian Blaga University of Sibiu, 550169 Sibiu, Romania; (A.B.); (S.R.F.); (S.B.); (C.B.); (A.H.)
| | - Adrian Hasegan
- Faculty of Medicine, Lucian Blaga University of Sibiu, 550169 Sibiu, Romania; (A.B.); (S.R.F.); (S.B.); (C.B.); (A.H.)
| |
Collapse
|
9
|
Mendes I, Vale N. How Can the Microbiome Induce Carcinogenesis and Modulate Drug Resistance in Cancer Therapy? Int J Mol Sci 2023; 24:11855. [PMID: 37511612 PMCID: PMC10380870 DOI: 10.3390/ijms241411855] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Revised: 07/18/2023] [Accepted: 07/21/2023] [Indexed: 07/30/2023] Open
Abstract
Over the years, cancer has been affecting the lives of many people globally and it has become one of the most studied diseases. Despite the efforts to understand the cell mechanisms behind this complex disease, not every patient seems to respond to targeted therapies or immunotherapies. Drug resistance in cancer is one of the limiting factors contributing to unsuccessful therapies; therefore, understanding how cancer cells acquire this resistance is essential to help cure individuals affected by cancer. Recently, the altered microbiome was observed to be an important hallmark of cancer and therefore it represents a promising topic of cancer research. Our review aims to provide a global perspective of some cancer hallmarks, for instance how genetic and epigenetic modifications may be caused by an altered human microbiome. We also provide information on how an altered human microbiome can lead to cancer development as well as how the microbiome can influence drug resistance and ultimately targeted therapies. This may be useful to develop alternatives for cancer treatment, i.e., future personalized medicine that can help in cases where traditional cancer treatment is unsuccessful.
Collapse
Affiliation(s)
- Inês Mendes
- OncoPharma Research Group, Center for Health Technology and Services Research (CINTESIS), Rua Doutor Plácido da Costa, 4200-450 Porto, Portugal;
- CINTESIS@RISE, Faculty of Medicine, University of Porto, Alameda Professor Hernâni Monteiro, 4200-319 Porto, Portugal
- School of Life and Environmental Sciences, University of Trás-os-Montes and Alto Douro (UTAD), Edifício de Geociências, 5000-801 Vila Real, Portugal
| | - Nuno Vale
- OncoPharma Research Group, Center for Health Technology and Services Research (CINTESIS), Rua Doutor Plácido da Costa, 4200-450 Porto, Portugal;
- CINTESIS@RISE, Faculty of Medicine, University of Porto, Alameda Professor Hernâni Monteiro, 4200-319 Porto, Portugal
- Department of Community Medicine, Information and Health Decision Sciences (MEDCIDS), Faculty of Medicine, University of Porto, Rua Doutor Plácido da Costa, 4200-450 Porto, Portugal
| |
Collapse
|
10
|
Bicknell B, Liebert A, Borody T, Herkes G, McLachlan C, Kiat H. Neurodegenerative and Neurodevelopmental Diseases and the Gut-Brain Axis: The Potential of Therapeutic Targeting of the Microbiome. Int J Mol Sci 2023; 24:9577. [PMID: 37298527 PMCID: PMC10253993 DOI: 10.3390/ijms24119577] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 04/28/2023] [Accepted: 05/23/2023] [Indexed: 06/12/2023] Open
Abstract
The human gut microbiome contains the largest number of bacteria in the body and has the potential to greatly influence metabolism, not only locally but also systemically. There is an established link between a healthy, balanced, and diverse microbiome and overall health. When the gut microbiome becomes unbalanced (dysbiosis) through dietary changes, medication use, lifestyle choices, environmental factors, and ageing, this has a profound effect on our health and is linked to many diseases, including lifestyle diseases, metabolic diseases, inflammatory diseases, and neurological diseases. While this link in humans is largely an association of dysbiosis with disease, in animal models, a causative link can be demonstrated. The link between the gut and the brain is particularly important in maintaining brain health, with a strong association between dysbiosis in the gut and neurodegenerative and neurodevelopmental diseases. This link suggests not only that the gut microbiota composition can be used to make an early diagnosis of neurodegenerative and neurodevelopmental diseases but also that modifying the gut microbiome to influence the microbiome-gut-brain axis might present a therapeutic target for diseases that have proved intractable, with the aim of altering the trajectory of neurodegenerative and neurodevelopmental diseases such as Alzheimer's disease, Parkinson's disease, multiple sclerosis, autism spectrum disorder, and attention-deficit hyperactivity disorder, among others. There is also a microbiome-gut-brain link to other potentially reversible neurological diseases, such as migraine, post-operative cognitive dysfunction, and long COVID, which might be considered models of therapy for neurodegenerative disease. The role of traditional methods in altering the microbiome, as well as newer, more novel treatments such as faecal microbiome transplants and photobiomodulation, are discussed.
Collapse
Affiliation(s)
- Brian Bicknell
- NICM Health Research Institute, University of Western Sydney, Westmead, NSW 2145, Australia; (A.L.); (H.K.)
| | - Ann Liebert
- NICM Health Research Institute, University of Western Sydney, Westmead, NSW 2145, Australia; (A.L.); (H.K.)
- Faculty of Medicine and Health, University of Sydney, Camperdown, NSW 2006, Australia
- Department of Governance and Research, Sydney Adventist Hospital, Wahroonga, NSW 2076, Australia;
| | - Thomas Borody
- Centre for Digestive Diseases, Five Dock, NSW 2046, Australia;
| | - Geoffrey Herkes
- Department of Governance and Research, Sydney Adventist Hospital, Wahroonga, NSW 2076, Australia;
| | - Craig McLachlan
- Centre for Healthy Futures, Torrens University Australia, Ultimo, NSW 2007, Australia;
| | - Hosen Kiat
- NICM Health Research Institute, University of Western Sydney, Westmead, NSW 2145, Australia; (A.L.); (H.K.)
- Centre for Healthy Futures, Torrens University Australia, Ultimo, NSW 2007, Australia;
- Macquarie Medical School, Macquarie University, Macquarie Park, NSW 2109, Australia
- ANU College of Health and Medicine, Australian National University, Canberra, ACT 2601, Australia
| |
Collapse
|
11
|
Boicean A, Bratu D, Bacila C, Tanasescu C, Fleacă RS, Mohor CI, Comaniciu A, Băluță T, Roman MD, Chicea R, Cristian AN, Hasegan A, Birsan S, Dura H, Mohor CI. Therapeutic Perspectives for Microbiota Transplantation in Digestive Diseases and Neoplasia-A Literature Review. Pathogens 2023; 12:766. [PMID: 37375456 PMCID: PMC10302701 DOI: 10.3390/pathogens12060766] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2023] [Revised: 05/23/2023] [Accepted: 05/25/2023] [Indexed: 06/29/2023] Open
Abstract
In a mutually beneficial connection with its host, the gut microbiota affects the host's nutrition, immunity, and metabolism. An increasing number of studies have shown links between certain types of disease and gut dysbiosis or specific microorganisms. Fecal microbiota transplantation (FMT) is strongly advised for the treatment of recurrent or resistant Clostridium difficile infection (CDI) due to its outstanding clinical effectiveness against CDI. The therapeutic potential of FMT for other disorders, particularly inflammatory bowel diseases and malignancies, is currently gaining more and more attention. We summarized the most recent preclinical and clinical evidence to show the promise of FMT in the management of cancer as well as complications related to cancer treatment after reviewing the most recent research on the gut microbiota and its relationship to cancer.
Collapse
Affiliation(s)
- Adrian Boicean
- County Clinical Emergency Hospital of Sibiu, 550245 Sibiu, Romania; (A.B.); (C.B.); (C.T.); (R.S.F.); (C.I.M.); (A.C.); (T.B.); (M.D.R.); (R.C.); (A.N.C.); (A.H.); (S.B.); (H.D.); (C.I.M.)
- Faculty of Medicine, Lucian Blaga University of Sibiu, 550169 Sibiu, Romania
| | - Dan Bratu
- County Clinical Emergency Hospital of Sibiu, 550245 Sibiu, Romania; (A.B.); (C.B.); (C.T.); (R.S.F.); (C.I.M.); (A.C.); (T.B.); (M.D.R.); (R.C.); (A.N.C.); (A.H.); (S.B.); (H.D.); (C.I.M.)
- Faculty of Medicine, Lucian Blaga University of Sibiu, 550169 Sibiu, Romania
| | - Ciprian Bacila
- County Clinical Emergency Hospital of Sibiu, 550245 Sibiu, Romania; (A.B.); (C.B.); (C.T.); (R.S.F.); (C.I.M.); (A.C.); (T.B.); (M.D.R.); (R.C.); (A.N.C.); (A.H.); (S.B.); (H.D.); (C.I.M.)
| | - Ciprian Tanasescu
- County Clinical Emergency Hospital of Sibiu, 550245 Sibiu, Romania; (A.B.); (C.B.); (C.T.); (R.S.F.); (C.I.M.); (A.C.); (T.B.); (M.D.R.); (R.C.); (A.N.C.); (A.H.); (S.B.); (H.D.); (C.I.M.)
- Faculty of Medicine, Lucian Blaga University of Sibiu, 550169 Sibiu, Romania
| | - Radu Sorin Fleacă
- County Clinical Emergency Hospital of Sibiu, 550245 Sibiu, Romania; (A.B.); (C.B.); (C.T.); (R.S.F.); (C.I.M.); (A.C.); (T.B.); (M.D.R.); (R.C.); (A.N.C.); (A.H.); (S.B.); (H.D.); (C.I.M.)
- Faculty of Medicine, Lucian Blaga University of Sibiu, 550169 Sibiu, Romania
| | - Calin Ilie Mohor
- County Clinical Emergency Hospital of Sibiu, 550245 Sibiu, Romania; (A.B.); (C.B.); (C.T.); (R.S.F.); (C.I.M.); (A.C.); (T.B.); (M.D.R.); (R.C.); (A.N.C.); (A.H.); (S.B.); (H.D.); (C.I.M.)
- Faculty of Medicine, Lucian Blaga University of Sibiu, 550169 Sibiu, Romania
| | - Andra Comaniciu
- County Clinical Emergency Hospital of Sibiu, 550245 Sibiu, Romania; (A.B.); (C.B.); (C.T.); (R.S.F.); (C.I.M.); (A.C.); (T.B.); (M.D.R.); (R.C.); (A.N.C.); (A.H.); (S.B.); (H.D.); (C.I.M.)
| | - Teodora Băluță
- County Clinical Emergency Hospital of Sibiu, 550245 Sibiu, Romania; (A.B.); (C.B.); (C.T.); (R.S.F.); (C.I.M.); (A.C.); (T.B.); (M.D.R.); (R.C.); (A.N.C.); (A.H.); (S.B.); (H.D.); (C.I.M.)
| | - Mihai Dan Roman
- County Clinical Emergency Hospital of Sibiu, 550245 Sibiu, Romania; (A.B.); (C.B.); (C.T.); (R.S.F.); (C.I.M.); (A.C.); (T.B.); (M.D.R.); (R.C.); (A.N.C.); (A.H.); (S.B.); (H.D.); (C.I.M.)
- Faculty of Medicine, Lucian Blaga University of Sibiu, 550169 Sibiu, Romania
| | - Radu Chicea
- County Clinical Emergency Hospital of Sibiu, 550245 Sibiu, Romania; (A.B.); (C.B.); (C.T.); (R.S.F.); (C.I.M.); (A.C.); (T.B.); (M.D.R.); (R.C.); (A.N.C.); (A.H.); (S.B.); (H.D.); (C.I.M.)
- Faculty of Medicine, Lucian Blaga University of Sibiu, 550169 Sibiu, Romania
| | - Adrian Nicolae Cristian
- County Clinical Emergency Hospital of Sibiu, 550245 Sibiu, Romania; (A.B.); (C.B.); (C.T.); (R.S.F.); (C.I.M.); (A.C.); (T.B.); (M.D.R.); (R.C.); (A.N.C.); (A.H.); (S.B.); (H.D.); (C.I.M.)
- Faculty of Medicine, Lucian Blaga University of Sibiu, 550169 Sibiu, Romania
| | - Adrian Hasegan
- County Clinical Emergency Hospital of Sibiu, 550245 Sibiu, Romania; (A.B.); (C.B.); (C.T.); (R.S.F.); (C.I.M.); (A.C.); (T.B.); (M.D.R.); (R.C.); (A.N.C.); (A.H.); (S.B.); (H.D.); (C.I.M.)
- Faculty of Medicine, Lucian Blaga University of Sibiu, 550169 Sibiu, Romania
| | - Sabrina Birsan
- County Clinical Emergency Hospital of Sibiu, 550245 Sibiu, Romania; (A.B.); (C.B.); (C.T.); (R.S.F.); (C.I.M.); (A.C.); (T.B.); (M.D.R.); (R.C.); (A.N.C.); (A.H.); (S.B.); (H.D.); (C.I.M.)
- Faculty of Medicine, Lucian Blaga University of Sibiu, 550169 Sibiu, Romania
| | - Horațiu Dura
- County Clinical Emergency Hospital of Sibiu, 550245 Sibiu, Romania; (A.B.); (C.B.); (C.T.); (R.S.F.); (C.I.M.); (A.C.); (T.B.); (M.D.R.); (R.C.); (A.N.C.); (A.H.); (S.B.); (H.D.); (C.I.M.)
- Faculty of Medicine, Lucian Blaga University of Sibiu, 550169 Sibiu, Romania
| | - Cosmin Ioan Mohor
- County Clinical Emergency Hospital of Sibiu, 550245 Sibiu, Romania; (A.B.); (C.B.); (C.T.); (R.S.F.); (C.I.M.); (A.C.); (T.B.); (M.D.R.); (R.C.); (A.N.C.); (A.H.); (S.B.); (H.D.); (C.I.M.)
- Faculty of Medicine, Lucian Blaga University of Sibiu, 550169 Sibiu, Romania
| |
Collapse
|
12
|
Kurt F, Leventhal GE, Spalinger MR, Anthamatten L, Rogalla von Bieberstein P, Menzi C, Reichlin M, Meola M, Rosenthal F, Rogler G, Lacroix C, de Wouters T. Co-cultivation is a powerful approach to produce a robust functionally designed synthetic consortium as a live biotherapeutic product (LBP). Gut Microbes 2023; 15:2177486. [PMID: 36794804 PMCID: PMC9980632 DOI: 10.1080/19490976.2023.2177486] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 01/31/2023] [Indexed: 02/17/2023] Open
Abstract
The success of fecal microbiota transplants (FMT) has provided the necessary proof-of-concept for microbiome therapeutics. Yet, feces-based therapies have many associated risks and uncertainties, and hence defined microbial consortia that modify the microbiome in a targeted manner have emerged as a promising safer alternative to FMT. The development of such live biotherapeutic products has important challenges, including the selection of appropriate strains and the controlled production of the consortia at scale. Here, we report on an ecology- and biotechnology-based approach to microbial consortium construction that overcomes these issues. We selected nine strains that form a consortium to emulate the central metabolic pathways of carbohydrate fermentation in the healthy human gut microbiota. Continuous co-culturing of the bacteria produces a stable and reproducible consortium whose growth and metabolic activity are distinct from an equivalent mix of individually cultured strains. Further, we showed that our function-based consortium is as effective as FMT in counteracting dysbiosis in a dextran sodium sulfate mouse model of acute colitis, while an equivalent mix of strains failed to match FMT. Finally, we showed robustness and general applicability of our approach by designing and producing additional stable consortia of controlled composition. We propose that combining a bottom-up functional design with continuous co-cultivation is a powerful strategy to produce robust functionally designed synthetic consortia for therapeutic use.
Collapse
Affiliation(s)
- Fabienne Kurt
- PharmaBiome AG, Schlieren, Switzerland
- Laboratory of Food Biotechnology, Institute of Food, Nutrition and Health, ETH Zurich, Zurich, Switzerland
| | | | - Marianne Rebecca Spalinger
- Department of Gastroenterology and Hepatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Laura Anthamatten
- PharmaBiome AG, Schlieren, Switzerland
- Laboratory of Food Biotechnology, Institute of Food, Nutrition and Health, ETH Zurich, Zurich, Switzerland
| | | | | | | | | | | | - Gerhard Rogler
- Department of Gastroenterology and Hepatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Christophe Lacroix
- Laboratory of Food Biotechnology, Institute of Food, Nutrition and Health, ETH Zurich, Zurich, Switzerland
| | | |
Collapse
|
13
|
Wang S, Cui J, Jiang S, Zheng C, Zhao J, Zhang H, Zhai Q. Early life gut microbiota: Consequences for health and opportunities for prevention. Crit Rev Food Sci Nutr 2022; 64:5793-5817. [PMID: 36537331 DOI: 10.1080/10408398.2022.2158451] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The gut microbiota influences many aspects of the host, including immune system maturation, nutrient absorption and metabolism, and protection from pathogens. Increasing evidences from cohort and animal studies indicate that changes in the gut microbiota early in life increases the risk of developing specific diseases early and later in life. Therefore, it is becoming increasingly important to identify specific disease prevention or therapeutic solutions targeting the gut microbiota, especially during infancy, which is the window of the human gut microbiota establishment process. In this review, we provide an overview of current knowledge concerning the relationship between disturbances in the gut microbiota early in life and health consequences later in life (e.g., necrotizing enterocolitis, celiac disease, asthma, allergies, autism spectrum disorders, overweight/obesity, diabetes and growth retardation), with a focus on changes in the gut microbiota prior to disease onset. In addition, we summarize and discuss potential microbiota-based interventions early in life (e.g., diet adjustments, probiotics, prebiotics, fecal microbiota transplantation, environmental changes) to promote health or prevent the development of specific diseases. This knowledge should aid the understanding of early life microbiology and inform the development of prediction and prevention measures for short- and long-term health disorders based on the gut microbiota.
Collapse
Affiliation(s)
- Shumin Wang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
| | - Jingjing Cui
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Shilong Jiang
- Nutrition and Metabolism Research Division, Innovation Center, Heilongjiang Feihe Dairy Co., Ltd, Beijing, China
- PKUHSC-China Feihe Joint Research Institute of Nutrition and Healthy Lifespan Development, Beijing, China
| | - Chengdong Zheng
- Nutrition and Metabolism Research Division, Innovation Center, Heilongjiang Feihe Dairy Co., Ltd, Beijing, China
- PKUHSC-China Feihe Joint Research Institute of Nutrition and Healthy Lifespan Development, Beijing, China
| | - Jianxin Zhao
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
| | - Heng Zhang
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
- Department of Child Health Care, Wuxi Maternity and Child Health Care Hospital, Wuxi, Jiangsu, China
| | - Qixiao Zhai
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
| |
Collapse
|
14
|
Dong Y, Xu T, Xiao G, Hu Z, Chen J. Opportunities and challenges for synthetic biology in the therapy of inflammatory bowel disease. Front Bioeng Biotechnol 2022; 10:909591. [PMID: 36032720 PMCID: PMC9399643 DOI: 10.3389/fbioe.2022.909591] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 07/15/2022] [Indexed: 11/13/2022] Open
Abstract
Inflammatory bowel disease (IBD) is a complex, chronic intestinal inflammatory disorder that primarily includes Crohn’s disease (CD) and ulcerative colitis (UC). Although traditional antibiotics and immunosuppressants are known as the most effective and commonly used treatments, some limitations may be expected, such as limited efficacy in a small number of patients and gut flora disruption. A great many research studies have been done with respect to the etiology of IBD, while the composition of the gut microbiota is suggested as one of the most influential factors. Along with the development of synthetic biology and the continuing clarification of IBD etiology, broader prospects for novel approaches to IBD therapy could be obtained. This study presents an overview of the currently existing treatment options and possible therapeutic targets at the preclinical stage with respect to microbial synthesis technology in biological therapy. This study is highly correlated to the following topics: microbiota-derived metabolites, microRNAs, cell therapy, calreticulin, live biotherapeutic products (LBP), fecal microbiota transplantation (FMT), bacteriophages, engineered bacteria, and their functional secreted synthetic products for IBD medical implementation. Considering microorganisms as the main therapeutic component, as a result, the related clinical trial stability, effectiveness, and safety analysis may be the major challenges for upcoming research. This article strives to provide pharmaceutical researchers and developers with the most up-to-date information for adjuvant medicinal therapies based on synthetic biology.
Collapse
Affiliation(s)
- Yumeng Dong
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
- Suzhou U-Synbio Co., Ltd., Suzhou, China
| | - Tiangang Xu
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| | - Guozheng Xiao
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| | - Ziyan Hu
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| | - Jingyu Chen
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
- *Correspondence: Jingyu Chen,
| |
Collapse
|
15
|
Rakotonirina A, Galperine T, Allémann E. Fecal microbiota transplantation: a review on current formulations in Clostridioides difficile infection and future outlooks. Expert Opin Biol Ther 2022; 22:929-944. [PMID: 35763604 DOI: 10.1080/14712598.2022.2095901] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION The role of the gut microbiota in health and the pathogenesis of several diseases has been highlighted in recent years. Even though the precise mechanisms involving the microbiome in these ailments are still unclear, microbiota-modulating therapies have been developed. Fecal microbiota transplantation (FMT) has shown significant results against Clostridioides difficile infection (CDI), and its potential has been investigated for other diseases. Unfortunately, the technical aspects of the treatment make it difficult to implement. Pharmaceutical technology approaches to encapsulate microorganisms could play an important role in providing this treatment and render the treatment modalities easier to handle. AREAS COVERED After an overview of CDI, this narrative review aims to discuss the current formulations for FMT and specifically addresses the technical aspects of the treatment. This review also distinguishes itself by focusing on the hurdles and emphasizing the possible improvements using pharmaceutical technologies. EXPERT OPINION FMT is an efficient treatment for recurrent CDI. However, its standardization is overlooked. The approach of industrial and hospital preparations of FMT are different, but both show promise in their respective methodologies. Novel FMT formulations could enable further research on dysbiotic diseases in the future.
Collapse
Affiliation(s)
- Adèle Rakotonirina
- School of Pharmaceutical Sciences, University of Geneva, Geneva, Switzerland.,Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, Geneva, Switzerland
| | - Tatiana Galperine
- Infectious Diseases Service, Department of Medicine, University Hospital and University of Lausanne, Lausanne, Switzerland.,French Group of Faecal Microbiota Transplantation
| | - Eric Allémann
- School of Pharmaceutical Sciences, University of Geneva, Geneva, Switzerland.,Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, Geneva, Switzerland
| |
Collapse
|
16
|
Fecal miR-142a-3p from dextran sulfate sodium-challenge recovered mice prevents colitis by promoting the growth of Lactobacillus reuteri. Mol Ther 2022; 30:388-399. [PMID: 34450255 PMCID: PMC8753372 DOI: 10.1016/j.ymthe.2021.08.025] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Revised: 07/07/2021] [Accepted: 07/29/2021] [Indexed: 02/07/2023] Open
Abstract
Feces are enriched with microRNAs (miRNAs) that shape the gut microbiota. These miRNAs are differentially expressed in the feces of healthy and diseased subjects. However, whether fecal miRNAs in subjects with inflammatory bowel diseases are involved in regulating microbiota composition and whether they have any beneficial effects remains unknown. Here, we studied the fecal microbiome composition and miRNA abundance in mice with dextran sulfate sodium (DSS)-induced colitis and mice at the recovery phase to explore different miRNAs expressed, their relations with microbial abundance, and their effects on colitis. We found that miR-142a-3p expression was significantly increased in the feces of mice recovered from colitis and that it could alleviate disease symptoms in mice treated with DSS in a microbiome-dependent manner. Specifically, miR-142a-3p promoted the growth of Lactobacillus reuteri, which had a high abundance in the feces of mice recovered from colitis, by regulating transcripts of polA and locus tag LREU_RS03575. Moreover, L. reuteri, as well as its metabolite reuterin, could alleviate DSS-induced disease symptoms. These results highlight the role of fecal miR-142a-3p in the prevention of colitis. We propose that the feces of subjects who have recovered from diseases might be enriched with miRNAs with preventive effects against those diseases.
Collapse
|
17
|
Zhang AN, Gaston JM, Dai CL, Zhao S, Poyet M, Groussin M, Yin X, Li LG, van Loosdrecht MCM, Topp E, Gillings MR, Hanage WP, Tiedje JM, Moniz K, Alm EJ, Zhang T. An omics-based framework for assessing the health risk of antimicrobial resistance genes. Nat Commun 2021; 12:4765. [PMID: 34362925 PMCID: PMC8346589 DOI: 10.1038/s41467-021-25096-3] [Citation(s) in RCA: 367] [Impact Index Per Article: 91.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Accepted: 07/23/2021] [Indexed: 12/24/2022] Open
Abstract
Antibiotic resistance genes (ARGs) are widespread among bacteria. However, not all ARGs pose serious threats to public health, highlighting the importance of identifying those that are high-risk. Here, we developed an ‘omics-based’ framework to evaluate ARG risk considering human-associated-enrichment, gene mobility, and host pathogenicity. Our framework classifies human-associated, mobile ARGs (3.6% of all ARGs) as the highest risk, which we further differentiate as ‘current threats’ (Rank I; 3%) - already present among pathogens - and ‘future threats’ (Rank II; 0.6%) - novel resistance emerging from non-pathogens. Our framework identified 73 ‘current threat’ ARG families. Of these, 35 were among the 37 high-risk ARGs proposed by the World Health Organization and other literature; the remaining 38 were significantly enriched in hospital plasmids. By evaluating all pathogen genomes released since framework construction, we confirmed that ARGs that recently transferred into pathogens were significantly enriched in Rank II (‘future threats’). Lastly, we applied the framework to gut microbiome genomes from fecal microbiota transplantation donors. We found that although ARGs were widespread (73% of genomes), only 8.9% of genomes contained high-risk ARGs. Our framework provides an easy-to-implement approach to identify current and future antimicrobial resistance threats, with potential clinical applications including reducing risk of microbiome-based interventions. Antibiotic resistance genes are common but not all are of high risk to human health. Here, the authors develop an omics-based framework for ranking genes by risk that incorporates level of enrichment in human associated environments, gene mobility, and host pathogenicity.
Collapse
Affiliation(s)
- An-Ni Zhang
- Environmental Microbiome Engineering and Biotechnology Laboratory, Department of Civil Engineering, The University of Hong Kong, Hong Kong SAR, China.,Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, USA
| | | | - Chengzhen L Dai
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, USA
| | - Shijie Zhao
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, USA
| | - Mathilde Poyet
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, USA.,Center for Microbiome Informatics and Therapeutics, Massachusetts Institute of Technology, Cambridge, USA.,The Broad Institute of MIT and Harvard, Cambridge, USA
| | - Mathieu Groussin
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, USA.,Center for Microbiome Informatics and Therapeutics, Massachusetts Institute of Technology, Cambridge, USA.,The Broad Institute of MIT and Harvard, Cambridge, USA
| | - Xiaole Yin
- Environmental Microbiome Engineering and Biotechnology Laboratory, Department of Civil Engineering, The University of Hong Kong, Hong Kong SAR, China
| | - Li-Guan Li
- Environmental Microbiome Engineering and Biotechnology Laboratory, Department of Civil Engineering, The University of Hong Kong, Hong Kong SAR, China
| | | | - Edward Topp
- London Research and Development Centre (LRDC), Agriculture and Agri-Food Canada, London, ON, Canada
| | - Michael R Gillings
- Department of Biological Sciences, Macquarie University, Sydney, NSW, Australia
| | - William P Hanage
- Center for Communicable Disease Dynamics, Department of Epidemiology, Harvard TH Chan School of Public Health, Boston, USA
| | - James M Tiedje
- Department of Plant, Soil and Microbial Sciences and of Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI, USA
| | - Katya Moniz
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, USA
| | - Eric J Alm
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, USA.,Center for Microbiome Informatics and Therapeutics, Massachusetts Institute of Technology, Cambridge, USA.,The Broad Institute of MIT and Harvard, Cambridge, USA
| | - Tong Zhang
- Environmental Microbiome Engineering and Biotechnology Laboratory, Department of Civil Engineering, The University of Hong Kong, Hong Kong SAR, China. .,School of Public Health, The University of Hong Kong, Hong Kong SAR, China. .,Center for Environmental Engineering Research, The University of Hong Kong, Hong Kong SAR, China.
| |
Collapse
|
18
|
High engraftment capacity of frozen ready-to-use human fecal microbiota transplants assessed in germ-free mice. Sci Rep 2021; 11:4365. [PMID: 33623056 PMCID: PMC7902644 DOI: 10.1038/s41598-021-83638-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Accepted: 02/01/2021] [Indexed: 11/15/2022] Open
Abstract
The number of indications for fecal microbiota transplantation is expected to rise, thus increasing the needs for production of readily available frozen or freeze-dried transplants. Using shotgun metagenomics, we investigated the capacity of two novel human fecal microbiota transplants prepared in maltodextrin-trehalose solutions (abbreviated MD and TR for maltodextrin:trehalose, 3:1, w/w, and trehalose:maltodextrin 3:1, w/w, respectively), to colonize a germ-free born mouse model. Gavage with frozen-thawed MD or TR suspensions gave the taxonomic profiles of mouse feces that best resembled those obtained with the fresh inoculum (Spearman correlations based on relative abundances of metagenomic species around 0.80 and 0.75 for MD and TR respectively), while engraftment capacity of defrosted NaCl transplants most diverged (Spearman correlations around 0.63). Engraftment of members of the family Lachnospiraceae and Ruminoccocaceae was the most challenging in all groups of mice, being improved with MD and TR transplants compared to NaCl, but still lower than with the fresh preparation. Improvement of engraftment of this important group in maintaining health represents a challenge that could benefit from further research on fecal microbiota transplant manufacturing.
Collapse
|
19
|
Iakupova AA, Abdulkhakov SR, Safin AG, Alieva IM, Oslopova JV, Abdulkhakov RA. [Fecal microbiota transplantation: donor selection criteria, storage and preparation of biomaterials (review of current recommendations)]. TERAPEVT ARKH 2021; 93:215-221. [PMID: 36286640 DOI: 10.26442/00403660.2021.02.200615] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Accepted: 04/06/2021] [Indexed: 12/12/2022]
Abstract
Fecal microbiota transplantation is a treatment method based on the introduction of donated fecal material to the recipient in order to restore the damaged composition of the intestinal microbiota. This review summarizes existing data on indications for fecal microbiota transplantation, recommendations for donor selection, processing and storage of donor biomaterial.
Collapse
Affiliation(s)
| | - S R Abdulkhakov
- Kazan (Volga Region) Federal University
- Kazan State Medical University
| | - A G Safin
- Kazan (Volga Region) Federal University
| | | | | | | |
Collapse
|
20
|
Zheng YM, Chen XY, Cai JY, Yuan Y, Xie WR, Xu JT, Xia HHX, Zhang M, He XX, Wu LH. Washed microbiota transplantation reduces proton pump inhibitor dependency in nonerosive reflux disease. World J Gastroenterol 2021; 27:513-522. [PMID: 33642825 PMCID: PMC7896436 DOI: 10.3748/wjg.v27.i6.513] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Revised: 12/20/2020] [Accepted: 01/12/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND The pathogenesis of gastroesophageal reflux disease (GERD) is closely associated with the intestinal bacteria composition and their metabolites. AIM To investigate whether washed microbiota transplantation (WMT) improves symptoms of nonerosive reflux disease (NERD) with proton pump inhibitor (PPI) dependency. METHODS Patients with recurrent NERD and PPI dependency at the First Affiliated Hospital of Guangdong Pharmaceutical University from 2017 to 2018 were included and divided into a WMT or PPI group treated with PPI with/without WMT. The endpoint was NERD symptom frequency evaluated 1 mo after WMT using reflux disease questionnaire (RDQ) and GERD questionnaire (GERDQ) scores, remission time, PPI dose, and the examination of intestinal mucosal barrier function. RESULTS In the WMT (n = 15) and PPI (n = 12) groups, the total remission rate at 1 mo after treatment was 93.3% vs 41.7%. Compared with the PPI group, the WMT group showed better results in GERDQ (P = 0.004) and RDQ (P = 0.003) and in remission months (8 vs 2, P = 0.002). The PPI dose was reduced to some extent for 80% of patients in the WMT group and 33.3% in the PPI group. In 24 patients, intestinal mucosal barrier function was examined before treatment, and changes in the degree of damage were observed in 13 of these patients after treatment. Only one of the 15 patients had minor side effects, including a mushy stool two or three times a day, which resolved on their own after 1 wk. CONCLUSION This study is the first to demonstrate that WMT may be safe and effective for relieving NERD symptoms and reducing PPI dependency and recurrence.
Collapse
Affiliation(s)
- Ya-Mei Zheng
- Department of Gastroenterology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Research Center for Engineering Techniques of Microbiota-Targeted Therapies of Guangdong Province, Guangzhou 510030, Guangdong Province, China
| | - Xian-Yun Chen
- Department of Gastroenterology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Research Center for Engineering Techniques of Microbiota-Targeted Therapies of Guangdong Province, Guangzhou 510030, Guangdong Province, China
| | - Jie-Yi Cai
- Department of Gastroenterology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Research Center for Engineering Techniques of Microbiota-Targeted Therapies of Guangdong Province, Guangzhou 510030, Guangdong Province, China
| | - Yu Yuan
- Department of Gastroenterology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Research Center for Engineering Techniques of Microbiota-Targeted Therapies of Guangdong Province, Guangzhou 510030, Guangdong Province, China
| | - Wen-Rui Xie
- Department of Gastroenterology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Research Center for Engineering Techniques of Microbiota-Targeted Therapies of Guangdong Province, Guangzhou 510030, Guangdong Province, China
| | - Jia-Ting Xu
- Department of Gastroenterology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Research Center for Engineering Techniques of Microbiota-Targeted Therapies of Guangdong Province, Guangzhou 510030, Guangdong Province, China
| | - Harry Hua-Xiang Xia
- Department of Science and Education, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou 510030, Guangdong Province, China
| | - Min Zhang
- Department of Epidemiology and Health Statistics, Guangdong Pharmaceutical University, Guangzhou 510220, Guangdong Province, China
| | - Xing-Xiang He
- Department of Gastroenterology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Research Center for Engineering Techniques of Microbiota-Targeted Therapies of Guangdong Province, Guangzhou 510030, Guangdong Province, China
| | - Li-Hao Wu
- Department of Gastroenterology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Research Center for Engineering Techniques of Microbiota-Targeted Therapies of Guangdong Province, Guangzhou 510030, Guangdong Province, China
| |
Collapse
|
21
|
Pession A, Zama D, Muratore E, Leardini D, Gori D, Guaraldi F, Prete A, Turroni S, Brigidi P, Masetti R. Fecal Microbiota Transplantation in Allogeneic Hematopoietic Stem Cell Transplantation Recipients: A Systematic Review. J Pers Med 2021; 11:100. [PMID: 33557125 PMCID: PMC7913807 DOI: 10.3390/jpm11020100] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 01/25/2021] [Accepted: 02/02/2021] [Indexed: 12/29/2022] Open
Abstract
The disruption of gut microbiota eubiosis has been linked to major complications in allogeneic hematopoietic stem cell transplantation (allo-HSCT) recipients. Various strategies have been developed to reduce dysbiosis and related complications. Fecal microbiota transplantation (FMT) consists of the infusion of fecal matter from a healthy donor to restore impaired intestinal homeostasis, and could be applied in the allo-HSCT setting. We conducted a systematic review of studies addressing the use of FMT in allo-HSCT patients. In the 23 papers included in the qualitative synthesis, FMT was used for the treatment of recurrent Clostridioides difficile infections or as a therapeutic strategy for steroid-resistant gut aGvHD. FMT was also performed with a preventive aim (e.g., to decolonize from antibiotic-resistant bacteria). Additional knowledge on the biological mechanisms underlying clinical findings is needed in order to employ FMT in clinical practice. There is also concern regarding the administration of microbial consortia in immune-compromised patients with altered gut permeability. Therefore, the safety profile and efficacy of the procedure must be determined to better assess the role of FMT in allo-HSCT recipients.
Collapse
Affiliation(s)
- Andrea Pession
- Pediatric Oncology and Hematology “Lalla Seràgnoli”, Pediatric Unit—IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; (A.P.); (D.Z.); (D.L.); (A.P.); (R.M.)
| | - Daniele Zama
- Pediatric Oncology and Hematology “Lalla Seràgnoli”, Pediatric Unit—IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; (A.P.); (D.Z.); (D.L.); (A.P.); (R.M.)
| | - Edoardo Muratore
- Pediatric Oncology and Hematology “Lalla Seràgnoli”, Pediatric Unit—IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; (A.P.); (D.Z.); (D.L.); (A.P.); (R.M.)
| | - Davide Leardini
- Pediatric Oncology and Hematology “Lalla Seràgnoli”, Pediatric Unit—IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; (A.P.); (D.Z.); (D.L.); (A.P.); (R.M.)
| | - Davide Gori
- Department of Biomedical and Neuromotor Sciences (DIBINEM), University of Bologna, 40126 Bologna, Italy; (D.G.); (F.G.)
| | - Federica Guaraldi
- Department of Biomedical and Neuromotor Sciences (DIBINEM), University of Bologna, 40126 Bologna, Italy; (D.G.); (F.G.)
- IRCCS Istituto delle Scienze Neurologiche di Bologna, 40126 Bologna, Italy
| | - Arcangelo Prete
- Pediatric Oncology and Hematology “Lalla Seràgnoli”, Pediatric Unit—IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; (A.P.); (D.Z.); (D.L.); (A.P.); (R.M.)
| | - Silvia Turroni
- Department of Pharmacy and Biotechnology (FABIT), University of Bologna, 40126 Bologna, Italy;
| | - Patrizia Brigidi
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40126 Bologna, Italy;
| | - Riccardo Masetti
- Pediatric Oncology and Hematology “Lalla Seràgnoli”, Pediatric Unit—IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; (A.P.); (D.Z.); (D.L.); (A.P.); (R.M.)
| |
Collapse
|
22
|
Zheng YM, He XX, Xia HHX, Yuan Y, Xie WR, Cai JY, Xu JT, Wu LH. Multi-donor multi-course faecal microbiota transplantation relieves the symptoms of chronic hemorrhagic radiation proctitis: A case report. Medicine (Baltimore) 2020; 99:e22298. [PMID: 32991434 PMCID: PMC7523865 DOI: 10.1097/md.0000000000022298] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2020] [Revised: 07/28/2020] [Accepted: 08/21/2020] [Indexed: 02/07/2023] Open
Abstract
RATIONALE There are many treatments for chronic hemorrhagic radiation colorectal inflammation, but only a few treatments are supported by high-quality research evidence. Studies have shown that the occurrence and development of radiation proctitis are closely associated with the intestinal flora. Animal studies have indicated that faecal microbiota transplantation (FMT) can improve radiation enteropathy in a mouse model. PATIENT CONCERNS A 45-year-old female patient suffered from recurrent hematochezia and diarrhea for half a year after radiotherapy and underwent recurrent transfusion treatments. Colonoscopy showed obvious congestion of the sigmoid colon and rectal mucosa, a smooth surface, and bleeding that was easily induced by touch, which are consistent with radiation proctitis. The pathological findings revealed chronic mucosal inflammation. The magnetic resonance imaging examination of the pelvic cavity with a plain scan and enhancement showed changes after radiotherapy and chemotherapy, and no obvious tumor recurrence or metastasis was found. The laboratory examinations excluded pathogen infection. DIAGNOSES Based on the history and examinations, the final diagnosis of this patient was chronic hemorrhagic radiation proctitis. INTERVENTIONS The patient was treated with a total of 4 individual courses of FMT. OUTCOMES After the six-month follow-up, her hematochezia, abdominal pain and diarrhea were relieved. Furthermore, 16S rRNA sequencing of the feces showed that the intestinal bacterial composition of the patient obviously changed after FMT and became similar to that of the donors. LESSONS This case report shows that FMT can relieve the symptoms of hematochezia and diarrhea by changing the bacterial community structure in patients with chronic hemorrhagic radiation proctitis.
Collapse
|
23
|
Tan Y, Shen J, Si T, Ho CL, Li Y, Dai L. Engineered Live Biotherapeutics: Progress and Challenges. Biotechnol J 2020; 15:e2000155. [DOI: 10.1002/biot.202000155] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 07/29/2020] [Indexed: 12/16/2022]
Affiliation(s)
- Yang Tan
- CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology (SIAT) Chinese Academy of Sciences Shenzhen 518055 China
| | - Juntao Shen
- CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology (SIAT) Chinese Academy of Sciences Shenzhen 518055 China
| | - Tong Si
- CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology (SIAT) Chinese Academy of Sciences Shenzhen 518055 China
- University of Chinese Academy of Sciences Beijing 100049 China
| | - Chun Loong Ho
- Department of Biomedical Engineering, SUSTech Academy for Advanced Interdisciplinary Studies Southern University of Science and Technology (SUSTech) Shenzhen 518055 China
| | - Yinqing Li
- School of Pharmaceutical Sciences Tsinghua University IDG‐McGovern Institute for Brain Research Beijing 100084 China
| | - Lei Dai
- CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology (SIAT) Chinese Academy of Sciences Shenzhen 518055 China
- University of Chinese Academy of Sciences Beijing 100049 China
| |
Collapse
|
24
|
Ianiro G, Mullish BH, Kelly CR, Kassam Z, Kuijper EJ, Ng SC, Iqbal TH, Allegretti JR, Bibbò S, Sokol H, Zhang F, Fischer M, Costello SP, Keller JJ, Masucci L, van Prehn J, Quaranta G, Quraishi MN, Segal J, Kao D, Satokari R, Sanguinetti M, Tilg H, Gasbarrini A, Cammarota G. Reorganisation of faecal microbiota transplant services during the COVID-19 pandemic. Gut 2020; 69:1555-1563. [PMID: 32620549 PMCID: PMC7456726 DOI: 10.1136/gutjnl-2020-321829] [Citation(s) in RCA: 116] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 06/23/2020] [Accepted: 06/24/2020] [Indexed: 12/11/2022]
Abstract
The COVID-19 pandemic has led to an exponential increase in SARS-CoV-2 infections and associated deaths, and represents a significant challenge to healthcare professionals and facilities. Individual countries have taken several prevention and containment actions to control the spread of infection, including measures to guarantee safety of both healthcare professionals and patients who are at increased risk of infection from COVID-19. Faecal microbiota transplantation (FMT) has a well-established role in the treatment of Clostridioides difficile infection. In the time of the pandemic, FMT centres and stool banks are required to adopt a workflow that continues to ensure reliable patient access to FMT while maintaining safety and quality of procedures. In this position paper, based on the best available evidence, worldwide FMT experts provide guidance on issues relating to the impact of COVID-19 on FMT, including patient selection, donor recruitment and selection, stool manufacturing, FMT procedures, patient follow-up and research activities.
Collapse
Affiliation(s)
- Gianluca Ianiro
- Digestive Disease Center, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Benjamin H Mullish
- Division of Digestive Diseases, Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, London, UK
| | - Colleen R Kelly
- Division of Gastroenterology, Alpert Medical School of Brown University, Providence, Rhode Island, USA
| | - Zain Kassam
- Finch Therapeutics Group, Somerville, Massachusetts, USA
| | - Ed J Kuijper
- Center for Microbiota Analysis and Therapy, Department of Medical Microbiology, Leiden University Medical Centre, Leiden, The Netherlands
- Netherlands Donor Feces Bank, Leiden University Medical Center, Leiden, The Netherlands
| | - Siew C Ng
- Center for Gut Microbiota Research, Institute of Digestive Disease, Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, Chinese University of Hong Kong, Hong Kong, Hong Kong
| | - Tariq H Iqbal
- Microbiome Treatment Centre, University of Birmingham, Birmingham, UK
- Department of Gastroenterology, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - Jessica R Allegretti
- Gastroenterology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Stefano Bibbò
- Digestive Disease Center, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Harry Sokol
- Service de Gastroenterologie; French Group of Fecal Microbiota Transplantation, Hôpital Saint Antoine, Sorbonne Université, Inserm, Centre de Recherche Saint-Antoine, Paris, France
- INRA, UMR1319 Micalis, AgroParisTech, Jouy-en-Josas, France
| | - Faming Zhang
- Medical Center for Digestive Diseases, the Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Monika Fischer
- Department of Medicine, Indiana University, Indianapolis, Indiana, USA
| | - Samuel Paul Costello
- Department of Gastroenterology, The Queen Elizabeth Hospital, University of Adelaide, Woodville, South Australia, Australia
| | - Josbert J Keller
- Netherlands Donor Feces Bank, Leiden University Medical Center, Leiden, The Netherlands
- Department of Gastroenterology, Haaglanden Medical Center, The Hague, The Netherlands
| | - Luca Masucci
- Institute of Microbiology, Fondazione Policlinico Universitario "A. Gemelli" IRCCS, Università Cattolica del Sacro Cuore, Roma, Italy
| | - Joffrey van Prehn
- Netherlands Donor Feces Bank, Leiden University Medical Center, Leiden, The Netherlands
| | - Gianluca Quaranta
- Institute of Microbiology, Fondazione Policlinico Universitario "A. Gemelli" IRCCS, Università Cattolica del Sacro Cuore, Roma, Italy
| | - Mohammed Nabil Quraishi
- Microbiome Treatment Centre, University of Birmingham, Birmingham, UK
- Department of Gastroenterology, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - Jonathan Segal
- Department of Gastroenterology, St Mark's Hospital, Harrow, UK
| | - Dina Kao
- Division of Gastroenterology, Department of Medicine, University of Alberta, Edmonton, Alberta, Canada
| | - Reetta Satokari
- Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Maurizio Sanguinetti
- Institute of Microbiology, Fondazione Policlinico Universitario "A. Gemelli" IRCCS, Università Cattolica del Sacro Cuore, Roma, Italy
| | - Herbert Tilg
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology & Metabolism, Innsbruck Medical University, Innsbruck, Austria
| | - Antonio Gasbarrini
- Digestive Disease Center, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Giovanni Cammarota
- Digestive Disease Center, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, Rome, Italy
| |
Collapse
|
25
|
Gulati M, Singh SK, Corrie L, Kaur IP, Chandwani L. Delivery routes for faecal microbiota transplants: Available, anticipated and aspired. Pharmacol Res 2020; 159:104954. [DOI: 10.1016/j.phrs.2020.104954] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Revised: 05/11/2020] [Accepted: 05/20/2020] [Indexed: 02/06/2023]
|
26
|
Tang LL, Feng WZ, Cheng JJ, Gong YN. Clinical remission of ulcerative colitis after different modes of faecal microbiota transplantation: a meta-analysis. Int J Colorectal Dis 2020; 35:1025-1034. [PMID: 32388604 DOI: 10.1007/s00384-020-03599-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/13/2020] [Indexed: 02/04/2023]
Abstract
BACKGROUND Ulcerative colitis (UC) is a chronic, recurrent and destructive disease of the gastrointestinal tract. Faecal microbiota transplantation (FMT) is a therapeutic measure in which faecal microbiota from healthy people is transplanted into patients. AIM To systematically evaluate the safety and effectiveness of treating UC with different modes of FMT. METHODS Seven databases were searched by two independent researchers and studies related to randomized controlled trials were included in the analysis. RESULTS Seven studies on UC involving 431 patients were included in the analysis. The results showed that FMT had better efficacy than placebo (OR = 2.29, 95% CI 1.48-3.53, P = 0.0002). Subgroup analyses of influencing factors showed that frozen faeces from multiple donors delivered via the lower gastrointestinal tract had a better curative effect than placebo (OR = 2.76, 95% CI 1.59-4.79, P = 0.0003; OR = 2.93, 95% CI 1.67-5.71, P = 0.0002; and OR = 2.70, 95% CI 1.67-4.37, P < 0.0001); the difference in efficacy between mixed faeces from a single donor transplanted through the upper gastrointestinal tract and placebo was not significant(P = 0.05, P = 0.09 and P = 0.98). The analysis of side effects showed no significant difference between FMT and placebo (P = 0.43). CONCLUSIONS It may be safe and effective to transplant frozen faeces from multiple donors through the lower gastrointestinal tract to treat UC.
Collapse
Affiliation(s)
- Li-Li Tang
- Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, China
| | - Wen-Zhe Feng
- Affiliated Hospital of Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, China.
| | - Jia-Jun Cheng
- Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, China
| | - Yan-Ni Gong
- Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, China
| |
Collapse
|
27
|
Lopetuso LR, Ianiro G, Allegretti JR, Bibbò S, Gasbarrini A, Scaldaferri F, Cammarota G. Fecal transplantation for ulcerative colitis: current evidence and future applications. Expert Opin Biol Ther 2020; 20:343-351. [PMID: 32083498 DOI: 10.1080/14712598.2020.1733964] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Introduction: Established evidence suggests that gut microbiota plays a role in ulcerative colitis (UC). Fecal microbiota transplantation (FMT) is clearly recognized as a highly effective treatment for patients with recurrent Clostridium difficile infection and has been investigated also in patients with UC, with promising results.Areas covered: Literature review was performed to select publications concerning current evidence on the role of gut microbiota in the pathogenesis of UC, and on the effectiveness of FMT in this disorder.Expert opinion: The randomized controlled trials published investigating the use of FMT suggested a potential role for FMT in the treatment of mild to moderate UC. However, given several unanswered questions regarding donor selection, dose, route of administration and duration of therapy, this is not yet recommended as a viable therapy option. FMT has allowed for more in depth investigation with regards to the role the gut microbiota may be playing in UC. This knowledge is critical to identifying where FMT may appropriately fit in the UC treatment paradigm. As our understanding of the role the microbiome plays in this chronic disease, FMT, and then eventually defined microbes, will hopefully serve in a complementary role to conventional IBD therapies.
Collapse
Affiliation(s)
- Loris R Lopetuso
- UOC MEDICINA INTERNA E GASTROENTEROLOGIA, Area Medicina Interna, Gastroenterologia ed Oncologia Medica, Dipartimento di Scienze Gastroenterologiche, Endocrino-Metaboliche e Nefro-Urologiche, Fondazione Policlinico Universitario A. Gemelli IRCCS, Roma, Italia.,Istituto di Patologia Speciale Medica, Università Cattolica del Sacro Cuore, Roma, Italia
| | - Gianluca Ianiro
- UOC MEDICINA INTERNA E GASTROENTEROLOGIA, Area Medicina Interna, Gastroenterologia ed Oncologia Medica, Dipartimento di Scienze Gastroenterologiche, Endocrino-Metaboliche e Nefro-Urologiche, Fondazione Policlinico Universitario A. Gemelli IRCCS, Roma, Italia.,Istituto di Patologia Speciale Medica, Università Cattolica del Sacro Cuore, Roma, Italia
| | - Jessica R Allegretti
- Division of Gastroenterology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Stefano Bibbò
- UOC MEDICINA INTERNA E GASTROENTEROLOGIA, Area Medicina Interna, Gastroenterologia ed Oncologia Medica, Dipartimento di Scienze Gastroenterologiche, Endocrino-Metaboliche e Nefro-Urologiche, Fondazione Policlinico Universitario A. Gemelli IRCCS, Roma, Italia.,Istituto di Patologia Speciale Medica, Università Cattolica del Sacro Cuore, Roma, Italia
| | - Antonio Gasbarrini
- UOC MEDICINA INTERNA E GASTROENTEROLOGIA, Area Medicina Interna, Gastroenterologia ed Oncologia Medica, Dipartimento di Scienze Gastroenterologiche, Endocrino-Metaboliche e Nefro-Urologiche, Fondazione Policlinico Universitario A. Gemelli IRCCS, Roma, Italia.,Istituto di Patologia Speciale Medica, Università Cattolica del Sacro Cuore, Roma, Italia
| | - Franco Scaldaferri
- UOC MEDICINA INTERNA E GASTROENTEROLOGIA, Area Medicina Interna, Gastroenterologia ed Oncologia Medica, Dipartimento di Scienze Gastroenterologiche, Endocrino-Metaboliche e Nefro-Urologiche, Fondazione Policlinico Universitario A. Gemelli IRCCS, Roma, Italia.,Istituto di Patologia Speciale Medica, Università Cattolica del Sacro Cuore, Roma, Italia
| | - Giovanni Cammarota
- UOC MEDICINA INTERNA E GASTROENTEROLOGIA, Area Medicina Interna, Gastroenterologia ed Oncologia Medica, Dipartimento di Scienze Gastroenterologiche, Endocrino-Metaboliche e Nefro-Urologiche, Fondazione Policlinico Universitario A. Gemelli IRCCS, Roma, Italia.,Istituto di Patologia Speciale Medica, Università Cattolica del Sacro Cuore, Roma, Italia
| |
Collapse
|
28
|
Liu S, Rezende RM, Moreira TG, Tankou SK, Cox LM, Wu M, Song A, Dhang FH, Wei Z, Costamagna G, Weiner HL. Oral Administration of miR-30d from Feces of MS Patients Suppresses MS-like Symptoms in Mice by Expanding Akkermansia muciniphila. Cell Host Microbe 2019; 26:779-794.e8. [PMID: 31784260 PMCID: PMC6948921 DOI: 10.1016/j.chom.2019.10.008] [Citation(s) in RCA: 142] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Revised: 09/08/2019] [Accepted: 10/14/2019] [Indexed: 01/15/2023]
Abstract
Fecal transfer from healthy donors is being explored as a microbiome modality. MicroRNAs (miRNAs) have been found to affect the microbiome. Multiple sclerosis (MS) patients have been shown to have an altered gut microbiome. Here, we unexpectedly found that transfer of feces harvested at peak disease from the experimental autoimmune encephalomyelitis (EAE) model of MS ameliorates disease in recipients in a miRNA-dependent manner. Specifically, we show that miR-30d is enriched in the feces of peak EAE and untreated MS patients. Synthetic miR-30d given orally ameliorates EAE through expansion of regulatory T cells (Tregs). Mechanistically, miR-30d regulates the expression of a lactase in Akkermansia muciniphila, which increases Akkermansia abundance in the gut. The expanded Akkermansia in turn increases Tregs to suppress EAE symptoms. Our findings report the mechanistic underpinnings of a miRNA-microbiome axis and suggest that the feces of diseased subjects might be enriched with miRNAs with therapeutic properties.
Collapse
Affiliation(s)
- Shirong Liu
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Partners Multiple Sclerosis Center, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA; Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA.
| | - Rafael M Rezende
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Partners Multiple Sclerosis Center, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA; Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Thais G Moreira
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Partners Multiple Sclerosis Center, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA; Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Stephanie K Tankou
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Partners Multiple Sclerosis Center, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA; Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Laura M Cox
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Partners Multiple Sclerosis Center, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA; Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Meng Wu
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Anya Song
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Partners Multiple Sclerosis Center, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA; Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Fyonn H Dhang
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Partners Multiple Sclerosis Center, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA; Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Zhiyun Wei
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Partners Multiple Sclerosis Center, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Gianluca Costamagna
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Partners Multiple Sclerosis Center, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA; Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Howard L Weiner
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Partners Multiple Sclerosis Center, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA; Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA
| |
Collapse
|
29
|
Cammarota G, Ianiro G, Kelly CR, Mullish BH, Allegretti JR, Kassam Z, Putignani L, Fischer M, Keller JJ, Costello SP, Sokol H, Kump P, Satokari R, Kahn SA, Kao D, Arkkila P, Kuijper EJ, Vehreschild MJG, Pintus C, Lopetuso L, Masucci L, Scaldaferri F, Terveer EM, Nieuwdorp M, López-Sanromán A, Kupcinskas J, Hart A, Tilg H, Gasbarrini A. International consensus conference on stool banking for faecal microbiota transplantation in clinical practice. Gut 2019; 68:2111-2121. [PMID: 31563878 PMCID: PMC6872442 DOI: 10.1136/gutjnl-2019-319548] [Citation(s) in RCA: 308] [Impact Index Per Article: 51.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Revised: 09/10/2019] [Accepted: 09/22/2019] [Indexed: 12/13/2022]
Abstract
Although faecal microbiota transplantation (FMT) has a well-established role in the treatment of recurrent Clostridioides difficile infection (CDI), its widespread dissemination is limited by several obstacles, including lack of dedicated centres, difficulties with donor recruitment and complexities related to regulation and safety monitoring. Given the considerable burden of CDI on global healthcare systems, FMT should be widely available to most centres.Stool banks may guarantee reliable, timely and equitable access to FMT for patients and a traceable workflow that ensures safety and quality of procedures. In this consensus project, FMT experts from Europe, North America and Australia gathered and released statements on the following issues related to the stool banking: general principles, objectives and organisation of the stool bank; selection and screening of donors; collection, preparation and storage of faeces; services and clients; registries, monitoring of outcomes and ethical issues; and the evolving role of FMT in clinical practice,Consensus on each statement was achieved through a Delphi process and then in a plenary face-to-face meeting. For each key issue, the best available evidence was assessed, with the aim of providing guidance for the development of stool banks in order to promote accessibility to FMT in clinical practice.
Collapse
Affiliation(s)
- Giovanni Cammarota
- Internal Medicine and Gastroenterology, Day Hospital of Gastroenterology and Intestinal Microbiota Transplantation, Fondazione Policlinico A Gemelli IRCCS, Catholic University of Medicine, Roma, Italy
| | - Gianluca Ianiro
- Internal Medicine and Gastroenterology, Day Hospital of Gastroenterology and Intestinal Microbiota Transplantation, Fondazione Policlinico A Gemelli IRCCS, Roma, Italy
| | - Colleen R Kelly
- Division of Gastroenterology, Alpert Medical School of Brown University, Providence, Rhode Island, United States of America
| | - Benjamin H Mullish
- Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Jessica R Allegretti
- Division of Gastroenterology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Zain Kassam
- Microbiome Informatics, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
- OpenBiome, Somerville, Massachusetts, United States of America
| | - Lorenza Putignani
- Parasitology Unit and Human Microbiome Unit, Bambino Gesù Pediatric Hospital, Roma, Italy
| | - Monika Fischer
- Department of Medicine, Indiana University, Indianapolis, Indiana, United States of America
| | - Josbert J Keller
- Department of Gastroenterologyand Hepatology, Haaglanden Medical Center, 2597 AX, The Hague, Netherlands
- National Donor Feces Bank, LUMC, Leiden, the Netherlands
| | - Samuel Paul Costello
- Department of Gastroenterology, The Queen Elizabeth Hospital, University of Adelaide, Woodville, South Australia, Australia
| | - Harry Sokol
- Service de Gastroenterologie, Hôpital Saint Antoine, Sorbonne Université, Inserm, Centre de Recherche Saint-Antoine, Paris, France
- French Group of Fecal Microbiota Transplantation, Paris, France
- INRA, UMR1319 Micalis, AgroParisTech, Jouy-en-Josas, France
| | - Patrizia Kump
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Reetta Satokari
- Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Stacy A Kahn
- Division of Gastroenterology, Hepatology and Nutrition, Boston Children's Hospital, Boston, Massachusetts, Uunited States of America
| | - Dina Kao
- Division of Gastroenterology, Department of Medicine, University of Alberta, Edmonton, Alberta, Canada
| | - Perttu Arkkila
- Department of Clinic of Gastroenterology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Ed J Kuijper
- Department of Medical Microbiology, Leiden University Medical Centre, Leiden, the Netherlands
| | - Maria J Gt Vehreschild
- Department I of Internal Medicine; German Centre for Infection Research, Partner site Bonn-Cologne, University Hospital of Cologne, Cologne, Germany
| | - Cristina Pintus
- Tissues and Cells Area, Italian National Transplant Center, Rome, Italy
| | - Loris Lopetuso
- Internal Medicine and Gastroenterology, Fondazione Policlinico A Gemelli IRCCS, Roma, Italy
| | - Luca Masucci
- Microbiology, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Catholic University of Medicine, Roma, Italy
| | - Franco Scaldaferri
- Internal Medicine and Gastroenterology, Fondazione Policlinico A Gemelli IRCCS, Roma, Italy
| | - E M Terveer
- National Donor Feces Bank, LUMC, Leiden, the Netherlands
- Department of Medical Microbiology, Leiden University Medical Centre, Leiden, the Netherlands
| | - Max Nieuwdorp
- Department of Internal Medicine, Amsterdam University Medical Centers, location AMC and VuMC, Amsterdam, Netherlands
| | - Antonio López-Sanromán
- Gastroenterology and Hepatology Department, Hospital Universitario Ramon y Cajal, Madrid, Spain
| | - Juozas Kupcinskas
- Department of Gastroenterology, Institute for Digestive Research, Medical Academy, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Ailsa Hart
- Department of Gastroenterology, St Mark's Hospital, London, United Kingdom
| | - Herbert Tilg
- Department of Internal Medicine I, Gastroenterology, Endocrinology & Metabolism, Innsbruck Medical University, Innsbruck, Austria
| | - Antonio Gasbarrini
- Internal Medicine and Gastroenterology, Fondazione Policlinico A Gemelli IRCCS, Catholic University of Medicine, Roma, Italy
| |
Collapse
|
30
|
Hasan N, Yang H. Factors affecting the composition of the gut microbiota, and its modulation. PeerJ 2019; 7:e7502. [PMID: 31440436 PMCID: PMC6699480 DOI: 10.7717/peerj.7502] [Citation(s) in RCA: 426] [Impact Index Per Article: 71.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Accepted: 07/17/2019] [Indexed: 12/13/2022] Open
Abstract
Gut microbiota have important functions in the body, and imbalances in the composition and diversity of those microbiota can cause several diseases. The host fosters favorable microbiota by releasing specific factors, such as microRNAs, and nonspecific factors, such as antimicrobial peptides, mucus and immunoglobulin A that encourage the growth of specific types of bacteria and inhibit the growth of others. Diet, antibiotics, and age can change gut microbiota, and many studies have shown the relationship between disorders of the microbiota and several diseases and reported some ways to modulate that balance. In this review, we highlight how the host shapes its gut microbiota via specific and nonspecific factors, how environmental and nutritional factors affect it, and how to modulate it using prebiotics, probiotics, and fecal microbiota transplantation.
Collapse
Affiliation(s)
- Nihal Hasan
- Department of Microbiology, Northeast Forestry University, Harbin, Heilongjiang, China.,Faculty of Health Science, Al-Baath University, Homs, Syria
| | - Hongyi Yang
- Department of Microbiology, Northeast Forestry University, Harbin, Heilongjiang, China
| |
Collapse
|
31
|
Chen J, Yu B, Chen D, Zheng P, Luo Y, Huang Z, Luo J, Mao X, Yu J, He J. Changes of porcine gut microbiota in response to dietary chlorogenic acid supplementation. Appl Microbiol Biotechnol 2019; 103:8157-8168. [PMID: 31401751 DOI: 10.1007/s00253-019-10025-8] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Revised: 07/07/2019] [Accepted: 07/09/2019] [Indexed: 12/15/2022]
Abstract
Chlorogenic acids (CGA), the most abundant natural polyphenol present in human diet and plants, have attracted considerable research interest because of their broad bioactivities including the antimicrobial activity. However, little is known about their influences on intestinal bacterial communities. Here, we described a response in intestinal microbiome to CGA using a porcine model. Twenty-four weaned pigs were allotted to two groups and fed with a basal diet or a basal diet containing 1000 mg/kg CGA. Results showed that CGA significantly increased the length of the small intestine (P < 0.05) and enhanced the activity of diamine oxidase (DAO) and the concentration of MHC-II in the jejunal and ileal mucosa (P < 0.05). Moreover, the acetate concentration in ileum and cecum digesta, and the propionate and butyrate concentrations in the cecum digesta, were significantly elevated by CGA (P < 0.05). Interestingly, CGA significantly increased the total 16S rRNA gene copies and bacterial alpha diversity in the cecum (P < 0.05). The relative abundance of bacteria from phyla Firmicutes and Bacteroidetes was increased in the cecum digesta (P < 0.05), whereas the abundance of bacteria from phylum Protebacteria was decreased by CGA (P < 0.05). Importantly, pigs on CGA-containing diet had higher abundance of Lactobacillus spp., Prevotella spp., Anaerovibrio spp., and Alloprevotella spp. in the cecum (P < 0.05). Not only did our study suggest a synergic response of intestinal barrier function and microbiota to the CGA, but the result will also contribute to understanding of the mechanisms behind the CGA-modulated gut health.
Collapse
Affiliation(s)
- Jiali Chen
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan, 611130, People's Republic of China.,Key Laboratory of Animal Disease-Resistance Nutrition, Chengdu, Sichuan, 611130, People's Republic of China
| | - Bing Yu
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan, 611130, People's Republic of China.,Key Laboratory of Animal Disease-Resistance Nutrition, Chengdu, Sichuan, 611130, People's Republic of China
| | - Daiwen Chen
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan, 611130, People's Republic of China.,Key Laboratory of Animal Disease-Resistance Nutrition, Chengdu, Sichuan, 611130, People's Republic of China
| | - Ping Zheng
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan, 611130, People's Republic of China.,Key Laboratory of Animal Disease-Resistance Nutrition, Chengdu, Sichuan, 611130, People's Republic of China
| | - Yuheng Luo
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan, 611130, People's Republic of China.,Key Laboratory of Animal Disease-Resistance Nutrition, Chengdu, Sichuan, 611130, People's Republic of China
| | - Zhiqing Huang
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan, 611130, People's Republic of China.,Key Laboratory of Animal Disease-Resistance Nutrition, Chengdu, Sichuan, 611130, People's Republic of China
| | - Junqiu Luo
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan, 611130, People's Republic of China.,Key Laboratory of Animal Disease-Resistance Nutrition, Chengdu, Sichuan, 611130, People's Republic of China
| | - Xiangbing Mao
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan, 611130, People's Republic of China.,Key Laboratory of Animal Disease-Resistance Nutrition, Chengdu, Sichuan, 611130, People's Republic of China
| | - Jie Yu
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan, 611130, People's Republic of China.,Key Laboratory of Animal Disease-Resistance Nutrition, Chengdu, Sichuan, 611130, People's Republic of China
| | - Jun He
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan, 611130, People's Republic of China. .,Key Laboratory of Animal Disease-Resistance Nutrition, Chengdu, Sichuan, 611130, People's Republic of China.
| |
Collapse
|
32
|
Mukhopadhya I, Segal JP, Carding SR, Hart AL, Hold GL. The gut virome: the 'missing link' between gut bacteria and host immunity? Therap Adv Gastroenterol 2019; 12:1756284819836620. [PMID: 30936943 PMCID: PMC6435874 DOI: 10.1177/1756284819836620] [Citation(s) in RCA: 115] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2019] [Accepted: 02/14/2019] [Indexed: 02/04/2023] Open
Abstract
The human gut virome includes a diverse collection of viruses that infect our own cells as well as other commensal organisms, directly impacting on our well-being. Despite its predominance, the virome remains one of the least understood components of the gut microbiota, with appropriate analysis toolkits still in development. Based on its interconnectivity with all living cells, it is clear that the virome cannot be studied in isolation. Here we review the current understanding of the human gut virome, specifically in relation to other constituents of the microbiome, its evolution and life-long association with its host, and our current understanding in the context of inflammatory bowel disease and associated therapies. We propose that the gut virome and the gut bacterial microbiome share similar trajectories and interact in both health and disease and that future microbiota studies should in parallel characterize the gut virome to uncover its role in health and disease.
Collapse
Affiliation(s)
- Indrani Mukhopadhya
- Gastrointestinal Research Group, Division of Applied Medicine, University of Aberdeen, Foresterhill, Aberdeen, UK Gut Health Group, The Rowett Institute, University of Aberdeen, Foresterhill, Aberdeen, UK
| | - Jonathan P. Segal
- St. Mark’s Hospital, Watford Road, Harrow, UK Imperial College London, South Kensington Campus, Department of Surgery and Cancer, London, UK
| | - Simon R. Carding
- Gut Microbes and Health Research Programme, The Quadram Institute, Norwich Research Park, Norwich, Norfolk, UK Norwich Medical School, University of East Anglia, Norwich Research Park, Norwich, Norfolk, UK
| | - Ailsa L. Hart
- St. Mark’s Hospital, Watford Road, Harrow, UK Imperial College London, South Kensington Campus, Department of Surgery and Cancer, London, UK
| | | |
Collapse
|
33
|
Cammarota G, Gallo A, Ianiro G, Montalto M. Emerging drugs for the treatment of clostridium difficile. Expert Opin Emerg Drugs 2019; 24:17-28. [PMID: 30841760 DOI: 10.1080/14728214.2019.1591371] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
INTRODUCTION Clostridium difficile or Clostridioides difficile (C. difficile) infection represents the most common cause of healthcare-associated infection. Over the last decades, the incidence and severity of C. difficile infection is rapidly increasing, with a significant impact on morbidity and mortality, and burden on health care system. Orally administered vancomycin and fidaxomicin are the therapeutic options of choice for initial C. difficile infection and fecal microbiota transplant for the recurrence infection. Furthermore, in recent years several new antibiotics with narrow-spectrum activity and low intestinal resorption have been developed, including surotomycin, cadazolid, and ridinilazol, and novel toxoid vaccines are expected to be efficacious in the prevention of C. difficile infection. Areas covered: Literature review was performed to select publications about current guidelines and phase-II/III trials on emerging drugs. These include novel antibiotics, monoclonal antibodies, vaccines, and fecal microbiota transplantation. Expert opinion: We have today a wide spectrum of promising therapeutic possibilities against infection. Pivotal future clinical trials may be crucial in developing effective strategies to optimize outcomes, mainly in high-risk population.
Collapse
Affiliation(s)
- Giovanni Cammarota
- a UOC di Medicina Interna , F. Policlinico Universitario A. Gemelli IRCCS, Università Cattolica del Sacro Cuore , Roma , Italy
| | - Antonella Gallo
- b UOC di Medicina Interna , F. Policlinico Universitario A. Gemelli IRCCS , Roma , Italy
| | - Gianluca Ianiro
- c UOC di Medicina Interna e Gastroenterologia , F. Policlinico Universitario A. Gemelli IRCCS , Roma , Italy
| | - Massimo Montalto
- a UOC di Medicina Interna , F. Policlinico Universitario A. Gemelli IRCCS, Università Cattolica del Sacro Cuore , Roma , Italy
| |
Collapse
|
34
|
Vaginal microbiota transplantation for the treatment of bacterial vaginosis: a conceptual analysis. FEMS Microbiol Lett 2019; 366:5304978. [DOI: 10.1093/femsle/fnz025] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Accepted: 01/30/2019] [Indexed: 12/14/2022] Open
|
35
|
Sun X, Jia Z. Microbiome modulates intestinal homeostasis against inflammatory diseases. Vet Immunol Immunopathol 2018; 205:97-105. [PMID: 30459007 DOI: 10.1016/j.vetimm.2018.10.014] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Revised: 10/21/2018] [Accepted: 10/27/2018] [Indexed: 02/07/2023]
Abstract
Eliminating prophylactic antibiotics in food animal production has exerted pressure on discovering antimicrobial alternatives (e.g. microbiome) to reduce elevated intestinal diseases. Intestinal tract is a complex ecosystem coupling host cells with microbiota. The microbiota and its metabolic activities and products are collectively called microbiome. Intestinal homeostasis is reached through dynamic and delicate crosstalk between host immunity and microbiome. However, this balance can be occasionally broken, which results in intestinal inflammatory diseases such as human Inflammatory Bowel Diseases, chicken necrotic enteritis, and swine postweaning diarrhea. In this review, we introduce the intestinal immune system, intestinal microbiome, and microbiome modulation of inflammation against intestinal diseases. The purpose of this review is to provide updated knowledge on host-microbe interaction and to promote using microbiome as new antimicrobial strategies to reduce intestinal diseases.
Collapse
Affiliation(s)
- Xiaolun Sun
- Center of Excellence for Poultry Science, University of Arkansas, Fayetteville, AR, 72701, United States.
| | - Zhenquan Jia
- Department of Biology, University of North Carolina at Greensboro, Greensboro, NC 27402, United States
| |
Collapse
|
36
|
|
37
|
D'Odorico I, Di Bella S, Monticelli J, Giacobbe DR, Boldock E, Luzzati R. Role of fecal microbiota transplantation in inflammatory bowel disease. J Dig Dis 2018; 19:322-334. [PMID: 29696802 DOI: 10.1111/1751-2980.12603] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Revised: 01/09/2018] [Accepted: 04/22/2018] [Indexed: 12/11/2022]
Abstract
There is increasing evidence of the key role played by altered intestinal microbiota in the pathogenesis of inflammatory bowel disease (IBD). Management strategies involving immune modulation are effective and widely used, but treatment failures and side effects occur. Fecal microbiota transplantation (FMT) provides a novel, perhaps complementary, strategy to restore the normal gut microbiota in patients with IBD. This review summarizes the available efficacy and safety data on the use of FMT in patients with IBD. Several aspects remain to be clarified about the clinical predictors of the response to FMT, its most appropriate route of administration, and the most appropriate quantity and quality of microbiota to be transplanted. Further studies focusing on long-term outcomes and safety are also warranted.
Collapse
Affiliation(s)
| | - Stefano Di Bella
- Infectious Diseases Division, University Hospital of Trieste, Trieste, Italy
| | - Jacopo Monticelli
- Infectious Diseases Division, University Hospital of Trieste, Trieste, Italy
| | - Daniele R Giacobbe
- Infectious Diseases Unit, OspedalePoliclinico San Martino-IRCCS per l'Oncologia and University of Genoa, Genoa, Italy
| | - Emma Boldock
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK
| | - Roberto Luzzati
- Infectious Diseases Division, University Hospital of Trieste, Trieste, Italy
| |
Collapse
|
38
|
Segal JP, Abbasi F, Kanagasundaram C, Hart A. Does the Internet promote the unregulated use of fecal microbiota transplantation: a potential public health issue? Clin Exp Gastroenterol 2018; 11:179-183. [PMID: 29750050 PMCID: PMC5935081 DOI: 10.2147/ceg.s159609] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
INTRODUCTION The Internet has become an increasingly popular resource for medical information. Fecal microbiota transplantation (FMT) has changed the treatment of Clostridium difficile with cure rates of 81% following one infusion of FMT, further studies have since validated these findings. The Medicines and Health care Products Regulatory Agency has classified FMT as a medicine and hence should be only utilized in strict clinical settings. METHODS We searched Facebook, Twitter, Google, and YouTube using the words "Faecal Microbiota Transplantation" and "FMT". We utilized the first 50 hits on each site. We analyzed the percentage of articles that fell outside regulated medical practice. We searched how many clinics in the UK advertised practice that falls outside suggested guidelines. RESULTS Google, YouTube, and Facebook had a variety of information regarding FMT available. Nine out of 50 (18%) of the top 50 google searches can be considered articles that fall outside regulated practice. YouTube highlighted four videos describing how to self-administer FMT, one of these was for ulcerative colitis. Fourteen percent of the top 50 YouTube videos fall outside regulated practice and 8% of the top 50 Facebook searches fall outside regulated clinical practice. There were two clinics in the UK advertising FMT for uses that fall outside regulated practice. CONCLUSION Clinicians and patients need to be aware of the resources available through social media and the Internet. It should be appreciated that some websites fall outside regulated clinical practice. Private clinics offering FMT need to ensure that they are offering FMT within a regulated framework.
Collapse
Affiliation(s)
| | | | | | - Ailsa Hart
- Department of Gastroenterology, St Mark’s Hospital, Harrow, UK
| |
Collapse
|
39
|
Testa A, Rispo A, Imperatore N, Nardone OM, Trinchese G, Cavaliere G, Castiglione F, Mollica MP. Gut microbiota and Crohn’s disease. MEDITERRANEAN JOURNAL OF NUTRITION AND METABOLISM 2018. [DOI: 10.3233/mnm-17182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
INTRODUCTION: Crohn’s disease (CD) is characterized by a chronic inflammation of the gastrointestinal tract causing abdominal pain, diarrhea, weight loss and systemic symptoms. Although the etiology of this disease is unknown, current knowledge suggests a multifactorial genesis involving genetic, environmental and immunological factors. EVIDENCE ACQUISITION: We focused our attention on critical analysis of the recent literature on the role of gut microbiota in inflammatory bowel disease (IBD), by evaluating the differences of composition, functions and role of intestinal flora. In particular, we focused on evidences about the interaction between gut microbiota and pathogenesis of IBD. In this setting, we conducted a PUBMED search for guidelines, systematic reviews (SR) and primary studies. EVIDENCE SYNTHESIS: Some data suggest that, in a significant percentage of patients, the microbiota plays an important role in the genesis and maintenance of CD. Probiotic supplementation and antibiotic treatment appear to be a valid therapeutic approach, although the clinical data remain controversial. CONCLUSIONS: Despite the exciting and growing research on the role of gut microbiota in IBD, our knowledge remains fairly limited. Further studies are needed to measure the diversity, function and resistance to antibiotics of the intestinal microbiota in CD.
Collapse
Affiliation(s)
- Anna Testa
- Department of Clinical Medicine and Surgery, Gastroenterology, School of Medicine “Federico II” of Naples, Naples, Italy
| | - Antonio Rispo
- Department of Clinical Medicine and Surgery, Gastroenterology, School of Medicine “Federico II” of Naples, Naples, Italy
| | - Nicola Imperatore
- Department of Clinical Medicine and Surgery, Gastroenterology, School of Medicine “Federico II” of Naples, Naples, Italy
| | - Olga Maria Nardone
- Department of Clinical Medicine and Surgery, Gastroenterology, School of Medicine “Federico II” of Naples, Naples, Italy
| | - Giovanna Trinchese
- Department of Biology, University of Naples “Federico II”, Naples, Italy
| | - Gina Cavaliere
- Department of Biology, University of Naples “Federico II”, Naples, Italy
| | - Fabiana Castiglione
- Department of Clinical Medicine and Surgery, Gastroenterology, School of Medicine “Federico II” of Naples, Naples, Italy
| | - Maria Pina Mollica
- Department of Biology, University of Naples “Federico II”, Naples, Italy
| |
Collapse
|
40
|
Shen ZH, Zhu CX, Quan YS, Yang ZY, Wu S, Luo WW, Tan B, Wang XY. Relationship between intestinal microbiota and ulcerative colitis: Mechanisms and clinical application of probiotics and fecal microbiota transplantation. World J Gastroenterol 2018; 24:5-14. [PMID: 29358877 PMCID: PMC5757125 DOI: 10.3748/wjg.v24.i1.5] [Citation(s) in RCA: 442] [Impact Index Per Article: 63.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2017] [Revised: 11/07/2017] [Accepted: 11/21/2017] [Indexed: 02/06/2023] Open
Abstract
Ulcerative colitis (UC) is an inflammatory disease that mainly affects the colon and rectum. It is believed that genetic factors, host immune system disorders, intestinal microbiota dysbiosis, and environmental factors contribute to the pathogenesis of UC. However, studies on the role of intestinal microbiota in the pathogenesis of UC have been inconclusive. Studies have shown that probiotics improve intestinal mucosa barrier function and immune system function and promote secretion of anti-inflammatory factors, thereby inhibiting the growth of harmful bacteria in the intestine. Fecal microbiota transplantation (FMT) can reduce bowel permeability and thus the severity of disease by increasing the production of short-chain fatty acids, especially butyrate, which help maintain the integrity of the epithelial barrier. FMT can also restore immune dysbiosis by inhibiting Th1 differentiation, activity of T cells, leukocyte adhesion, and production of inflammatory factors. Probiotics and FMT are being increasingly used to treat UC, but their use is controversial because of uncertain efficacy. Here, we briefly review the role of intestinal microbiota in the pathogenesis and treatment of UC.
Collapse
Affiliation(s)
- Zhao-Hua Shen
- Department of Gastroenterology, Third Xiangya Hospital, Central South University, Changsha 410013, Hunan Province, China
- Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Changsha 410008, Hunan Province, China
| | - Chang-Xin Zhu
- Department of Gastroenterology, Third Xiangya Hospital, Central South University, Changsha 410013, Hunan Province, China
- Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Changsha 410008, Hunan Province, China
| | - Yong-Sheng Quan
- Department of Gastroenterology, Third Xiangya Hospital, Central South University, Changsha 410013, Hunan Province, China
- Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Changsha 410008, Hunan Province, China
| | - Zhen-Yu Yang
- Department of Gastroenterology, Third Xiangya Hospital, Central South University, Changsha 410013, Hunan Province, China
- Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Changsha 410008, Hunan Province, China
| | - Shuai Wu
- Department of Gastroenterology, Third Xiangya Hospital, Central South University, Changsha 410013, Hunan Province, China
- Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Changsha 410008, Hunan Province, China
| | - Wei-Wei Luo
- Department of Gastroenterology, Third Xiangya Hospital, Central South University, Changsha 410013, Hunan Province, China
- Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Changsha 410008, Hunan Province, China
| | - Bei Tan
- Department of Gastroenterology, Third Xiangya Hospital, Central South University, Changsha 410013, Hunan Province, China
- Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Changsha 410008, Hunan Province, China
| | - Xiao-Yan Wang
- Department of Gastroenterology, Third Xiangya Hospital, Central South University, Changsha 410013, Hunan Province, China
- Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Changsha 410008, Hunan Province, China
| |
Collapse
|
41
|
Maida M, Mcilroy J, Ianiro G, Cammarota G. Faecal Microbiota Transplantation as Emerging Treatment in European Countries. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1050:177-195. [PMID: 29383670 DOI: 10.1007/978-3-319-72799-8_11] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Clostridium difficile infection (CDI) is one of the most common healthcare-associated infections in the world and is a leading cause of morbidity and mortality in hospitalized patients.Although several antibiotics effectively treat CDI, some individuals do not respond to these drugs and may be cured by transplanting stool from healthy donors. This procedure, termed Faecal Microbiota Transplantation (FMT), has demonstrated remarkable efficacy as a treatment for recurrent CDI.FMT has also been investigated in other diseases and disorders where perturbations to the gut microbiota have been theorized to play a causative role in pathogenesis and severity, such as inflammatory bowel disease (IBD). Although FMT is currently not recommended to cure IBD patients in clinical practice, several studies have recently been carried out with promising results. The aim of future research is therefore to standardize protocols and develop FMT as a therapeutic option for these patients.This review summarizes data on the use of FMT as a treatment for CDI and IBD, with special attention given to studies conducted in European countries.
Collapse
Affiliation(s)
- Marcello Maida
- Section of Gastroenterology, S.Elia - Raimondi Hospital, Caltanissetta, Italy
| | - James Mcilroy
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen, UK
| | - Gianluca Ianiro
- Gastroenterological Area, Fondazione Policlinico Universitario Gemelli, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Giovanni Cammarota
- Gastroenterological Area, Fondazione Policlinico Universitario Gemelli, Università Cattolica del Sacro Cuore, Rome, Italy.
| |
Collapse
|
42
|
Basson AR, Lam M, Cominelli F. Complementary and Alternative Medicine Strategies for Therapeutic Gut Microbiota Modulation in Inflammatory Bowel Disease and their Next-Generation Approaches. Gastroenterol Clin North Am 2017; 46:689-729. [PMID: 29173517 PMCID: PMC5909826 DOI: 10.1016/j.gtc.2017.08.002] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The human gut microbiome exerts a major impact on human health and disease, and therapeutic gut microbiota modulation is now a well-advocated strategy in the management of many diseases, including inflammatory bowel disease (IBD). Scientific and clinical evidence in support of complementary and alternative medicine, in targeting intestinal dysbiosis among patients with IBD, or other disorders, has increased dramatically over the past years. Delivery of "artificial" stool replacements for fecal microbiota transplantation (FMT) could provide an effective, safer alternative to that of human donor stool. Nevertheless, optimum timing of FMT administration in IBD remains unexplored, and future investigations are essential.
Collapse
Affiliation(s)
- Abigail R Basson
- Digestive Health Research Institute, Case Western Reserve University, Cleveland, OH, USA; Department of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Minh Lam
- Digestive Health Research Institute, Case Western Reserve University, Cleveland, OH, USA; Department of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Fabio Cominelli
- Digestive Health Research Institute, Case Western Reserve University, Cleveland, OH, USA; Department of Medicine, Case Western Reserve University, Cleveland, OH, USA; Department of Pathology, Case Western Reserve University, Cleveland, OH, USA.
| |
Collapse
|
43
|
Holleran G, Lopetuso L, Petito V, Graziani C, Ianiro G, McNamara D, Gasbarrini A, Scaldaferri F. The Innate and Adaptive Immune System as Targets for Biologic Therapies in Inflammatory Bowel Disease. Int J Mol Sci 2017; 18:E2020. [PMID: 28934123 PMCID: PMC5666702 DOI: 10.3390/ijms18102020] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2017] [Revised: 09/10/2017] [Accepted: 09/14/2017] [Indexed: 12/12/2022] Open
Abstract
Inflammatory bowel disease (IBD) is an immune-mediated inflammatory condition causing inflammation of gastrointestinal and systemic cells, with an increasing prevalence worldwide. Many factors are known to trigger and maintain inflammation in IBD including the innate and adaptive immune systems, genetics, the gastrointestinal microbiome and several environmental factors. Our knowledge of the involvement of the immune system in the pathophysiology of IBD has advanced rapidly over the last two decades, leading to the development of several immune-targeted treatments with a biological source, known as biologic agents. The initial focus of these agents was directed against the pro-inflammatory cytokine tumor necrosis factor-α (TNF-α) leading to dramatic changes in the disease course for a proportion of patients with IBD. However, more recently, it has been shown that a significant proportion of patients do not respond to anti-TNF-α directed therapies, leading a shift to other inflammatory pathways and targets, including those of both the innate and adaptive immune systems, and targets linking both systems including anti-leukocyte trafficking agents-integrins and adhesion molecules. This review briefly describes the molecular basis of immune based gastrointestinal inflammation in IBD, and then describes how several current and future biologic agents work to manipulate these pathways, and their clinical success to date.
Collapse
Affiliation(s)
- Grainne Holleran
- Internal Medicine, Gastroenterology and Liver Unit, Gastroenterology Area, Fondazione Policlinico Universitario Gemelli, Università Cattolica del Sacro Cuore, 00168 Rome, Italy.
- Gastroenterology Department, Department of Clinical Medicine, Trinity College Dublin, Dublin 2, Ireland.
| | - Loris Lopetuso
- Internal Medicine, Gastroenterology and Liver Unit, Gastroenterology Area, Fondazione Policlinico Universitario Gemelli, Università Cattolica del Sacro Cuore, 00168 Rome, Italy.
| | - Valentina Petito
- Internal Medicine, Gastroenterology and Liver Unit, Gastroenterology Area, Fondazione Policlinico Universitario Gemelli, Università Cattolica del Sacro Cuore, 00168 Rome, Italy.
| | - Cristina Graziani
- Internal Medicine, Gastroenterology and Liver Unit, Gastroenterology Area, Fondazione Policlinico Universitario Gemelli, Università Cattolica del Sacro Cuore, 00168 Rome, Italy.
| | - Gianluca Ianiro
- Internal Medicine, Gastroenterology and Liver Unit, Gastroenterology Area, Fondazione Policlinico Universitario Gemelli, Università Cattolica del Sacro Cuore, 00168 Rome, Italy.
| | - Deirdre McNamara
- Gastroenterology Department, Department of Clinical Medicine, Trinity College Dublin, Dublin 2, Ireland.
| | - Antonio Gasbarrini
- Internal Medicine, Gastroenterology and Liver Unit, Gastroenterology Area, Fondazione Policlinico Universitario Gemelli, Università Cattolica del Sacro Cuore, 00168 Rome, Italy.
| | - Franco Scaldaferri
- Internal Medicine, Gastroenterology and Liver Unit, Gastroenterology Area, Fondazione Policlinico Universitario Gemelli, Università Cattolica del Sacro Cuore, 00168 Rome, Italy.
| |
Collapse
|
44
|
Zhang M, Sun K, Wu Y, Yang Y, Tso P, Wu Z. Interactions between Intestinal Microbiota and Host Immune Response in Inflammatory Bowel Disease. Front Immunol 2017; 8:942. [PMID: 28855901 PMCID: PMC5558048 DOI: 10.3389/fimmu.2017.00942] [Citation(s) in RCA: 233] [Impact Index Per Article: 29.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Accepted: 07/24/2017] [Indexed: 12/13/2022] Open
Abstract
Inflammatory bowel disease (IBD) is a chronic inflammatory disorder of the gastrointestinal tract. Although the etiology and pathogenesis of IBD remain unclear, both genetic susceptibility and environmental factors are implicated in the initiation and progression of IBD. Recent studies with experimental animal models and clinical patients indicated that the intestinal microbiota is one of the critical environmental factors that influence nutrient metabolism, immune responses, and the health of the host in various intestinal diseases, including ulcerative colitis and Crohn’s disease. The objective of this review is to highlight the crosstalk between gut microbiota and host immune response and the contribution of this interaction to the pathogenesis of IBD. In addition, potential therapeutic strategies targeting the intestinal micro-ecosystem in IBD are discussed.
Collapse
Affiliation(s)
- Ming Zhang
- State Key Laboratory of Animal Nutrition, Department of Animal Nutrition and Feed Science, China Agricultural University, Beijing, China
| | - Kaiji Sun
- State Key Laboratory of Animal Nutrition, Department of Animal Nutrition and Feed Science, China Agricultural University, Beijing, China
| | - Yujun Wu
- State Key Laboratory of Animal Nutrition, Department of Animal Nutrition and Feed Science, China Agricultural University, Beijing, China
| | - Ying Yang
- State Key Laboratory of Animal Nutrition, Department of Animal Nutrition and Feed Science, China Agricultural University, Beijing, China
| | - Patrick Tso
- Department of Pathology and Laboratory Medicine, Metabolic Diseases Institute, University of Cincinnati, Cincinnati, OH, United States
| | - Zhenlong Wu
- State Key Laboratory of Animal Nutrition, Department of Animal Nutrition and Feed Science, China Agricultural University, Beijing, China.,Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing, China
| |
Collapse
|
45
|
Lane ER, Zisman TL, Suskind DL. The microbiota in inflammatory bowel disease: current and therapeutic insights. J Inflamm Res 2017; 10:63-73. [PMID: 28652796 PMCID: PMC5473501 DOI: 10.2147/jir.s116088] [Citation(s) in RCA: 165] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Inflammatory bowel disease is a heterogeneous group of chronic disorders that result from the interaction of the intestinal immune system with the gut microbiome. Until recently, most investigative efforts and therapeutic breakthroughs were centered on understanding and manipulating the altered mucosal immune response that characterizes these diseases. However, more recent studies have highlighted the important role of environmental factors, and in particular the microbiota, in disease onset and disease exacerbation. Advances in genomic sequencing technology and bioinformatics have facilitated an explosion of investigative inquiries into the composition and function of the intestinal microbiome in health and disease and have advanced our understanding of the interplay between the gut microbiota and the host immune system. The gut microbiome is dynamic and changes with age and in response to diet, antibiotics and other environmental factors, and these alterations in the microbiome contribute to disease onset and exacerbation. Strategies to manipulate the microbiome through diet, probiotics, antibiotics or fecal microbiota transplantation may potentially be used therapeutically to influence modulate disease activity. This review will characterize the factors involved in the development of the intestinal microbiome and will describe the typical alterations in the microbiota that are characteristic of inflammatory bowel disease. Additionally, this manuscript will summarize the early but promising literature on the role of the gut microbiota in the pathogenesis of inflammatory bowel disease with implications for utilizing this data for diagnostic or therapeutic application in the clinical management of patients with these diseases.
Collapse
Affiliation(s)
- Erin R Lane
- Division of Gastroenterology and Hepatology, Seattle Children's Hospital
| | - Timothy L Zisman
- Division of Gastroenterology, University of Washington, Seattle, WA, USA
| | - David L Suskind
- Division of Gastroenterology and Hepatology, Seattle Children's Hospital
| |
Collapse
|
46
|
Patients' views on fecal microbiota transplantation: an acceptable therapeutic option in inflammatory bowel disease? Eur J Gastroenterol Hepatol 2017; 29:322-330. [PMID: 27879485 DOI: 10.1097/meg.0000000000000783] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
BACKGROUND Fecal microbiota transplantation (FMT) represents a new therapeutic option that has been studied in two randomized-controlled trials in ulcerative colitis patients. Our study aimed to identify patients' views on the use of this novel therapeutic approach. METHODS Using an anonymous questionnaire, we obtained data from 574 inflammatory bowel disease (IBD) patients on their knowledge and willingness to undergo FMT. RESULTS A large proportion of IBD patients (53.5%) are unaware that FMT is a therapeutic option in Clostridium difficile infection and potentially IBD. More responders preferred FMT (31.5%) to a study with a new medication (28.9%), although the difference was not significant (P=0.37), and the preferred way of transplantation was colonoscopy (49.7%). In all, 38.3% preferred a family member as a donor, but there was fear about the procedure (41.5% mentioned fear of infectious diseases, 26.5% expressed disgust). The knowledge of successful FMT treatment in other patients was important for 82.2% of responders and for 50.7%, a discussion with a specialist would likely change their opinion about FMT. CONCLUSION FMT represents a therapeutic procedure that is of interest for IBD patients. As FMT has been receiving increasing interest as an alternative treatment in IBD and more studies on FMT in IBD are being carried out, it is important to learn about the knowledge, attitude, and preferences of patients to provide better education to patients on this topic. However, there are reservations because of the fact that data on the benefits of FMT in IBD are controversial and several limitations exist on the use of FMT in IBD.
Collapse
|
47
|
Scaldaferri F, Pecere S, Petito V, Zambrano D, Fiore L, Lopetuso LR, Schiavoni E, Bruno G, Gerardi V, Laterza L, Pizzoferrato M, Ianiro G, Stojanovic J, Poscia A, Papa A, Paroni Sterbini F, Sanguinetti M, Masucci L, Cammarota G, Gasbarrini A. Efficacy and Mechanisms of Action of Fecal Microbiota Transplantation in Ulcerative Colitis: Pitfalls and Promises From a First Meta-Analysis. Transplant Proc 2017; 48:402-7. [PMID: 27109966 DOI: 10.1016/j.transproceed.2015.12.040] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Accepted: 12/30/2015] [Indexed: 12/25/2022]
Abstract
BACKGROUND Inflammatory bowel disease (IBD) is the results of a chronic inflammatory process deriving from disequilibrium between self-microbiota composition and immune response. METHODS New evidence, coming from Clostridium difficile infection, clearly showed that active and powerful modulation of microbiota composition by fecal microbiota composition (FMT) is safe, easy to perform, and efficacious, opening new frontiers in gastrointestinal and extra-intestinal diseases. FMT has been proposed also for IBD as well as other non-gastrointestinal conditions related to intestinal microbiota dysfunctions, with good preliminary data. RESULTS In this setting, ulcerative colitis (UC) represents one of the most robust potential indications for FMT after C difficile colitis. CONCLUSIONS In the present review, we focus on FMT and its application on ulcerative colitis, clarifying mechanisms of actions and efficacy data, trough completion of a meta-analysis on available randomized, controlled trial data in UC. Because microbiota is so crucially involved in this topic, a short review of microbial alterations in UC will also be performed.
Collapse
Affiliation(s)
- F Scaldaferri
- Institute of Special Pathology, Gastroenterology Division, Catholic University of Sacred Hearth, Rome, Italy.
| | - S Pecere
- Institute of Special Pathology, Gastroenterology Division, Catholic University of Sacred Hearth, Rome, Italy
| | - V Petito
- Institute of Special Pathology, Gastroenterology Division, Catholic University of Sacred Hearth, Rome, Italy
| | - D Zambrano
- Institute of Special Pathology, Gastroenterology Division, Catholic University of Sacred Hearth, Rome, Italy
| | - L Fiore
- Institute of Special Pathology, Gastroenterology Division, Catholic University of Sacred Hearth, Rome, Italy
| | - L R Lopetuso
- Institute of Special Pathology, Gastroenterology Division, Catholic University of Sacred Hearth, Rome, Italy
| | - E Schiavoni
- Institute of Special Pathology, Gastroenterology Division, Catholic University of Sacred Hearth, Rome, Italy
| | - G Bruno
- Institute of Special Pathology, Gastroenterology Division, Catholic University of Sacred Hearth, Rome, Italy
| | - V Gerardi
- Institute of Special Pathology, Gastroenterology Division, Catholic University of Sacred Hearth, Rome, Italy
| | - L Laterza
- Institute of Special Pathology, Gastroenterology Division, Catholic University of Sacred Hearth, Rome, Italy
| | - M Pizzoferrato
- Institute of Special Pathology, Gastroenterology Division, Catholic University of Sacred Hearth, Rome, Italy
| | - G Ianiro
- Institute of Special Pathology, Gastroenterology Division, Catholic University of Sacred Hearth, Rome, Italy
| | - J Stojanovic
- Institute of Hygiene, Catholic University of Sacred Hearth, Rome, Italy
| | - A Poscia
- Institute of Hygiene, Catholic University of Sacred Hearth, Rome, Italy
| | - A Papa
- Institute of Special Pathology, Gastroenterology Division, Catholic University of Sacred Hearth, Rome, Italy
| | - F Paroni Sterbini
- Institute of Microbiology, Catholic University of Sacred Hearth, Rome, Italy
| | - M Sanguinetti
- Institute of Microbiology, Catholic University of Sacred Hearth, Rome, Italy
| | - L Masucci
- Institute of Microbiology, Catholic University of Sacred Hearth, Rome, Italy
| | - G Cammarota
- Institute of Special Pathology, Gastroenterology Division, Catholic University of Sacred Hearth, Rome, Italy
| | - A Gasbarrini
- Institute of Special Pathology, Gastroenterology Division, Catholic University of Sacred Hearth, Rome, Italy
| |
Collapse
|
48
|
|
49
|
Liu S. The Development of Our Organ of Other Kinds-The Gut Microbiota. Front Microbiol 2016; 7:2107. [PMID: 28066404 PMCID: PMC5179505 DOI: 10.3389/fmicb.2016.02107] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Accepted: 12/13/2016] [Indexed: 01/31/2023] Open
Affiliation(s)
- Shirong Liu
- Department of Neurology, Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School Boston, MA, USA
| |
Collapse
|
50
|
Link A, Lachmund T, Schulz C, Weigt J, Malfertheiner P. Endoscopic peroral jejunal fecal microbiota transplantation. Dig Liver Dis 2016; 48:1336-1339. [PMID: 27575658 DOI: 10.1016/j.dld.2016.08.110] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Revised: 07/17/2016] [Accepted: 08/03/2016] [Indexed: 12/11/2022]
Abstract
BACKGROUND Fecal microbiota transplantation (FMT) is a valuable treatment modality for recurrent Clostridium difficile (C. difficile) colitis. Multiple questions including the best delivery route and volume remain unanswered. Here, we report a case series of high-volume FMT using endoscopic jejunal application route. METHODS In prospective observational study, FMT was performed using fresh specimen from healthy unrelated donors to the patients with recurrent or refractory C. difficile colitis. Selection of the route was based on the patient's preferences. Specimens of at least 50g were dissolved in 500ml of electrolyte solution and administered using endoscope directly in jejunum. RESULTS All procedures led to cure of C. difficile colitis. With exception of one case the procedure was well tolerated. In two cases, we observed FMT-reflux into the stomach despite deep jejunal application and in single case the FMT-reflux led to tracheal aspiration and severe pneumonia. CONCLUSIONS High-volume FMT via endoscopic jejunal route is an effective treatment option that is well tolerated and easy to perform. Nevertheless, aspiration is potential life-threatening event that needs to be kept in mind during the FMT-procedure.
Collapse
Affiliation(s)
- Alexander Link
- Department of Gastroenterology, Hepatology and Infectious Diseases, Otto-von-Guericke University, Magdeburg, Germany.
| | - Tim Lachmund
- Department of Gastroenterology, Hepatology and Infectious Diseases, Otto-von-Guericke University, Magdeburg, Germany
| | - Christian Schulz
- Department of Gastroenterology, Hepatology and Infectious Diseases, Otto-von-Guericke University, Magdeburg, Germany
| | - Jochen Weigt
- Department of Gastroenterology, Hepatology and Infectious Diseases, Otto-von-Guericke University, Magdeburg, Germany
| | - Peter Malfertheiner
- Department of Gastroenterology, Hepatology and Infectious Diseases, Otto-von-Guericke University, Magdeburg, Germany
| |
Collapse
|