151
|
Chen J, Zaman A, Ramakrishna B, Olesen SW. Stool Banking for Fecal Microbiota Transplantation: Methods and Operations at a Large Stool Bank. Front Cell Infect Microbiol 2021; 11:622949. [PMID: 33937092 PMCID: PMC8082449 DOI: 10.3389/fcimb.2021.622949] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Accepted: 03/22/2021] [Indexed: 12/15/2022] Open
Abstract
Objectives Fecal microbiota transplantation (FMT) is a recommended therapy for recurrent Clostridioides difficile infection and is being investigated as a potential therapy for dozens of microbiota-mediated indications. Stool banks centralize FMT donor screening and FMT material preparation with the goal of expanding access to FMT material while simultaneously improving its safety, quality, and convenience. Although there are published consensuses on donor screening guidelines, there are few reports about the implementation of those guidelines in functioning stool banks. Methods To help inform consensus standards with data gathered from real-world settings and, in turn, to improve patient care, here we describe the general methodology used in 2018 by OpenBiome, a large stool bank, and its outputs in that year. Results In 2018, the stool bank received 7,536 stool donations from 210 donors, a daily average of 20.6 donations, and processed 4,271 of those donations into FMT preparations. The median time a screened and enrolled stool donor actively donated stool was 5.8 months. The median time between the manufacture of an FMT preparation and its shipment to a hospital or physician was 8.9 months. Half of the stool bank's partner hospitals and physicians ordered an average of 0.75 or fewer FMT preparations per month. Conclusions Further knowledge sharing should help inform refinements of stool banking guidelines and best practices.
Collapse
|
152
|
Mustansir Dawoodbhoy F, Patel BK, Patel K, Bhatia M, Lee CN, Moochhala SM. Gut Microbiota Dysbiosis as a Target for Improved Post-Surgical Outcomes and Improved Patient Care: A Review of Current Literature. Shock 2021; 55:441-454. [PMID: 32881759 DOI: 10.1097/shk.0000000000001654] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Critical illness results in significant changes in the human gut microbiota, leading to the breakdown of the intestinal barrier function, which plays a role in the pathogenesis of multiple organ dysfunction. Patients with sepsis/acute respiratory distress syndrome (ARDS) have a profoundly distorted intestinal microbiota rhythm, which plays a considerable role in the development of gut-derived infections and intestinal dysbiosis. Despite recent medical developments, postsurgical complications are associated with a high morbidity and mortality rate. Bacterial translocation, which is the movement of bacteria and bacterial products across the intestinal barrier, was shown to be a mechanism behind sepsis. Current research is focusing on a solution by addressing significant factors that contribute to intestinal dysbiosis, which subsequently leads to multiple organ failure and, thus, mortality. It may, however, be challenging to manipulate the microbiota in critically ill patients for enhanced therapeutic gain. Probiotic manipulation is advantageous for maintaining the gut-barrier defense and for modulating the immune response. Based on available published research, this review aims to address the application of potential strategies in the intensive care unit, supplemented with current therapeutics by the administration of probiotics, prebiotics, and fecal microbiota transplant, to reduce post-surgical complications of sepsis/ARDS in critically ill patients.
Collapse
Affiliation(s)
| | | | - Kadamb Patel
- School of Applied Sciences, Temasek Polytechnic, Singapore
| | - Madhav Bhatia
- Department of Pathology and Biomedical Science, University of Otago Christchurch, Christchurch, New Zealand
| | - Chuen Neng Lee
- Department of Surgery, National University of Singapore, Singapore
| | | |
Collapse
|
153
|
Schmidt EKA, Raposo PJF, Madsen KL, Fenrich KK, Kabarchuk G, Fouad K. What Makes a Successful Donor? Fecal Transplant from Anxious-Like Rats Does Not Prevent Spinal Cord Injury-Induced Dysbiosis. BIOLOGY 2021; 10:254. [PMID: 33804928 PMCID: PMC8063845 DOI: 10.3390/biology10040254] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 03/16/2021] [Accepted: 03/23/2021] [Indexed: 12/12/2022]
Abstract
Spinal cord injury (SCI) causes gut dysbiosis and an increased prevalence of depression and anxiety. Previous research showed a link between these two consequences of SCI by using a fecal transplant from healthy rats which prevented both SCI-induced microbiota changes and the subsequent development of anxiety-like behaviour. However, whether the physical and mental state of the donor are important factors in the efficacy of FMT therapy after SCI remains unknown. In the present study, rats received a fecal transplant following SCI from uninjured donors with increased baseline levels of anxiety-like behaviour and reduced proportion of Lactobacillus in their stool. This fecal transplant increased intestinal permeability, induced anxiety-like behaviour, and resulted in minor but long-term alterations in the inflammatory state of the recipients compared to vehicle controls. There was no significant effect of the fecal transplant on motor recovery in rehabilitative training, suggesting that anxiety-like behaviour did not affect the motivation to participate in rehabilitative therapy. The results of this study emphasize the importance of considering both the microbiota composition and the mental state of the donor for fecal transplants following spinal cord injury.
Collapse
Affiliation(s)
- Emma K. A. Schmidt
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB T6G 2R3, Canada; (E.K.A.S.); (K.K.F.); (G.K.)
| | - Pamela J. F. Raposo
- Faculty of Rehabilitation Medicine, University of Alberta, Edmonton, AB T6G 2R3, Canada;
- Department of Physical Therapy, University of Alberta, Edmonton, AB T6G 2R3, Canada
| | - Karen L. Madsen
- Division of Gastroenterology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2R3, Canada;
| | - Keith K. Fenrich
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB T6G 2R3, Canada; (E.K.A.S.); (K.K.F.); (G.K.)
- Faculty of Rehabilitation Medicine, University of Alberta, Edmonton, AB T6G 2R3, Canada;
| | - Gillian Kabarchuk
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB T6G 2R3, Canada; (E.K.A.S.); (K.K.F.); (G.K.)
| | - Karim Fouad
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB T6G 2R3, Canada; (E.K.A.S.); (K.K.F.); (G.K.)
- Faculty of Rehabilitation Medicine, University of Alberta, Edmonton, AB T6G 2R3, Canada;
- Department of Physical Therapy, University of Alberta, Edmonton, AB T6G 2R3, Canada
| |
Collapse
|
154
|
Olesen SW, Gerardin Y. Re-Evaluating the Evidence for Faecal Microbiota Transplantation 'Super-Donors' in Inflammatory Bowel Disease. J Crohns Colitis 2021; 15:453-461. [PMID: 32808030 DOI: 10.1093/ecco-jcc/jjaa170] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
BACKGROUND Faecal microbiota transplantation [FMT] is a recommended treatment for recurrent Clostridioides difficile infection, and there is promise that FMT may be effective for conditions such as inflammatory bowel disease [IBD]. Previous FMT clinical trials have considered the possibility of a 'donor effect', that is, that FMT material from different donors has different clinical efficacies. METHODS Here we re-evaluate evidence for donor effects in published FMT clinical trials for IBD. RESULTS In ten of 12 published studies, no statistically significant donor effect was detected when rigorously re-evaluating the original analyses. One study showed statistically significant separation of microbiota composition of pools of donor stool when stratified by patient outcome. One study reported a significant effect but did not have underlying data available for re-evaluation. When quantifying the uncertainty on the magnitude of the donor effect, confidence intervals were large, including both zero donor effects and very substantial donor effects. CONCLUSION Although we found very little evidence for donor effects, the existing data cannot rule out the possibility that donor effects are clinically important. Large clinical trials prospectively designed to detect donor effects are probably needed to determine if donor effects are clinically relevant for IBD.
Collapse
Affiliation(s)
- Scott W Olesen
- OpenBiome, Cambridge, MA, USA.,Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | | |
Collapse
|
155
|
Wu W, Shen N, Luo L, Deng Z, Chen J, Tao Y, Mo X, Cao Q. Fecal microbiota transplantation before hematopoietic stem cell transplantation in a pediatric case of chronic diarrhea with a FOXP3 mutation. Pediatr Neonatol 2021; 62:172-180. [PMID: 33358585 DOI: 10.1016/j.pedneo.2020.11.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2020] [Revised: 08/23/2020] [Accepted: 11/27/2020] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND Immune dysregulation, polyendocrinopathy, enteropathy, X-linked (IPEX) syndrome is a rare disorder caused by mutation of the forkhead box protein 3 (FOXP3) gene, often leading to intractable and life-threatening diarrhea. Fecal microbiota transplantation (FMT), has been regarded in recent years as an available approach to reconstruct disrupted gut microbiome and successfully used to attenuates diarrhea induced by different underlying diseases. Therefore, FMT may have curative potential on the symptoms of enteropathy in patients with IPEX syndrome. METHODS Physical and laboratory examinations were performed, and clinical data were collected. FMT was administered via frozen fecal microbial solution, and the fecal microbiota composition was analyzed using 16S rDNA sequencing before and after FMT. RESULTS The patient was diagnosed with IPEX syndrome with a mutation detected in the FOXP3 gene, which was identified as c.767T > C (p.M256T). He presented with recurrent watery diarrhea and respiratory infections after birth and developed a significant failure to thrive. Disturbances in the gut microbiota composition and marked decreased bacterial diversity were observed to be involved in the persistent and refractory diarrhea. After receiving FMT treatment, the patient responded with remission of the diarrhea without apparent side effects. His stool output significantly decreased, corresponding to increased microbial diversity and modification of his microbiota composition. The patient finally achieved full recovery after hematopoietic stem cell transplantation (HSCT). CONCLUSION Our data suggest an association between the gut microbiota and clinical symptoms of patient with IPEX syndrome and demonstrate FMT as an alternative therapy for severe diarrhea unresponsive to routine therapy in these patients.
Collapse
Affiliation(s)
- Wenyan Wu
- Department of Infectious Diseases, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Nan Shen
- Department of Infectious Diseases, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lijuan Luo
- Department of Infectious Diseases, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhaohui Deng
- Department of Gastroenterology, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jing Chen
- Department of Hematology/Oncology, Shanghai Children's Medical Center, Shanghai Jiao Tong University, Shanghai, China
| | - Yue Tao
- The Laboratory of Pediatric Infectious Diseases, Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Xi Mo
- The Laboratory of Pediatric Infectious Diseases, Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Qing Cao
- The Laboratory of Pediatric Infectious Diseases, Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
156
|
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
|
157
|
Lee JJ, Yong D, Suk KT, Kim DJ, Woo HJ, Lee SS, Kim BS. Alteration of Gut Microbiota in Carbapenem-Resistant Enterobacteriaceae Carriers during Fecal Microbiota Transplantation According to Decolonization Periods. Microorganisms 2021; 9:microorganisms9020352. [PMID: 33578974 PMCID: PMC7916679 DOI: 10.3390/microorganisms9020352] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 02/05/2021] [Accepted: 02/08/2021] [Indexed: 02/06/2023] Open
Abstract
Fecal microbiota transplantation (FMT) has been suggested as an alternative therapeutic option to decolonize carbapenem-resistant Enterobacteriaceae (CRE). However, the analysis of gut microbiota alteration in CRE carriers during FMT is still limited. Here, gut microbiota changes in CRE carriers were evaluated during FMT according to decolonization periods. The decolonization of 10 CRE carriers was evaluated after FMT, using serial consecutive rectal swab cultures. Alterations of gut microbiota before and after FMT (56 serial samples) were analyzed using high-throughput sequencing. The decolonization rates of CRE carriers were 40%, 50%, and 90% within 1, 3 and 5 months after initial FMT, respectively. Gut microbiota significantly changed after FMT (p = 0.003). Microbiota alteration was different between the early decolonization carriers (EDC) and late decolonization carriers (LDC). Microbiota convergence in carriers to donors was detected in EDC within 4 weeks, and keystone genera within the Bacteroidetes were found in the gut microbiota of EDC before FMT. The relative abundance of Klebsiella was lower in EDC than in LDC, before and after FMT. Our results indicate that FMT is a potential option for CRE decolonization. The gut microbiota of CRE carriers could be used to predict decolonization timing after FMT, and determine repeated FMT necessity.
Collapse
Affiliation(s)
- Jin-Jae Lee
- Department of Life Science and Multidisciplinary Genome Institute, Hallym University, Chuncheon 24252, Korea;
- Institute for Liver and Digestive Diseases, Hallym University, Chuncheon 24252, Korea; (K.T.S.); (D.J.K.)
| | - Dongeun Yong
- Department of Laboratory Medicine and Research Institute of Bacterial Resistance, Yonsei University College of Medicine, Seoul 03722, Korea;
| | - Ki Tae Suk
- Institute for Liver and Digestive Diseases, Hallym University, Chuncheon 24252, Korea; (K.T.S.); (D.J.K.)
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, Hallym University, Chuncheon Sacred Heart Hospital, Chuncheon 24253, Korea
| | - Dong Joon Kim
- Institute for Liver and Digestive Diseases, Hallym University, Chuncheon 24252, Korea; (K.T.S.); (D.J.K.)
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, Hallym University, Chuncheon Sacred Heart Hospital, Chuncheon 24253, Korea
| | - Heung-Jeong Woo
- Department of Internal Medicine, Division of Infectious Diseases, Hallym University Dongtan Sacred Heart Hospital, Hallym University College of Medicine, Hwaseong 18450, Korea;
| | - Seung Soon Lee
- Institute for Liver and Digestive Diseases, Hallym University, Chuncheon 24252, Korea; (K.T.S.); (D.J.K.)
- Department of Internal Medicine, Division of Infectious Diseases, Hallym University Chuncheon Sacred Heart Hospital, Hallym University College of Medicine, Chuncheon 24252, Korea
- Correspondence: (S.S.L.); (B.-S.K.)
| | - Bong-Soo Kim
- Department of Life Science and Multidisciplinary Genome Institute, Hallym University, Chuncheon 24252, Korea;
- Institute for Liver and Digestive Diseases, Hallym University, Chuncheon 24252, Korea; (K.T.S.); (D.J.K.)
- Correspondence: (S.S.L.); (B.-S.K.)
| |
Collapse
|
158
|
Saha S, Khanna S. Stool banking for fecal microbiota transplantation: ready for prime time? Hepatobiliary Surg Nutr 2021; 10:110-112. [PMID: 33575297 DOI: 10.21037/hbsn-20-371] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2020] [Accepted: 04/01/2020] [Indexed: 01/18/2023]
Affiliation(s)
- Srishti Saha
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA
| | - Sahil Khanna
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
159
|
Gheorghe CE, Ritz NL, Martin JA, Wardill HR, Cryan JF, Clarke G. Investigating causality with fecal microbiota transplantation in rodents: applications, recommendations and pitfalls. Gut Microbes 2021; 13:1941711. [PMID: 34328058 PMCID: PMC8331043 DOI: 10.1080/19490976.2021.1941711] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 06/02/2021] [Accepted: 06/04/2021] [Indexed: 02/04/2023] Open
Abstract
In recent years, studies investigating the role of the gut microbiota in health and diseases have increased enormously - making it essential to deepen and question the research methodology employed. Fecal microbiota transplantation (FMT) in rodent studies (either from human or animal donors) allows us to better understand the causal role of the intestinal microbiota across multiple fields. However, this technique lacks standardization and requires careful experimental design in order to obtain optimal results. By comparing several studies in which rodents are the final recipients of FMT, we summarize the common practices employed. In this review, we document the limitations of this method and highlight different parameters to be considered while designing FMT Studies. Standardizing this method is challenging, as it differs according to the research topic, but avoiding common pitfalls is feasible. Several methodological questions remain unanswered to this day and we offer a discussion on issues to be explored in future studies.
Collapse
Affiliation(s)
- Cassandra E. Gheorghe
- Department of Psychiatry and Neurobehavioral Science, University College Cork, Cork, Ireland
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Nathaniel L. Ritz
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Jason A. Martin
- Department of Psychiatry and Neurobehavioral Science, University College Cork, Cork, Ireland
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Hannah R. Wardill
- Precision Medicine, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, Australia
- Adelaide Medical School, the University of Adelaide, Adelaide, Australia
| | - John F. Cryan
- Department of Psychiatry and Neurobehavioral Science, University College Cork, Cork, Ireland
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Gerard Clarke
- Department of Psychiatry and Neurobehavioral Science, University College Cork, Cork, Ireland
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- INFANT Research Centre, University College Cork, Cork, Ireland
| |
Collapse
|
160
|
Utay NS, Monczor AN, Somasunderam A, Lupo S, Jiang ZD, Alexander AS, Finkelman M, Vigil KJ, Lake JE, Hanson B, DuPont HL, Arduino RC. Evaluation of Six Weekly Oral Fecal Microbiota Transplants in People with HIV. Pathog Immun 2020; 5:364-381. [PMID: 33501400 PMCID: PMC7815055 DOI: 10.20411/pai.v5i1.388] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Accepted: 11/13/2020] [Indexed: 12/12/2022] Open
Abstract
Background Reduced microbiota diversity (dysbiosis) in people with HIV (PWH) likely contributes to inflammation, a driver of morbidity and mortality. We aimed to evaluate the safety and tolerability of 6 weekly oral fecal microbiota transplants (FMT) administered to reverse this dysbiosis. Methods Six PWH on suppressive antiretroviral therapy (ART) received 6 weekly doses of lyophilized fecal microbiota product from healthy donors. Shotgun sequencing on stool before, after last FMT, and 20 weeks thereafter was performed. Inflammation and gut permeability biomarkers were measured. Results Median age at week 0 was 39 years, CD4+ T cell count 496 cells/mm3, HIV RNA levels <20 copies/mL. FMT was safe and well-tolerated. α diversity increased in 4 participants from weeks 0 to 6, including the 3 with the lowest α diversity at week 0. At week 26, α diversity more closely resembled week 0 than week 6 in these 4 participants. Metagenomic analysis showed no consistent changes across all participants. One participant had high gut permeability and inflammation biomarker levels and low α diversity that improved between weeks 0 and 6 with a shift in distribution. Conclusions Weekly FMT was safe and well-tolerated. α diversity increased in participants with the lowest baseline α diversity during the treatment period. Future randomized, controlled trials of FMT should consider evaluating PWH with greater inflammation, gut damage, or dysbiosis as this population may be most likely to show a significant response.ClinicalTrials.gov Identifier: NCT03329560.
Collapse
Affiliation(s)
- Netanya S Utay
- Division of General Medicine, Department of Internal Medicine, McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, Texas.,Kelsey Research Foundation, Houston, Texas
| | - Ana N Monczor
- Division of Infectious Diseases, Department of Internal Medicine, McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, Texas
| | - Anoma Somasunderam
- Division of General Medicine, Department of Internal Medicine, McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, Texas
| | - Sofia Lupo
- Division of Infectious Diseases, Department of Internal Medicine, McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, Texas
| | - Zhi-Dong Jiang
- School of Public Health at The University of Texas Health Science Center at Houston, Houston, Texas
| | | | | | - Karen J Vigil
- Division of Infectious Diseases, Department of Internal Medicine, McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, Texas
| | - Jordan E Lake
- Division of Infectious Diseases, Department of Internal Medicine, McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, Texas
| | - Blake Hanson
- School of Public Health at The University of Texas Health Science Center at Houston, Houston, Texas
| | - Herbert L DuPont
- Kelsey Research Foundation, Houston, Texas.,School of Public Health at The University of Texas Health Science Center at Houston, Houston, Texas
| | - Roberto C Arduino
- Division of Infectious Diseases, Department of Internal Medicine, McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, Texas
| |
Collapse
|
161
|
Ebbers M, Hemmer CJ, Müller-Hilke B, Reisinger EC. Immunotherapy and vaccination against infectious diseases. Wien Klin Wochenschr 2020; 133:714-720. [PMID: 33326055 PMCID: PMC7738774 DOI: 10.1007/s00508-020-01746-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Accepted: 09/07/2020] [Indexed: 11/29/2022]
Abstract
Due to the overuse of antibiotics, infections, in particular those caused by multidrug-resistant bacteria, are becoming more and more frequent. Despite the worldwide introduction of antibiotic therapy, vaccines and constant improvements in hygiene, the burden of multidrug-resistant bacterial infections is increasing and is expected to rise in the future. The development of monoclonal therapeutic antibodies and specific immunomodulatory drugs represent new treatment options in the fight against infectious diseases. This article provides a brief overview of recent advances in immunomodulatory therapy and other strategies in the treatment of infectious disease.
Collapse
Affiliation(s)
- Meinolf Ebbers
- Department of Tropical Medicine and Infectious Diseases, Rostock University Medical Center, Ernst-Heydemann-Str. 6, 18057, Rostock, Germany.,Core Facility for Cell Sorting and Cell Analysis, Rostock University Medical Center, Schillingallee 70, 18057, Rostock, Germany
| | - Christoph J Hemmer
- Department of Tropical Medicine and Infectious Diseases, Rostock University Medical Center, Ernst-Heydemann-Str. 6, 18057, Rostock, Germany
| | - Brigitte Müller-Hilke
- Core Facility for Cell Sorting and Cell Analysis, Rostock University Medical Center, Schillingallee 70, 18057, Rostock, Germany
| | - Emil C Reisinger
- Department of Tropical Medicine and Infectious Diseases, Rostock University Medical Center, Ernst-Heydemann-Str. 6, 18057, Rostock, Germany.
| |
Collapse
|
162
|
Chen L, Li J, Zhu W, Kuang Y, Liu T, Zhang W, Chen X, Peng C. Skin and Gut Microbiome in Psoriasis: Gaining Insight Into the Pathophysiology of It and Finding Novel Therapeutic Strategies. Front Microbiol 2020; 11:589726. [PMID: 33384669 PMCID: PMC7769758 DOI: 10.3389/fmicb.2020.589726] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 11/23/2020] [Indexed: 12/11/2022] Open
Abstract
Psoriasis affects the health of myriad populations around the world. The pathogenesis is multifactorial, and the exact driving factor remains unclear. This condition arises from the interaction between hyperproliferative keratinocytes and infiltrating immune cells, with poor prognosis and high recurrence. Better clinical treatments remain to be explored. There is much evidence that alterations in the skin and intestinal microbiome play an important role in the pathogenesis of psoriasis, and restoration of the microbiome is a promising preventive and therapeutic strategy for psoriasis. Herein, we have reviewed recent studies on the psoriasis-related microbiome in an attempt to confidently identify the “core” microbiome of psoriasis patients, understand the role of microbiome in the pathogenesis of psoriasis, and explore new therapeutic strategies for psoriasis through microbial intervention.
Collapse
Affiliation(s)
- Lihui Chen
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China.,Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China.,Hunan Key Laboratory of Pharmacogenetics, Institute of Clinical Pharmacology, Central South University, Changsha, China.,Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, China.,Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, China
| | - Jie Li
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China.,Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, China.,Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, China
| | - Wu Zhu
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China.,Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, China.,Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, China
| | - Yehong Kuang
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China.,Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, China.,Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, China
| | - Tao Liu
- Central Laboratory, Shenzhen Center for Chronic Disease Control and Prevention, Shenzhen, China
| | - Wei Zhang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China.,Hunan Key Laboratory of Pharmacogenetics, Institute of Clinical Pharmacology, Central South University, Changsha, China
| | - Xiang Chen
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China.,Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, China.,Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, China
| | - Cong Peng
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China.,Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, China.,Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, China
| |
Collapse
|
163
|
Basson AR, Zhou Y, Seo B, Rodriguez-Palacios A, Cominelli F. Autologous fecal microbiota transplantation for the treatment of inflammatory bowel disease. Transl Res 2020; 226:1-11. [PMID: 32585148 PMCID: PMC7308243 DOI: 10.1016/j.trsl.2020.05.008] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 05/20/2020] [Accepted: 05/23/2020] [Indexed: 12/12/2022]
Abstract
The term autologous fecal microbiota transplantation (a-FMT) refers herein to the use of one's feces during a healthy state for later use to restore gut microbial communities after perturbations. Generally, heterologous fecal microbiota transplantation (h-FMT), where feces from a ``healthy" donor is transplanted into a person with illness, has been used to treat infectious diseases such as recurrent Clostridioides difficile infection (CDI), with cure rates of up to 90%. In humans, due to limited response to medicines, h-FMT has become a hallmark intervention to treat CDI. Extrapolating the benefits from CDI, h-FMT has been attempted in various diseases, including inflammatory bowel disease (IBD), but clinical response has been variable and less effective (ranging between 24% and 50%). Differences in h-FMT clinical response could be because CDI is caused by a Clostridial infection, whereas IBD is a complex, microbiome-driven immunological inflammatory disorder that presents predominantly within the gut wall of genetically-susceptible hosts. FMT response variability could also be due to differences in microbiome composition between donors, recipients, and within individuals, which vary with diet, and environments, across regions. While donor selection has emerged as a key factor in FMT success, the use of heterologous donor stool still places the recipient at risk of exposure to infectious/pathogenic microorganisms. As an implementable solution, herein we review the available literature on a-FMT, and list some considerations on the benefits of a-FMT for IBD.
Collapse
Key Words
- a-fmt, autologous fecal microbiota transplantation
- cd, crohn's disease
- cdi, clostridium difficile infection
- ci, confidence interval
- fmt, fecal microbiota transplantation
- hgm, human gut microbiota
- h-fmt, heterologous fecal microbiota transplantation
- ibd, inflammatory bowel disease
- ibs, irritable bowel syndrome
- rct, randomized controlled trial
- uc, ulcerative colitis
Collapse
Affiliation(s)
- Abigail R Basson
- Division of Gastroenterology & Liver Diseases, Case Western Reserve University School of Medicine, Cleveland, Ohio; Digestive Health Research Institute, University Hospitals Cleveland Medical Center, Cleveland, Ohio
| | - Yibing Zhou
- Division of Gastroenterology & Liver Diseases, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Brian Seo
- Division of Gastroenterology & Liver Diseases, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Alexander Rodriguez-Palacios
- Division of Gastroenterology & Liver Diseases, Case Western Reserve University School of Medicine, Cleveland, Ohio; Digestive Health Research Institute, University Hospitals Cleveland Medical Center, Cleveland, Ohio
| | - Fabio Cominelli
- Division of Gastroenterology & Liver Diseases, Case Western Reserve University School of Medicine, Cleveland, Ohio; Digestive Health Research Institute, University Hospitals Cleveland Medical Center, Cleveland, Ohio.
| |
Collapse
|
164
|
Holster S, Rode J, Bohr J, Kumawat AK, Veress G, Hultgren Hörnquist E, Brummer RJ, König J. Faecal microbiota transfer in patients with microscopic colitis - a pilot study in collagenous colitis. Scand J Gastroenterol 2020; 55:1454-1466. [PMID: 33142068 DOI: 10.1080/00365521.2020.1839544] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
OBJECTIVES Faecal microbiota transfer (FMT) consists of the infusion of donor faecal material into the intestine of patients with the aim to restore a disturbed gut microbiota. METHODS In this pilot study (NCT03275467), the effect of three repeated FMTs (day 0, two weeks, four weeks) was studied and followed up for six months in nine collagenous colitis (CC) patients, using two stool donors. RESULTS Five patients had an active disease at the time of baseline sampling. The primary endpoint (remission at six weeks, defined as <3 stools whereof <1 watery stool per day) was achieved by two of these patients, and by one at eight weeks. Overall, in all nine patients, FMT did not result in a significant reduction of watery stools, assessed by daily diary. However, diarrhoea (assessed by gastrointestinal symptom rating scale) was significantly improved at four (p = .038) and eight weeks (p = .038), indigestion at eight (p = .045) and 12 weeks (p = .006), disease-related worries at four (p = .027) and eight weeks (p = .027), and quality of life at six months (p = .009). FMT resulted in an increased number of lamina propria lymphocytes, possibly indicating an initial mucosal immune activation. No serious adverse events, no systemic effects, and no changes in faecal calprotectin and psychological symptoms were observed. CONCLUSIONS FMT is able to improve symptoms in a yet undefined subset of CC patients. Further studies could help to characterise this subset and to understand if these results can be generalised to all microscopic colitis patients.
Collapse
Affiliation(s)
- Savanne Holster
- Nutrition-Gut-Brain Interactions Research Centre, Faculty of Medicine and Health, School of Medical Sciences, Örebro University, Örebro, Sweden
| | - Julia Rode
- Nutrition-Gut-Brain Interactions Research Centre, Faculty of Medicine and Health, School of Medical Sciences, Örebro University, Örebro, Sweden
| | - Johan Bohr
- Department of Gastroenterology, Faculty of Medicine and Health, Örebro University, Örebro, Sweden, Örebro University Hospital, Örebro, Sweden
| | - Ashok Kumar Kumawat
- Cardiovascular Research Centre, Faculty of Medicine and Health, School of Medical Sciences, Örebro University, Örebro, Sweden
| | - Gábor Veress
- Department of Laboratory Medicine, Faculty for Medicine and Health, Örebro University, Örebro, Sweden, Örebro University Hospital, Örebro, Sweden
| | - Elisabeth Hultgren Hörnquist
- Nutrition-Gut-Brain Interactions Research Centre, Faculty of Medicine and Health, School of Medical Sciences, Örebro University, Örebro, Sweden
| | - Robert Jan Brummer
- Nutrition-Gut-Brain Interactions Research Centre, Faculty of Medicine and Health, School of Medical Sciences, Örebro University, Örebro, Sweden
| | - Julia König
- Nutrition-Gut-Brain Interactions Research Centre, Faculty of Medicine and Health, School of Medical Sciences, Örebro University, Örebro, Sweden
| |
Collapse
|
165
|
Vandeputte D. Personalized Nutrition Through The Gut Microbiota: Current Insights And Future Perspectives. Nutr Rev 2020; 78:66-74. [PMID: 33259623 DOI: 10.1093/nutrit/nuaa098] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
This narrative review discusses how to preserve or increase health through personalized nutritional products and services using microbiome data. In contrast to other reviews, which discuss this subject in the light of metabolic disorders and/or with a nutrition-affects-the-microbiota view, this review takes the perspective that the gut microbiota (GM) affects nutrition. Gut microbes affect host nutritional status through their role in energy harvest and nutrient availability. Consequently, GM modulation could contribute to fulfil nutritional requirements and in this way conquer malnutrition and disease. This review provides an overview of microbiota modulation methods that could be used to improve nutritional status as well as the personalization of these approaches. While some of these methods are immediately applicable, others require more development to assess their feasibility and safety.
Collapse
Affiliation(s)
- Doris Vandeputte
- Center for Microbiology, VIB, Leuven, Belgium.,Department of Microbiology, Immunology and Transplantation, Rega Institute, KU Leuven - University of Leuven, Leuven, Belgium.,Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| |
Collapse
|
166
|
Ross CN, Reveles KR. Feasibility of fecal microbiota transplantation via oral gavage to safely alter gut microbiome composition in marmosets. Am J Primatol 2020; 82:e23196. [PMID: 32970852 PMCID: PMC7679041 DOI: 10.1002/ajp.23196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 08/17/2020] [Accepted: 08/22/2020] [Indexed: 11/10/2022]
Abstract
Disruption of microbial communities within human hosts has been associated with infection, obesity, cognitive decline, cancer risk and frailty, suggesting that microbiome-targeted therapies may be an option for improving healthspan and lifespan. The objectives of this study were to determine the feasibility of delivering fecal microbiota transplants (FMTs) to marmosets via oral gavage and to evaluate if alteration of the gut microbiome post-FMT could be achieved. This was a prospective study of marmosets housed at the Barshop Institute for Longevity and Aging Studies in San Antonio, Texas. Eligible animals included healthy young adult males (age 2-5 years) with no recent medication use. Stool from two donors was combined and administered in 0.5 ml doses to five young recipients once weekly for 3 weeks. Safety outcomes and alterations in the gut microbiome composition via 16S ribosomal RNA sequencing were compared at baseline and monthly up to 6 months post-FMT. Overall, significant differences in the percent relative abundance was seen in FMT recipients at the phylum and family levels from baseline to 1 month and baseline to 6 months post-FMT. In permutational multivariate analysis of variance analyses, treatment status (donor vs. recipient) (p = .056) and time course (p = .019) predicted β diversity (p = .056). The FMT recipients did not experience any negative health outcomes over the course of the treatment. FMT via oral gavage was safe to administer to young adult marmosets. The marmoset microbiome may be altered by FMT; however, progressive changes in the microbiome are strongly driven by the host and its baseline microbiome composition.
Collapse
Affiliation(s)
- Corinna N Ross
- Department of Science and Mathematics, Texas A&M University San Antonio, San Antonio, Texas, USA
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, Texas, USA
- Barshop Institute for Longevity and Aging Research, University of Texas Health San Antonio, San Antonio, Texas, USA
| | - Kelly R Reveles
- College of Pharmacy, The University of Texas at Austin, Austin, Texas, USA
- Pharmacotherapy Education & Research Center, University of Texas Health San Antonio, San Antonio, Texas, USA
| |
Collapse
|
167
|
Kong L, Lloyd-Price J, Vatanen T, Seksik P, Beaugerie L, Simon T, Vlamakis H, Sokol H, Xavier RJ. Linking Strain Engraftment in Fecal Microbiota Transplantation With Maintenance of Remission in Crohn's Disease. Gastroenterology 2020; 159:2193-2202.e5. [PMID: 32860788 PMCID: PMC7725862 DOI: 10.1053/j.gastro.2020.08.045] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 07/02/2020] [Accepted: 08/21/2020] [Indexed: 02/07/2023]
Abstract
BACKGROUND & AIMS Crohn's disease (CD) is a chronic gastrointestinal disease resulting from the dysfunctional interplay between genetic susceptibility, the immune system, and commensal intestinal microbiota. Emerging evidence suggests that treatment by suppression of the immune response and replacement of the microbiota through fecal microbiota transplantation (FMT) is a promising approach for the treatment of CD. METHODS We obtained stool metagenomes from CD patients in remission and assessed gut microbiome composition before and after FMT at the species and strain levels. Longitudinal follow-up evaluation allowed us to identify the gain, loss, and strain replacement of specific species and link these events to the maintenance of remission in CD. RESULTS We found that FMT had a significant long-term effect on patient microbial compositions, although this was primarily driven by the engraftment of donor species, which remained at low abundance. Thirty-eight percent of FMT-driven changes were strain replacements, emphasizing the importance of detailed profiling methods, such as metagenomics. Several instances of long-term coexistence between donor and patient strains were also observed. Engraftment of some Actinobacteria, and engraftment or loss of Proteobacteria, were related to better disease outcomes in CD patients who received FMT, and transmission of Bacteroidetes was deleterious. CONCLUSIONS Our results suggest clades that may be beneficial to transmit/eliminate through FMT, and provide criteria that may help identify personalized FMT donors to more effectively maintain remission in CD patients. The framework established here creates a foundation for future studies centered around the application of FMT and defined microbial communities as a therapeutic approach for treating CD.
Collapse
Affiliation(s)
- Lingjia Kong
- Broad Institute of MIT and Harvard, Cambridge, MA, USA,Center for Computational and Integrative Biology and Department of Molecular Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | | | - Tommi Vatanen
- Broad Institute of MIT and Harvard, Cambridge, MA, USA,Liggins Institute, University of Auckland, Auckland, New Zealand
| | - Philippe Seksik
- Centre de Recherche Saint-Antoine, CRSA, AP-HP, Hôpital Saint Antoine, Service de Gastroenterologie, Sorbonne Université, Inserm, 75012, Paris, France.,Department of Gastroenterology, Saint Antoine Hospital, Assistance Publique-Hopitaux de Paris (APHP), 184 rue du Faubourg Saint-Antoine, 75571, Paris, CEDEX 12, France.,French Group of Fecal Transplantation (GFTF), Paris, France.,Paris Center for Microbiome Medicine (FHU PaCeMM), Paris, France
| | - Laurent Beaugerie
- Department of Gastroenterology, Saint Antoine Hospital, Assistance Publique-Hopitaux de Paris (APHP), 184 rue du Faubourg Saint-Antoine, 75571, Paris, CEDEX 12, France.,Paris Center for Microbiome Medicine (FHU PaCeMM), Paris, France
| | - Tabassome Simon
- Clinical Research Platform (URC-CRC-CRB), AP-HP Saint-Antoine Hospital, Paris, France.,Department of Clinical Pharmacology, APHP, Saint Antoine Hospital, Paris, France
| | - Hera Vlamakis
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Harry Sokol
- Centre de Recherche Saint-Antoine, Assistance Publique-Hôpitaux de Paris, Hôpital Saint Antoine, Service de Gastroenterologie, Sorbonne Université, INSERM 75012, Paris, France; Department of Gastroenterology, Saint Antoine Hospital, Assistance Publique-Hopitaux de Paris, Paris, France; French Group of Fecal Transplantation, Paris, France; Paris Center for Microbiome Medicine, Paris, France.
| | - Ramnik J. Xavier
- Broad Institute of MIT and Harvard, Cambridge, MA, USA,Center for Microbiome Informatics and Therapeutics, Massachusetts Institute of Technology, Cambridge, MA, USA,Center for Computational and Integrative Biology and Department of Molecular Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA,Correspondence:;
| |
Collapse
|
168
|
Olesen SW. Power calculations for detecting differences in efficacy of fecal microbiota donors. Contemp Clin Trials Commun 2020; 20:100674. [PMID: 33241161 PMCID: PMC7672275 DOI: 10.1016/j.conctc.2020.100674] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 10/27/2020] [Indexed: 12/26/2022] Open
Abstract
Fecal microbiota transplantation (FMT) is a recommended therapy for recurrent Clostridioides difficile infection and is being investigated as a potential therapy for dozens of other indications, notably inflammatory bowel disease. The immense variability in human stool, combined with anecdotal reports from FMT studies, have suggested the existence of “donor effects”, in which stool from some FMT donors is more efficacious than stool from other donors. In this study, simulated clinical trials were used to estimate the number of patients that would be required to detect donor effects under a variety of study designs. In most cases, reliable detection of donor effects required more than 100 patients treated with FMT. These results suggest that previous reports of donor effects need to be verified with results from large clinical trials and that patient biomarkers may be the most promising route to robustly identifying donor effects.
Collapse
Affiliation(s)
- Scott W Olesen
- OpenBiome, 2067 Massachusetts Avenue, Cambridge, MA, 02140, USA
| |
Collapse
|
169
|
Dang XF, Qing-Xi Wang, Yin Z, Sun L, Yang WH. Recurrence of moderate to severe ulcerative colitis after fecal microbiota transplantation treatment and the efficacy of re-FMT: a case series. BMC Gastroenterol 2020; 20:401. [PMID: 33243141 PMCID: PMC7691068 DOI: 10.1186/s12876-020-01548-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Accepted: 11/18/2020] [Indexed: 12/17/2022] Open
Abstract
Background
Ulcerative colitis (UC) is a chronic inflammatory bowel disease (IBD), the pathogenesis of which is complicated, and it is difficult to treat. In recent years, the emerging fecal microbiota transplantation (FMT) has shown good effects in UC treatment and is therefore accepted by increasing numbers of patients. Our hospital has carried out FMT since 2017, and has achieved good results in UC treatment. We have found in our clinical work that the efficacy of re-FMT after recurrence decreased. This is difference from reported literatures. In order to attract clinical attention, here we selected typical cases for analysis. Methods Among all UC patients who received FMT in our hospital, 12 patients with moderate to severe UC were selected. They all received multiple FMT and were followed up for 52 weeks. Besides, none of them had other underlying diseases. Colonoscopy images of patients were presentated, SCCAI and UCDAI were used assess the effect of FMT. Results On the whole, FMT has a significant effect on moderate to severe UC. Of the 12 patients, 11 (91.7%) achieved a clinical response, 9 (75.0%) achieved clinical remission, and only one patient did not respond to FMT treatment. However, 6 patients relapsed within 52 weeks after remission, with a recurrence rate of 54.5%. Four of the six relapsed patients received FMT again, but the efficacy of FMT after relapse was significantly lower than that of the initial FMT. Fortunately, compared to before the initial FMT treatment, the severity of the disease after relapse was significantly reduced. Conclusion FMT has a good effect on the relief of moderate to severe UC. However, the effect of FMT treatment after relapse is reduced. For patients who relapse after remission, the efficacy of FMT reapplication requires more experiments to verify.
Collapse
Affiliation(s)
- Xiao-Fei Dang
- Department of Clinical Microbiology, Medical Research & Laboratory Diagnostic Center, Jinan Central Hospital Affiliated to Shandong First Medical University, 105 Jiefang Road, Jinan, Shandong, China
| | - Qing-Xi Wang
- Department of Clinical Microbiology, Medical Research & Laboratory Diagnostic Center, Jinan Central Hospital Affiliated to Shandong First Medical University, 105 Jiefang Road, Jinan, Shandong, China
| | - Zhao Yin
- Gastroenterology, Jinan Central Hospital Affiliated to Shandong First Medical University, 105 Jiefang Road, Jinan, China
| | - Lin Sun
- Department of Clinical Microbiology, Medical Research & Laboratory Diagnostic Center, Jinan Central Hospital Affiliated to Shandong First Medical University, 105 Jiefang Road, Jinan, Shandong, China
| | - Wei-Hua Yang
- Department of Clinical Microbiology, Medical Research & Laboratory Diagnostic Center, Jinan Central Hospital Affiliated to Shandong First Medical University, 105 Jiefang Road, Jinan, Shandong, China.
| |
Collapse
|
170
|
Rodríguez C, Romero E, Garrido-Sanchez L, Alcaín-Martínez G, Andrade RJ, Taminiau B, Daube G, García-Fuentes E. MICROBIOTA INSIGHTS IN CLOSTRIDIUM DIFFICILE INFECTION AND INFLAMMATORY BOWEL DISEASE. Gut Microbes 2020; 12:1725220. [PMID: 32129694 PMCID: PMC7524151 DOI: 10.1080/19490976.2020.1725220] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Inflammatory bowel disease (IBD) is characterized by chronic intestinal inflammation that includes Crohn´s disease (CD) and ulcerative colitis (UC). Although the etiology is still unknown, some specific factors have been directly related to IBD, including genetic factors, abnormal intestinal immunity, and/or gut microbiota modifications. Recent findings highlight the primary role of the gut microbiota closely associated with a persistent inappropriate inflammatory response. This gut environment of dysbiosis in a susceptible IBD host can increasingly worsen and lead to colonization and infection with some opportunistic pathogens, especially Clostridium difficile. C. difficile is an intestinal pathogen considered the main cause of antibiotic-associated diarrhea and colitis and an important complication of IBD, which can trigger or worsen an IBD flare. Recent findings have highlighted the loss of bacterial cooperation in the gut ecosystem, as well as the pronounced intestinal dysbiosis, in patients suffering from IBD and concomitant C. difficile infection (CDI). The results of intestinal microbiota studies are still limited and often difficult to compare because of the variety of disease conditions. However, these data provide important clues regarding the main modifications and interrelations in the complicated gut ecosystem to better understand both diseases and to take advantage of the development of new therapeutic strategies. In this review, we analyze in depth the gut microbiota changes associated with both forms of IBD and CDI and their similarity with the dysbiosis that occurs in CDI. We also discuss the metabolic pathways that favor the proliferation or decrease in several important taxa directly related to the disease.
Collapse
Affiliation(s)
- C. Rodríguez
- Instituto de Investigación Biomédica de Málaga-IBIMA, Málaga, Spain,Unidad de Gestión Clínica de Aparato Digestivo, Hospital Universitario Virgen de la Victoria, Málaga, Spain,CONTACT C. Rodríguez Instituto de Investigación Biomédica de Málaga-IBIMA, Málaga, SpainUnidad de Gestión Clínica de Aparato Digestivo, Hospital Universitario Virgen de la Vitoria, Málaga, Spain
| | - E. Romero
- Instituto de Investigación Biomédica de Málaga-IBIMA, Málaga, Spain
| | - L. Garrido-Sanchez
- Instituto de Investigación Biomédica de Málaga-IBIMA, Málaga, Spain,Unidad de Gestión Clínica de Endocrinología y Nutrición, Hospital Universitario Virgen de la Victoria, Málaga, Spain
| | - G. Alcaín-Martínez
- Instituto de Investigación Biomédica de Málaga-IBIMA, Málaga, Spain,Unidad de Gestión Clínica de Aparato Digestivo, Hospital Universitario Virgen de la Victoria, Málaga, Spain
| | - RJ. Andrade
- Instituto de Investigación Biomédica de Málaga-IBIMA, Málaga, Spain,Unidad de Gestión Clínica de Aparato Digestivo, Hospital Universitario Virgen de la Victoria, Málaga, Spain,Department of Medicine and Dermatology, Universidad de Málaga, Málaga, Spain,Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Málaga, Spain
| | - B. Taminiau
- Fundamental and Applied Research for Animals & Health (FARAH), Department of Food Microbiology, Faculty of Veterinary Medicine, University of Liège, Liège, Belgium
| | - G. Daube
- Fundamental and Applied Research for Animals & Health (FARAH), Department of Food Microbiology, Faculty of Veterinary Medicine, University of Liège, Liège, Belgium
| | - E. García-Fuentes
- Instituto de Investigación Biomédica de Málaga-IBIMA, Málaga, Spain,Unidad de Gestión Clínica de Aparato Digestivo, Hospital Universitario Virgen de la Victoria, Málaga, Spain
| |
Collapse
|
171
|
Dale HF, Lied GA. Gut microbiota and therapeutic approaches for dysbiosis in irritable bowel syndrome: recent developments and future perspectives. Turk J Med Sci 2020; 50:1632-1641. [PMID: 32222124 PMCID: PMC7672353 DOI: 10.3906/sag-2002-57] [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] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Accepted: 03/22/2020] [Indexed: 11/19/2022] Open
Abstract
Increased knowledge regarding the implications of gut microbiota in irritable bowel syndrome (IBS) suggests that a disturbed intestinal microenvironment (dysbiosis) might promote the development and maintenance of IBS symptoms and affects several pathways in the pathology of this multifactorial disease. Accordingly, manipulation of the gut microbiota in order to improve IBS symptoms has evolved as a novel treatment strategy in the last decade. Several different approaches have been investigated in order to improve the gut microbiota composition. Dietary modifications including supplementation with fibers, prebiotics, and probiotics are shown to improve symptoms and composition of gut microbiota in IBS; however, the exact probiotic mixture beneficial for each individual remains to be identified. The use of antibiotics still needs confirmation, although promising results have been reported with use of rifaximin. Fecal microbiota transplantation (FMT) has recently gained a lot of attention, and several placebo-controlled trials investigating FMT obtain promising results regarding symptom reduction and gut microbiota manipulation in IBS. However, more data regarding long-term effects are needed before FMT can be integrated as a customized treatment for IBS in the clinical routine.
Collapse
Affiliation(s)
- Hanna Fjeldheim Dale
- Department of Clinical Medicine, Centre for Nutrition, University of Bergen, Bergen, Norway
- Department of National Centre of Functional Gastrointestinal Disorders, Haukeland University Hospital, Bergen, Norway
| | - Gülen Arslan Lied
- Department of Clinical Medicine, Centre for Nutrition, University of Bergen, Bergen, Norway
- Department of National Centre of Functional Gastrointestinal Disorders, Haukeland University Hospital, Bergen, Norway
| |
Collapse
|
172
|
Sherrill-Mix S, Connors K, Aldrovandi GM, Brenchley JM, Boucher C, Bushman FD, Collman RG, Dandekar S, Klatt NR, Lagenaur LA, Paredes R, Tachedjian G, Turpin JA, Landay AL, Ghosh M. A Summary of the Fifth Annual Virology Education HIV Microbiome Workshop. AIDS Res Hum Retroviruses 2020; 36:886-895. [PMID: 32777940 PMCID: PMC7869876 DOI: 10.1089/aid.2020.0121] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
In October of 2019, researchers and community members from around the world met at the NIH for the fifth annual International Workshop on Microbiome in HIV. New research was presented on the role of the microbiome on chronic inflammation and vaccine design, interactions of genetics, environment, sexual practice and HIV infection with the microbiome and the development and clinical trials of microbiome-based therapeutic approaches intended to decrease the probability of HIV acquisition/transmission or ameliorate sequelae of HIV. The keynote address by Dr. Jacques Ravel focused on his work on the vaginal microbiome and efforts to improve the analysis and resolution of microbiome data.
Collapse
Affiliation(s)
- Scott Sherrill-Mix
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Kaleigh Connors
- Department of Epidemiology, The George Washington University, Washington, District of Columbia, USA
| | - Grace M. Aldrovandi
- Department of Pediatrics, University of California, Los Angeles, Los Angeles, California, USA
| | | | - Charles Boucher
- Department of Virosciences, Erasmus Medical Center, Erasmus University Rotterdam, Rotterdam, the Netherlands
| | - Frederic D. Bushman
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Ronald G. Collman
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Satya Dandekar
- Department of Medical Microbiology and Immunology, University of California, Davis, Davis, California, USA
| | - Nichole R. Klatt
- Department of Medicine, University of Minnesota, Minneapolis, Minnesota, USA
| | | | - Roger Paredes
- Institut de Recerca de la SIDA IrsiCaixa i Unitat VIH, Universitat Autònoma de Barcelona, Universitat de Vic, Vic, Spain
| | | | - Jim A. Turpin
- Divison of AIDS, NIAID, NIH, Bethesda, Maryland, USA
| | - Alan L. Landay
- Division of Gerontology, Department of Internal Medicine, Rush University Medical Center, Chicago, Illinois, USA
| | - Mimi Ghosh
- Department of Epidemiology, The George Washington University, Washington, District of Columbia, USA
| |
Collapse
|
173
|
Coman V, Vodnar DC. Gut microbiota and old age: Modulating factors and interventions for healthy longevity. Exp Gerontol 2020; 141:111095. [PMID: 32979504 PMCID: PMC7510636 DOI: 10.1016/j.exger.2020.111095] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 09/11/2020] [Accepted: 09/15/2020] [Indexed: 02/06/2023]
Abstract
Our gut microbiota is a complex and dynamic ecosystem with a paramount role in shaping our metabolic and immunological functions. Recent research suggests that aging may negatively affect the composition, diversity, and function of our microbiota mainly due to alterations in diet and immunologic reactivity (i.e. immunosenescence), and increased incidence of certain diseases and, therefore, increased exposure to certain medication (e.g. antibiotics, proton pump inhibitors). In turn, this aging-related gut dysbiosis may contribute to the initiation and/or progress of other metabolic diseases, and consequently, to a decrease in healthy longevity. On the positive side, promising therapeutic interventions, such as diet supplementation with prebiotics, probiotics and synbiotics, or fecal microbiota transplantation, aimed to counteract these aging-related deleterious consequences, could improve our health, and extend our healthy lifespan. In this context, the current review aims to assess the latest progress in identifying the key elements affecting the gut microbiota of the older adults and their mechanism of action, and the effectiveness of the therapeutic interventions aimed at restoring the diversity and healthy functions of the gut microbiota in older individuals.
Collapse
Affiliation(s)
- Vasile Coman
- Institute of Life Sciences, University of Agricultural Sciences and Veterinary Medicine Cluj-Napoca, Calea Mănăştur 3-5, 400372 Cluj-Napoca, Romania.
| | - Dan Cristian Vodnar
- Institute of Life Sciences, University of Agricultural Sciences and Veterinary Medicine Cluj-Napoca, Calea Mănăştur 3-5, 400372 Cluj-Napoca, Romania; Faculty of Food Science and Technology, University of Agricultural Sciences and Veterinary Medicine Cluj-Napoca, Calea Mănăștur 3-5, 400372 Cluj-Napoca, Romania.
| |
Collapse
|
174
|
Gut Microbiota in Patients with Morbid Obesity Before and After Bariatric Surgery: a Ten-Year Review Study (2009-2019). Obes Surg 2020; 31:317-326. [PMID: 33130944 DOI: 10.1007/s11695-020-05074-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 10/14/2020] [Accepted: 10/21/2020] [Indexed: 02/07/2023]
Abstract
The changes in the composition and function of gut microbiota affect the metabolic functions (which are mediated by microbial effects) in patients with obesity, resulting in significant physiological regulation in these patients. Most of the studies emphasize that the Western-style diet (high fat and low vegetable consumption) leads to significant changes in the intestinal microbiome in individuals with metabolic syndrome. A deeper understanding of the profiles of gut microbes will contribute to the development of new therapeutic strategies for the management of metabolic syndrome and other metabolic diseases and related disorders. The aim of this review is to evaluate recent experimental evidence outlining the alterations of gut microbiota composition and function in recovery from bariatric surgical operations with an emphasis on sleeve gastrectomy and gastric bypass.
Collapse
|
175
|
Schierová D, Březina J, Mrázek J, Fliegerová KO, Kvasnová S, Bajer L, Drastich P. Gut Microbiome Changes in Patients with Active Left-Sided Ulcerative Colitis after Fecal Microbiome Transplantation and Topical 5-aminosalicylic Acid Therapy. Cells 2020; 9:cells9102283. [PMID: 33066233 PMCID: PMC7602113 DOI: 10.3390/cells9102283] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 10/01/2020] [Accepted: 10/09/2020] [Indexed: 12/12/2022] Open
Abstract
Ulcerative colitis (UC) is an inflammatory bowel disease, and intestinal bacteria are implicated in the pathogenesis of this disorder. The administration of aminosalicylates (5-ASA) is a conventional treatment that targets the mucosa, while fecal microbial transplantation (FMT) is a novel treatment that directly targets the gut microbiota. The aim of this study was to identify changes in fecal bacterial composition after both types of treatments and evaluate clinical responses. Sixteen patients with active left-sided UC underwent enema treatment using 5-ASA (n = 8) or FMT (n = 8) with a stool from a single donor. Fecal microbiota were analyzed by 16S rDNA high-throughput sequencing, and clinical indices were used to assess the efficacy of treatments. 5-ASA therapy resulted in clinical remission in 50% (4/8) of patients, but no correlation with changes in fecal bacteria was observed. In FMT, remission was achieved in 37.5% (3/8) of patients and was associated with a significantly increased relative abundance of the families Lachnospiraceae, Ruminococcaceae, and Clostridiaceae of the phylum Firmicutes, and Bifidobacteriaceae and Coriobacteriaceae of the phylum Actinobacteria. At the genus level, Faecalibacterium, Blautia, Coriobacteria, Collinsela, Slackia, and Bifidobacterium were significantly more frequent in patients who reached clinical remission. However, the increased abundance of beneficial taxa was not a sufficient factor to achieve clinical improvement in all UC patients. Nevertheless, our preliminary results indicate that FMT as non-drug-using method is thought to be a promising treatment for UC patients.
Collapse
Affiliation(s)
- Dagmar Schierová
- Institute of Animal Physiology and Genetics of the Czech Academy of Science, v.v.i., 142 20 Prague, Czech Republic; (K.O.F.); (S.K.)
- Correspondence: (D.S.); (J.M.); Tel.: +420-2-6709-0509 (D.S.); +420-2-6709-0506 (J.M.)
| | - Jan Březina
- Hepatogastroenterology Department, Institute for Clinical and Experimental Medicine, 140 21 Prague, Czech Republic; (J.B.); (L.B.); (P.D.)
| | - Jakub Mrázek
- Institute of Animal Physiology and Genetics of the Czech Academy of Science, v.v.i., 142 20 Prague, Czech Republic; (K.O.F.); (S.K.)
- Correspondence: (D.S.); (J.M.); Tel.: +420-2-6709-0509 (D.S.); +420-2-6709-0506 (J.M.)
| | - Kateřina Olša Fliegerová
- Institute of Animal Physiology and Genetics of the Czech Academy of Science, v.v.i., 142 20 Prague, Czech Republic; (K.O.F.); (S.K.)
| | - Simona Kvasnová
- Institute of Animal Physiology and Genetics of the Czech Academy of Science, v.v.i., 142 20 Prague, Czech Republic; (K.O.F.); (S.K.)
| | - Lukáš Bajer
- Hepatogastroenterology Department, Institute for Clinical and Experimental Medicine, 140 21 Prague, Czech Republic; (J.B.); (L.B.); (P.D.)
| | - Pavel Drastich
- Hepatogastroenterology Department, Institute for Clinical and Experimental Medicine, 140 21 Prague, Czech Republic; (J.B.); (L.B.); (P.D.)
| |
Collapse
|
176
|
Khan S, Hauptman R, Kelly L. Engineering the Microbiome to Prevent Adverse Events: Challenges and Opportunities. Annu Rev Pharmacol Toxicol 2020; 61:159-179. [PMID: 33049161 DOI: 10.1146/annurev-pharmtox-031620-031509] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
In the past decade of microbiome research, we have learned about numerous adverse interactions between the microbiome and medical interventions such as drugs, radiation, and surgery. What if we could alter our microbiomes to prevent these events? In this review, we discuss potential routes to mitigate microbiome adverse events, including applications from the emerging field of microbiome engineering. We highlight cases where the microbiome acts directly on a treatment, such as via differential drug metabolism, and cases where a treatment directly harms the microbiome, such as in radiation therapy. Understanding and preventing microbiome adverse events is a difficult challenge that will require a data-driven approach involving causal statistics, multiomics techniques, and a personalized means of mitigating adverse events. We propose research considerations to encourage productive work in preventing microbiome adverse events, and we highlight the many challenges and opportunities that await.
Collapse
Affiliation(s)
- Saad Khan
- Department of Systems and Computational Biology, Albert Einstein College of Medicine, New York, NY 10461, USA;
| | - Ruth Hauptman
- Department of Systems and Computational Biology, Albert Einstein College of Medicine, New York, NY 10461, USA;
| | - Libusha Kelly
- Department of Systems and Computational Biology, Albert Einstein College of Medicine, New York, NY 10461, USA; .,Department of Microbiology and Immunology, Albert Einstein College of Medicine, New York, NY 10461, USA
| |
Collapse
|
177
|
Wu J, Zhao Y, Wang X, Kong L, Johnston LJ, Lu L, Ma X. Dietary nutrients shape gut microbes and intestinal mucosa via epigenetic modifications. Crit Rev Food Sci Nutr 2020; 62:783-797. [PMID: 33043708 DOI: 10.1080/10408398.2020.1828813] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Jianmin Wu
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Ying Zhao
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Xian Wang
- College of Animal Science and Technology, Beijing University of Agriculture, Beijing, China
| | - Lingchang Kong
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Lee J. Johnston
- West Central Research & Outreach Centre, University of Minnesota, Morris, Minnesota, USA
| | - Lin Lu
- College of Animal Science and Technology, Beijing University of Agriculture, Beijing, China
| | - Xi Ma
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| |
Collapse
|
178
|
Terveer EM, Vendrik KE, Ooijevaar RE, Lingen EV, Boeije-Koppenol E, Nood EV, Goorhuis A, Bauer MP, van Beurden YH, Dijkgraaf MG, Mulder CJ, Vandenbroucke-Grauls CM, Seegers JF, van Prehn J, Verspaget HW, Kuijper EJ, Keller JJ. Faecal microbiota transplantation for Clostridioides difficile infection: Four years' experience of the Netherlands Donor Feces Bank. United European Gastroenterol J 2020; 8:1236-1247. [PMID: 32990503 PMCID: PMC7724536 DOI: 10.1177/2050640620957765] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND The Netherlands Donor Feces Bank provides standardized ready-to-use donor faecal suspensions for faecal microbiota transplantation treatment of patients with recurrent Clostridioides difficile infection. OBJECTIVE The purpose of this study was evaluation of safety, feasibility and outcome of faecal microbiota transplantation facilitated by a national stool bank. METHODS The methods used included: observational cohort study of donors and recipients of faecal suspensions; assessment of donor screening and patient selection performed by an expert panel of medical microbiologists, gastroenterologists and infectious disease specialists; and patient outcome evaluated at different timepoints after faecal microbiota transplantation. RESULTS Of 871 volunteers who registered as a potential faeces donor, 16 (2%) became active donors. Nine donors stopped or were excluded after a mean donation period of 5.7 months. In 2016-2019, 47 (27%) of 176 requests for faecal microbiota transplantations were deemed not indicated by the expert panel. In total, 129 patients with recurrent C. difficile infection were treated with 143 faecal suspensions in 40 different hospitals. The cure rate at two months after a single infusion was 89% (107/120). Of 84 patients, long-term follow-up (median 42 weeks) was available and sustained cure was achieved in 61 (73%). Early C. difficile infection relapses (within two months after faecal microbiota transplantation) and late recurrences (after more than two months) occurred more frequently in patients who received non-C. difficile antibiotics within three weeks after faecal microbiota transplantation and in moderately to severely immunocompromised patients. Of 21 patients with C. difficile infection after faecal microbiota transplantation, 14 were cured with anti-C. difficile antibiotics and seven with a second transplantation. No faecal microbiota transplantation-related serious adverse events were observed, but gastro-intestinal complaints (nausea, abdominal pain or diarrhoea) persisted in 32% of the treated patients at long-term follow-up. CONCLUSION Faecal suspensions provided by a centralized stool bank, supported by a multidisciplinary expert team, resulted in effective, appropriate and safe application of faecal microbiota transplantation for recurrent C. difficile infection. LEVEL OF EVIDENCE Level II, prospective cohort study.
Collapse
Affiliation(s)
- Elisabeth M Terveer
- Department of Medical Microbiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Karuna Ew Vendrik
- Department of Medical Microbiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Rogier E Ooijevaar
- Department of Gastroenterology and Hepatology, Amsterdam University Medical Centers, Amsterdam, the Netherlands
| | - Emilie van Lingen
- Department of Gastroenterology, Leiden University Medical Center, Leiden, the Netherlands
| | - Eline Boeije-Koppenol
- Department of Medical Microbiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Els van Nood
- Department of Internal Medicine, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Abraham Goorhuis
- Department of Internal Medicine, Amsterdam University Medical Centers, Amsterdam, the Netherlands
| | - Martijn P Bauer
- Department of Internal Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Yvette H van Beurden
- Department of Gastroenterology and Hepatology, Amsterdam University Medical Centers, Amsterdam, the Netherlands.,Department of Gastroenterology and Hepatology, Spaarne Gasthuis, Hoofddorp, the Netherlands
| | - Marcel Gw Dijkgraaf
- Department of Epidemiology and Data Science, Amsterdam University Medical Centers, Amsterdam, the Netherlands
| | - Chris Jj Mulder
- Department of Gastroenterology and Hepatology, Amsterdam University Medical Centers, Amsterdam, the Netherlands
| | | | | | - Joffrey van Prehn
- Department of Medical Microbiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Hein W Verspaget
- Department of Gastroenterology, Leiden University Medical Center, Leiden, the Netherlands.,Department of Biobanking, Leiden University Medical Center, Leiden, the Netherlands
| | - Ed J Kuijper
- Department of Medical Microbiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Josbert J Keller
- Department of Gastroenterology, Leiden University Medical Center, Leiden, the Netherlands.,Department of Gastroenterology, Haaglanden Medisch Centrum, The Hague, the Netherlands
| |
Collapse
|
179
|
Cold F, Kousgaard SJ, Halkjaer SI, Petersen AM, Nielsen HL, Thorlacius-Ussing O, Hansen LH. Fecal Microbiota Transplantation in the Treatment of Chronic Pouchitis: A Systematic Review. Microorganisms 2020; 8:E1433. [PMID: 32962069 PMCID: PMC7565999 DOI: 10.3390/microorganisms8091433] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 09/09/2020] [Accepted: 09/16/2020] [Indexed: 12/13/2022] Open
Abstract
The objective was to evaluate available literature on treatment of chronic pouchitis with fecal microbiota transplantation (FMT) focusing on clinical outcomes, safety, and different approaches to FMT preparation and delivery. A systematic review of electronic databases was conducted using Medline, EMBASE, and the Cochrane Central Register of Controlled Trials Library from inception through April 2020. Human studies of all study types reporting results of FMT to treat chronic pouchitis were included. Nine studies, reporting FMT treatment of 69 patients with chronic pouchitis were found eligible for the review. Most studies were case series and cohort studies rated as having fair to poor quality due to high risk of bias and small sample size. Only one randomized controlled trial was included, finding no beneficial effect of FMT. In total clinical response after FMT was reported in 14 (31.8%) out of 44 evaluated patients at various timepoints after FMT, and clinical remission in ten (22.7%) patients. Only minor self-limiting adverse events were reported. FMT varied greatly regarding preparation, length of treatment, and route of delivery. The effects of FMT on symptoms of chronic pouchitis are not established, though some studies show promising results. Future controlled well-designed studies are warranted.
Collapse
Affiliation(s)
- Frederik Cold
- Department of Plant and Environmental Sciences, Section for Microbial Ecology and Biotechnology, Copenhagen University, Thorvaldsensvej 40, 1871 Frederiksberg C, Denmark;
- Gastrounit, Medical Division, Copenhagen University Hospital Hvidovre, 2650 Hvidovre, Denmark; (S.I.H.); (A.M.P.)
| | - Sabrina Just Kousgaard
- Department of Gastrointestinal Surgery, Aalborg University Hospital, 9100 Aalborg, Denmark; (S.J.K.); (O.T.-U.)
- Department of Clinical Medicine, Aalborg University, 9100 Aalborg, Denmark;
| | - Sofie Ingdam Halkjaer
- Gastrounit, Medical Division, Copenhagen University Hospital Hvidovre, 2650 Hvidovre, Denmark; (S.I.H.); (A.M.P.)
| | - Andreas Munk Petersen
- Gastrounit, Medical Division, Copenhagen University Hospital Hvidovre, 2650 Hvidovre, Denmark; (S.I.H.); (A.M.P.)
- Department of Clinical Microbiology, Copenhagen University Hospital Hvidovre, 2650 Hvidovre, Denmark
| | - Hans Linde Nielsen
- Department of Clinical Medicine, Aalborg University, 9100 Aalborg, Denmark;
- Department of Clinical Microbiology, Aalborg University Hospital, 9100 Aalborg, Denmark
| | - Ole Thorlacius-Ussing
- Department of Gastrointestinal Surgery, Aalborg University Hospital, 9100 Aalborg, Denmark; (S.J.K.); (O.T.-U.)
- Department of Clinical Medicine, Aalborg University, 9100 Aalborg, Denmark;
| | - Lars Hestbjerg Hansen
- Department of Plant and Environmental Sciences, Section for Microbial Ecology and Biotechnology, Copenhagen University, Thorvaldsensvej 40, 1871 Frederiksberg C, Denmark;
| |
Collapse
|
180
|
Segal JP, Mullish BH, Quraishi MN, Iqbal T, Marchesi JR, Sokol H. Mechanisms underpinning the efficacy of faecal microbiota transplantation in treating gastrointestinal disease. Therap Adv Gastroenterol 2020; 13:1756284820946904. [PMID: 32952613 PMCID: PMC7475788 DOI: 10.1177/1756284820946904] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2020] [Accepted: 07/13/2020] [Indexed: 02/04/2023] Open
Abstract
Faecal microbiota transplantation (FMT) is currently a recommended therapy for recurrent/refractory Clostridioides difficile infection (CDI). The success of FMT for CDI has led to interest in its therapeutic potential in many other disorders. The mechanisms that underpin the efficacy of FMT are not fully understood. Importantly, FMT remains a crucial treatment in managing CDI and understanding the mechanisms that underpin its success will be critical to improve its clinical efficacy, safety and usability. Furthermore, a deeper understanding of this may allow us to expose FMT's full potential as a therapeutic tool for other disease states. This review will explore the current understanding of the mechanisms underlying the efficacy of FMT across a variety of diseases.
Collapse
Affiliation(s)
- Jonathan P. Segal
- Departments of Gastroenterology and Hepatology, St Mary’s Hospital, Imperial College Healthcare NHS Trust, South Wharf Rd, London W2 1NY, UK
- Division of Digestive Diseases, Department of Metabolism, Digestion and Reproduction, Imperial College London, UK
| | - Benjamin H. Mullish
- Departments of Gastroenterology and Hepatology, St Mary’s Hospital, Imperial College Healthcare NHS Trust, London, UK
- Department of Metabolism, Digestion and Reproduction, Division of Digestive Diseases, Imperial College London, UK
| | - Mohammed N. Quraishi
- University of Birmingham Microbiome Treatment Centre, University of Birmingham, Birmingham, UK
- Department of Gastroenterology, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - Tariq Iqbal
- University of Birmingham Microbiome Treatment Centre, University of Birmingham, Birmingham, UK
- Department of Gastroenterology, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - Julian R. Marchesi
- Department of Metabolism, Digestion and Reproduction, Division of Digestive Diseases, Imperial College London, UK
- School of Biosciences, Cardiff University, Cardiff, UK
| | - Harry Sokol
- Gastroenterology Department, INSERM, Centre de Recherche Saint Antoine, CRSA, AP-HP, Sorbonne Université, Saint Antoine Hospital, Paris, France
- INRA, UMR1319 Micalis and AgroParisTech, Jouy en Josas, France Paris Centre for Microbiome Medicine (PaCeMM) FHU, Paris, France
| |
Collapse
|
181
|
Raghu Subramanian C, Talluri S, Khan SU, Katz JA, Georgetson M, Sinh P. Fecal Microbiota Transplantation for Recurrent Clostridium difficile Infection in Patients With Multiple Comorbidities: Long-Term Safety and Efficacy Results From a Tertiary Care Community Hospital. Gastroenterology Res 2020; 13:138-145. [PMID: 32864024 PMCID: PMC7433372 DOI: 10.14740/gr1275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Accepted: 04/29/2020] [Indexed: 11/19/2022] Open
Abstract
Background Cure rates of Clostridium difficile infection (CDI) with fecal microbiota transplant (FMT) have been promising. However, there is debate regarding success of FMT in patients with comorbidities. Methods Electronic chart review was done to collect data on patients who underwent FMT from January 2015 to August 2017. Charts were analyzed in November 2018 with a median follow-up of 25.4 months (interquartile range 20 - 31 months). Results Twenty patients underwent FMT. The primary success rate at our institution was 90% and overall success rate was 100%. Six patients (43%) had FMT failure (two early and four late). Conclusions This case series is a description of our center’s initial experience with FMT for treatment of recurrent CDI. Our high success rate reiterates the efficacy and safety of FMT in this population including patients with comorbidities.
Collapse
Affiliation(s)
- Charumathi Raghu Subramanian
- Department of Medicine, Guthrie Clinic/Robert Packer Hospital, Sayre, PA, USA.,Washington Hospital Healthcare System, Fremont, CA, USA
| | - Swapna Talluri
- Department of Medicine, Guthrie Clinic/Robert Packer Hospital, Sayre, PA, USA
| | - Safi Ullah Khan
- Department of Medicine, Guthrie Clinic/Robert Packer Hospital, Sayre, PA, USA.,Department of Medicine, West Virginia University, Morgantown, WV, USA
| | - Jeffry A Katz
- Division of Gastroenterology and Liver Diseases, Case Western Reserve University, University Hospitals Cleveland Medical Center, Cleveland, OH, USA
| | - Michael Georgetson
- Division of Gastroenterology, Department of Medicine, Guthrie Clinic, Sayre, PA, USA
| | - Preetika Sinh
- Division of Gastroenterology, Department of Medicine, Guthrie Clinic, Sayre, PA, USA.,Division of Gastroenterology and Hepatology, Medical College of Wisconsin, Milwaukee, WI, USA
| |
Collapse
|
182
|
Ronai I, Greslehner GP, Boem F, Carlisle J, Stencel A, Suárez J, Bayir S, Bretting W, Formosinho J, Guerrero AC, Morgan WH, Prigot-Maurice C, Rodeck S, Vasse M, Wallis JM, Zacks O. "Microbiota, symbiosis and individuality summer school" meeting report. MICROBIOME 2020; 8:117. [PMID: 32795355 PMCID: PMC7427737 DOI: 10.1186/s40168-020-00898-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Accepted: 07/24/2020] [Indexed: 05/13/2023]
Abstract
How does microbiota research impact our understanding of biological individuality? We summarize the interdisciplinary summer school on "Microbiota, symbiosis and individuality: conceptual and philosophical issues" (July 2019), which was supported by a European Research Council starting grant project "Immunity, DEvelopment, and the Microbiota" (IDEM). The summer school centered around interdisciplinary group work on four facets of microbiota research: holobionts, individuality, causation, and human health. The conceptual discussion of cutting-edge empirical research provided new insights into microbiota and highlights the value of incorporating into meetings experts from other disciplines, such as philosophy and history of science. Video Abstract.
Collapse
Affiliation(s)
- Isobel Ronai
- Columbia University, 1200 Amsterdam Ave, New York, 10027 NY USA
| | - Gregor P. Greslehner
- ImmunoConcept, UMR5164, CNRS & University of Bordeaux, 146 Rue Léo Saignat, Bordeaux, 33076 France
| | - Federico Boem
- Dipartimento di Filosofia e Scienze dell’Educazione, Università degli Studi di Torino, Palazzo Nuovo, Via Sant’Ottavio, 20, Torino, 10124 Italy
| | - Judith Carlisle
- Washington University in St. Louis, Department of Philosophy, One Brookings Drive, St. Louis, 63130-4899 MO USA
| | - Adrian Stencel
- Institute of Philosophy, Jagiellonian University, Grodzka 52, Kraków, 33-332 Poland
| | - Javier Suárez
- Abteilung Philosophie, Universität Bielefeld, Universitätsstraße 25, Bielefeld, 33615 Germany
| | - Saliha Bayir
- Institut für Philosophie,Universität Kassel, Henschelstr. 2, Kassel, 34127 Germany
| | - Wiebke Bretting
- ImmunoConcept, UMR5164, CNRS & University of Bordeaux, 146 Rue Léo Saignat, Bordeaux, 33076 France
| | - Joana Formosinho
- Medical Museion, Department of Public Health, University of Copenhagen, Fredericiagade 18, Copenhagen, 1310 Denmark
| | - Anna C. Guerrero
- Arizona State University, Center for Biology and Society, 427 E Tyler Mall, Tempe, 85281 AZ USA
| | - William H. Morgan
- The University of Sheffield, Department of Philosophy, 45 Victoria Street, Sheffield, S3 7QB UK
| | - Cybèle Prigot-Maurice
- Université de Poitiers, Laboratoire Écologie et Biologie des Interactions, UMR CNRS 7267, Bâtiment B35, 5 rue Albert Turpain, TSA 51106, Poitiers Cedex 9, 86073 France
| | - Salome Rodeck
- Leibniz Center for Literary and Cultural Research, Schützenstr. 18, Berlin, 10117 Germany
| | - Marie Vasse
- Institute for Integrative Biology, ETH Zürich, Universitätstrasse 16, Zürich, 8092 Switzerland
| | - Jacqueline M. Wallis
- University of Bristol, Department of Philosophy, Cotham House, Bristol, BS6 6JL UK
| | - Oryan Zacks
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, 6997801 Israel
| |
Collapse
|
183
|
Yan Y, Nguyen LH, Franzosa EA, Huttenhower C. Strain-level epidemiology of microbial communities and the human microbiome. Genome Med 2020; 12:71. [PMID: 32791981 PMCID: PMC7427293 DOI: 10.1186/s13073-020-00765-y] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Accepted: 07/14/2020] [Indexed: 02/07/2023] Open
Abstract
The biological importance and varied metabolic capabilities of specific microbial strains have long been established in the scientific community. Strains have, in the past, been largely defined and characterized based on microbial isolates. However, the emergence of new technologies and techniques has enabled assessments of their ecology and phenotypes within microbial communities and the human microbiome. While it is now more obvious how pathogenic strain variants are detrimental to human health, the consequences of subtle genetic variation in the microbiome have only recently been exposed. Here, we review the operational definitions of strains (e.g., genetic and structural variants) as they can now be identified from microbial communities using different high-throughput, often culture-independent techniques. We summarize the distribution and diversity of strains across the human body and their emerging links to health maintenance, disease risk and progression, and biochemical responses to perturbations, such as diet or drugs. We list methods for identifying, quantifying, and tracking strains, utilizing high-throughput sequencing along with other molecular and “culturomics” technologies. Finally, we discuss implications of population studies in bridging experimental gaps and leading to a better understanding of the health effects of strains in the human microbiome.
Collapse
Affiliation(s)
- Yan Yan
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, 677 Huntington Ave, Boston, MA, 02115, USA.,Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Long H Nguyen
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, 677 Huntington Ave, Boston, MA, 02115, USA.,Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.,Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Eric A Franzosa
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, 677 Huntington Ave, Boston, MA, 02115, USA.,Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Curtis Huttenhower
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, 677 Huntington Ave, Boston, MA, 02115, USA. .,Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| |
Collapse
|
184
|
Alagna L, Palomba E, Mangioni D, Bozzi G, Lombardi A, Ungaro R, Castelli V, Prati D, Vecchi M, Muscatello A, Bandera A, Gori A. Multidrug-Resistant Gram-Negative Bacteria Decolonization in Immunocompromised Patients: A Focus on Fecal Microbiota Transplantation. Int J Mol Sci 2020; 21:ijms21165619. [PMID: 32764526 PMCID: PMC7460658 DOI: 10.3390/ijms21165619] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 07/28/2020] [Accepted: 08/03/2020] [Indexed: 02/07/2023] Open
Abstract
Antimicrobial resistance is an important issue for global health; in immunocompromised patients, such as solid organ and hematological transplant recipients, it poses an even bigger threat. Colonization by multidrug-resistant (MDR) bacteria was acknowledged as a strong risk factor to subsequent infections, especially in individuals with a compromised immune system. A growing pile of studies has linked the imbalance caused by the dominance of certain taxa populating the gut, also known as intestinal microbiota dysbiosis, to an increased risk of MDR bacteria colonization. Several attempts were proposed to modulate the gut microbiota. Particularly, fecal microbiota transplantation (FMT) was successfully applied to treat conditions like Clostridioides difficile infection and other diseases linked to gut microbiota dysbiosis. In this review we aimed to provide a look at the data gathered so far on FMT, focusing on its possible role in treating MDR colonization in the setting of immunocompromised patients and analyzing its efficacy and safety.
Collapse
Affiliation(s)
- Laura Alagna
- Infectious Disease Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (L.A.); (D.M.); (G.B.); (A.L.); (R.U.); (V.C.); (A.M.); (A.B.); (A.G.)
| | - Emanuele Palomba
- Infectious Disease Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (L.A.); (D.M.); (G.B.); (A.L.); (R.U.); (V.C.); (A.M.); (A.B.); (A.G.)
- Department of Pathophysiology and Transplantation, University of Milan, 20122 Milan, Italy;
- Centre for Multidisciplinary Research in Health Science, University of Milan, 20122 Milan, Italy
- Correspondence: ; Tel.: +39-3494073517
| | - Davide Mangioni
- Infectious Disease Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (L.A.); (D.M.); (G.B.); (A.L.); (R.U.); (V.C.); (A.M.); (A.B.); (A.G.)
- Department of Medical Biotechnology and Translational Medicine, University of Milan, 20122 Milan, Italy
| | - Giorgio Bozzi
- Infectious Disease Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (L.A.); (D.M.); (G.B.); (A.L.); (R.U.); (V.C.); (A.M.); (A.B.); (A.G.)
| | - Andrea Lombardi
- Infectious Disease Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (L.A.); (D.M.); (G.B.); (A.L.); (R.U.); (V.C.); (A.M.); (A.B.); (A.G.)
| | - Riccardo Ungaro
- Infectious Disease Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (L.A.); (D.M.); (G.B.); (A.L.); (R.U.); (V.C.); (A.M.); (A.B.); (A.G.)
| | - Valeria Castelli
- Infectious Disease Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (L.A.); (D.M.); (G.B.); (A.L.); (R.U.); (V.C.); (A.M.); (A.B.); (A.G.)
- Department of Pathophysiology and Transplantation, University of Milan, 20122 Milan, Italy;
- Centre for Multidisciplinary Research in Health Science, University of Milan, 20122 Milan, Italy
| | - Daniele Prati
- Department of Transfusion Medicine and Hematology, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy;
| | - Maurizio Vecchi
- Department of Pathophysiology and Transplantation, University of Milan, 20122 Milan, Italy;
- Gastroenterology and Endoscopy Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Antonio Muscatello
- Infectious Disease Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (L.A.); (D.M.); (G.B.); (A.L.); (R.U.); (V.C.); (A.M.); (A.B.); (A.G.)
| | - Alessandra Bandera
- Infectious Disease Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (L.A.); (D.M.); (G.B.); (A.L.); (R.U.); (V.C.); (A.M.); (A.B.); (A.G.)
- Department of Pathophysiology and Transplantation, University of Milan, 20122 Milan, Italy;
- Centre for Multidisciplinary Research in Health Science, University of Milan, 20122 Milan, Italy
| | - Andrea Gori
- Infectious Disease Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (L.A.); (D.M.); (G.B.); (A.L.); (R.U.); (V.C.); (A.M.); (A.B.); (A.G.)
- Department of Pathophysiology and Transplantation, University of Milan, 20122 Milan, Italy;
- Centre for Multidisciplinary Research in Health Science, University of Milan, 20122 Milan, Italy
| |
Collapse
|
185
|
Van Laar T, Boertien JM, Herranz AH. Faecal Transplantation, Pro- and Prebiotics in Parkinson's Disease; Hope or Hype? JOURNAL OF PARKINSONS DISEASE 2020; 9:S371-S379. [PMID: 31609702 PMCID: PMC6839600 DOI: 10.3233/jpd-191802] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Abstract Faecal microbiome transplantation (FMT) is an attractive technique, because the administration is relatively simple and in general has a mild adverse effect pattern. Moreover, FMT consists of a broad mixture, which could be beneficial, because at this moment it is not known what type of changes in the microbiome are needed. However, except from a few cases no clinical data in Parkinson’s disease (PD) is available yet. There is some indication that FMT might be beneficial in severe constipated PD patients, but the clinical data to support this are very scarce. So, actually there are no good data in the public domain to support FMT at this moment in PD patients. FMT at this moment is a black box with too many unanswered questions, also with respect to safety concerns. Only the administration of species of Lactobacillus and Bifidobacterium over a time period of four to twelve weeks has repeatedly proven to be effective in treating constipation in PD. Also, no solid clinical data are available about the possible effects of probiotic treatment on motor symptoms or progression of PD. Therefore, also probiotic treatments in PD should wait until better clinical data become available, in order to select the right target populations and to have good estimates of the clinical effects to be expected.
Collapse
Affiliation(s)
- T Van Laar
- Department of Neurology, University Medical Center Groningen, University of Groningen, The Netherlands
| | - J M Boertien
- Department of Neurology, University Medical Center Groningen, University of Groningen, The Netherlands
| | - A Horta Herranz
- Behavioral and Cognitive Neurosciences, University of Groningen, The Netherlands
| |
Collapse
|
186
|
Jung S, Seo DJ, Yeo D, Wang Z, Min A, Zhao Z, Song M, Choi IS, Myoung J, Choi C. Experimental infection of hepatitis E virus induces pancreatic necroptosis in miniature pigs. Sci Rep 2020; 10:12022. [PMID: 32694702 PMCID: PMC7374588 DOI: 10.1038/s41598-020-68959-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Accepted: 06/16/2020] [Indexed: 02/07/2023] Open
Abstract
Infection by hepatitis E virus (HEV) via the oral route causes acute hepatitis. Extra-hepatic manifestations of HEV infection may stem from various causes; however, its distribution in organs such as the liver, as well as the mechanisms underlying HEV-induced cell injury, remain unclear. The objective of this study was to determine the chronological distribution of HEV in various tissues of HEV-challenged miniature pigs and to investigate the mechanisms underlying HEV-induced cell death in the pancreas and liver. Virological and serological analyses were performed on blood and faecal samples. Histopathology of the liver and extra-hepatic tissues was analysed. Cell death pathways and immune cell characterisation in inflammatory lesions were analysed using immunohistochemistry. The liver and pancreas displayed inflammation and cellular injury, and a large amount of HEV was observed in the lesions. The liver was infiltrated by T and natural killer cells. HEV was identified in all organs except the heart, and was associated with immune cells. Although the liver and the pancreas strongly expressed TNF-α and TRAIL, TUNEL assay results were negative. RIP3 and pMLKL were expressed in the pancreas. RIP3, but not pMLKL, was expressed in the liver. Pancreatitis induced in HEV-infected miniature pigs is associated with necroptosis.
Collapse
Affiliation(s)
- Soontag Jung
- Department of Food and Nutrition, School of Food Science and Technology, College of Biotechnology and Natural Resources, Chung-Ang University, 4726 Seodongdaero, Daedeok-myeon, Anseong-si, Gyeonggi-do, 17546, Republic of Korea
| | - Dong Joo Seo
- Department of Food and Nutrition, Gwangju University, Gwangju, 61743, Republic of Korea
| | - Daseul Yeo
- Department of Food and Nutrition, School of Food Science and Technology, College of Biotechnology and Natural Resources, Chung-Ang University, 4726 Seodongdaero, Daedeok-myeon, Anseong-si, Gyeonggi-do, 17546, Republic of Korea
| | - Zhaoqi Wang
- Department of Food and Nutrition, School of Food Science and Technology, College of Biotechnology and Natural Resources, Chung-Ang University, 4726 Seodongdaero, Daedeok-myeon, Anseong-si, Gyeonggi-do, 17546, Republic of Korea
| | - Ae Min
- Department of Food and Nutrition, School of Food Science and Technology, College of Biotechnology and Natural Resources, Chung-Ang University, 4726 Seodongdaero, Daedeok-myeon, Anseong-si, Gyeonggi-do, 17546, Republic of Korea
| | - Ziwei Zhao
- Department of Food and Nutrition, School of Food Science and Technology, College of Biotechnology and Natural Resources, Chung-Ang University, 4726 Seodongdaero, Daedeok-myeon, Anseong-si, Gyeonggi-do, 17546, Republic of Korea
| | - Mengxiao Song
- Department of Food and Nutrition, School of Food Science and Technology, College of Biotechnology and Natural Resources, Chung-Ang University, 4726 Seodongdaero, Daedeok-myeon, Anseong-si, Gyeonggi-do, 17546, Republic of Korea
| | - In-Soo Choi
- Department of Infectious Disease, College of Veterinary Medicine, Konkuk University, Seoul, 05029, Republic of Korea
| | - Jinjong Myoung
- Korea Zoonosis Research Institute, Chonbuk National University, Jeonju, 54896, Republic of Korea
| | - Changsun Choi
- Department of Food and Nutrition, School of Food Science and Technology, College of Biotechnology and Natural Resources, Chung-Ang University, 4726 Seodongdaero, Daedeok-myeon, Anseong-si, Gyeonggi-do, 17546, Republic of Korea.
- Bio and Environmental Technology Research Institute, Chung-Ang University, 4726 Seodongdaero, Daedeok-myeon, Anseong-si, Gyeonggi-do, 17546, Republic of Korea.
| |
Collapse
|
187
|
An ecological framework to understand the efficacy of fecal microbiota transplantation. Nat Commun 2020; 11:3329. [PMID: 32620839 PMCID: PMC7334230 DOI: 10.1038/s41467-020-17180-x] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Accepted: 06/11/2020] [Indexed: 02/08/2023] Open
Abstract
Human gut microbiota plays critical roles in physiology and disease. Our understanding of ecological principles that govern the dynamics and resilience of this highly complex ecosystem remains rudimentary. This knowledge gap becomes more problematic as new approaches to modifying this ecosystem, such as fecal microbiota transplantation (FMT), are being developed as therapeutic interventions. Here we present an ecological framework to understand the efficacy of FMT in treating conditions associated with a disrupted gut microbiota, using the recurrent Clostridioides difficile infection as a prototype disease. This framework predicts several key factors that determine the efficacy of FMT. Moreover, it offers an efficient algorithm for the rational design of personalized probiotic cocktails to decolonize pathogens. We analyze data from both preclinical mouse experiments and a clinical trial of FMT to validate our theoretical framework. The presented results significantly improve our understanding of the ecological principles of FMT and have a positive translational impact on the rational design of general microbiota-based therapeutics. Here, the authors present a theoretical framework based on community ecology and network science to investigate the efficacy of fecal microbiota transplantation in conditions associated with a disrupted gut microbiota, using the recurrent Clostridioides difficile infection as a prototype disease.
Collapse
|
188
|
Leong KSW, O'Sullivan JM, Derraik JGB, Cutfield WS. Gut microbiome transfer-Finding the perfect fit. Clin Endocrinol (Oxf) 2020; 93:3-10. [PMID: 32181906 DOI: 10.1111/cen.14183] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 03/02/2020] [Accepted: 03/12/2020] [Indexed: 12/12/2022]
Abstract
Gut microbiome transfer (GMT; also referred to as faecal microbiota transplantation or FMT) has been propelled from fringe therapy to mainstream science as a highly effective treatment for recurrent Clostridioides difficile infection. As a result, there has been great interest in the potential efficacy and safety of GMT in treating other medical conditions, for example inflammatory bowel disease, and more recently as a novel therapy for obesity and metabolic diseases. For these chronic conditions, the results from clinical trials have been mixed. Further, specifically in obesity and metabolic diseases, there are limited available data, with only a few published studies with a small number of participants and short duration of follow-up. Therefore, this review aims to explore the human, microbial and formulation factors that may affect the success of GMT. This includes various aspects in the preparation and administration of GMT, such as stool processing, modes of delivery, pretreatment with antibiotics and/or bowel lavage, frequency of GMT and possible use of precision bacteriotherapy. In addition, we examine the potential use of GMT in obesity, type 2 diabetes and metabolic diseases based on current available literature, highlighting some recent advances in GMT research in this area, as well as potential adverse effects after GMT therapy.
Collapse
Affiliation(s)
- Karen S W Leong
- Liggins Institute, University of Auckland, Auckland, New Zealand
- A Better Start - National Science Challenge, Auckland, New Zealand
| | - Justin M O'Sullivan
- Liggins Institute, University of Auckland, Auckland, New Zealand
- A Better Start - National Science Challenge, Auckland, New Zealand
| | - José G B Derraik
- Liggins Institute, University of Auckland, Auckland, New Zealand
- A Better Start - National Science Challenge, Auckland, New Zealand
- Department of Women's and Children's Health, Uppsala University, Uppsala, Sweden
- Department of Endocrinology, Children's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Wayne S Cutfield
- Liggins Institute, University of Auckland, Auckland, New Zealand
- A Better Start - National Science Challenge, Auckland, New Zealand
- Department of Endocrinology, Children's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
189
|
Bilinski J, Dziurzynski M, Grzesiowski P, Podsiadly E, Stelmaszczyk-Emmel A, Dzieciatkowski T, Dziewit L, Basak GW. Multimodal Approach to Assessment of Fecal Microbiota Donors based on Three Complementary Methods. J Clin Med 2020; 9:E2036. [PMID: 32610522 PMCID: PMC7409046 DOI: 10.3390/jcm9072036] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 06/25/2020] [Accepted: 06/26/2020] [Indexed: 01/27/2023] Open
Abstract
Methods of stool assessment are mostly focused on next-generation sequencing (NGS) or classical culturing, but only rarely both. We conducted a series of experiments using a multi-method approach to trace the stability of gut microbiota in various donors over time, to find the best method for the proper selection of fecal donors and to find "super-donor" indicators. Ten consecutive stools donated by each of three donors were used for the experiments (30 stools in total). The experiments assessed bacterial viability measured by flow cytometry, stool culturing on different media and in various conditions, and NGS (90 samples in total). There were no statistically significant differences between live and dead cell numbers; however, we found a group of cells classified as not-dead-not-alive, which may be possibly important in selection of "good" donors. Donor C, being a regular stool donor, was characterized by the largest number of cultivable species (64). Cultivable core microbiota (shared by all donors) was composed of only 16 species. ANCOM analysis of NGS data highlighted particular genera to be more abundant in one donor vs. the others. There was a correlation between the not-dead-not-alive group found in flow cytometry and Anaeroplasma found by NGS, and we could distinguish a regular stool donor from the others. In this work, we showed that combining various methods of microbiota assessment gives more information than each method separately.
Collapse
Affiliation(s)
- Jaroslaw Bilinski
- Department of Hematology, Oncology and Internal Medicine, Medical University of Warsaw, 02-091 Warsaw, Poland;
| | - Mikolaj Dziurzynski
- Department of Environmental Microbiology and Biotechnology, Institute of Microbiology, Faculty of Biology, University of Warsaw, 02-096 Warsaw, Poland
| | - Pawel Grzesiowski
- Foundation for the Infection Prevention Institute, 02-991 Warsaw, Poland;
| | - Edyta Podsiadly
- Department of Microbiology, Institute of Medical Sciences, University of Rzeszów, 35-310 Rzeszów, Poland;
| | - Anna Stelmaszczyk-Emmel
- Department of Laboratory Diagnostics and Clinical Immunology of Developmental Age, Medical University of Warsaw, 02-091 Warsaw, Poland;
| | - Tomasz Dzieciatkowski
- Department of Medical Microbiology, Medical University of Warsaw, 02-091 Warsaw, Poland;
| | - Lukasz Dziewit
- Department of Environmental Microbiology and Biotechnology, Institute of Microbiology, Faculty of Biology, University of Warsaw, 02-096 Warsaw, Poland
| | - Grzegorz W. Basak
- Department of Hematology, Oncology and Internal Medicine, Medical University of Warsaw, 02-091 Warsaw, Poland;
| |
Collapse
|
190
|
Abstract
Abstract
Purpose of Review
We examine recent developments in the treatment of cirrhosis by gut microbiome manipulation specifically focusing on the phase 1 safety and feasibility trials of faecal microbiota transplantation (FMT). We interrogate the published data so far on its feasibility, safety and efficacy.
Recent Findings
A large number of trials have demonstrated the efficacy of FMT in treating recurrent Clostridium difficile infection which is now considered standard of care. In cirrhosis, FMT is still being evaluated and there are a number of clinical trials underway. There are two phase 1 pilot safety studies that have been published with promising findings. However, the importance of rigorously testing donor stool for the presence of multi-drug resistant species has been highlighted and lessons have been learned.
Summary
For those patients with cirrhosis, replacing an unhealthy gut microbiome with a healthy one offers a promising antibiotic-free treatment that may reduce bacterial translocation and endotoxemia.
Collapse
|
191
|
McQuade JL, Ologun GO, Arora R, Wargo JA. Gut Microbiome Modulation Via Fecal Microbiota Transplant to Augment Immunotherapy in Patients with Melanoma or Other Cancers. Curr Oncol Rep 2020; 22:74. [PMID: 32577835 PMCID: PMC7685568 DOI: 10.1007/s11912-020-00913-y] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
PURPOSE OF REVIEW We review emerging evidence regarding the impact of gut microbes on antitumor immunity, and ongoing efforts to translate this in clinical trials. RECENT FINDINGS Pre-clinical models and human cohort studies support a role for gut microbes in modulating overall immunity and immunotherapy response, and numerous trials are now underway exploring strategies to modulate gut microbes to enhance responses to cancer therapy. This includes the use of fecal microbiota transplant (FMT), which is being used to treat patients with Clostridium difficile infection among other non-cancer indications. The use of FMT is now being extended to modulate gut microbes in patients being treated with cancer immunotherapy, with the goal of enhancing responses and/or to ameliorate toxicity. However, significant complexities exist with such an approach and will be discussed herein. Data from ongoing studies of FMT in cancer will provide critical insights for optimization of this approach.
Collapse
Affiliation(s)
- Jennifer L McQuade
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Unit 0430, Houston, 77030, TX, USA.
| | - Gabriel O Ologun
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, 1400 Pressler Street, Unit Number 1484, Houston, TX, 77030, USA
| | - Reetakshi Arora
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, 1400 Pressler Street, Unit Number 1484, Houston, TX, 77030, USA
| | - Jennifer A Wargo
- Department of Surgical Oncology and Genomic Medicine, The University of Texas MD Anderson Cancer Center, 1400 Pressler Street, Unit Number 1484, Houston, TX, 77030, USA
| |
Collapse
|
192
|
Bibbò S, Settanni CR, Porcari S, Bocchino E, Ianiro G, Cammarota G, Gasbarrini A. Fecal Microbiota Transplantation: Screening and Selection to Choose the Optimal Donor. J Clin Med 2020; 9:jcm9061757. [PMID: 32517023 PMCID: PMC7356099 DOI: 10.3390/jcm9061757] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 05/30/2020] [Accepted: 06/02/2020] [Indexed: 12/11/2022] Open
Abstract
In the past decade, fecal microbiota transplantation (FMT) has rapidly spread worldwide in clinical practice as a highly effective treatment option against recurrent Clostridioides difficile infection. Moreover, new evidence also supports a role for FMT in other conditions, such as inflammatory bowel disease, functional gastrointestinal disorders, or metabolic disorders. Recently, some studies have identified specific microbial characteristics associated with clinical improvement after FMT, in different disorders, paving the way for a microbiota-based precision medicine approach. Moreover, donor screening has become increasingly more complex over years, along with standardization of FMT and the increasing number of stool banks. In this narrative review, we discuss most recent evidence on the screening and selection of the stool donor, with reference to recent studies that have identified specific microbiological features for clinical conditions such as Clostridioides difficile infection, irritable bowel syndrome, inflammatory bowel disease, and metabolic disorders.
Collapse
Affiliation(s)
- Stefano Bibbò
- UOC Medicina Interna e Gastroenterologia, Dipartimento di Scienze Mediche e Chirurgiche, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Roma, Italy; (S.B.); (C.R.S.); (S.P.); (G.I.); (G.C.)
- Istituto di Patologia Speciale Medica, Università Cattolica del Sacro Cuore, 00168 Roma, Italy;
| | - Carlo Romano Settanni
- UOC Medicina Interna e Gastroenterologia, Dipartimento di Scienze Mediche e Chirurgiche, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Roma, Italy; (S.B.); (C.R.S.); (S.P.); (G.I.); (G.C.)
- Istituto di Patologia Speciale Medica, Università Cattolica del Sacro Cuore, 00168 Roma, Italy;
| | - Serena Porcari
- UOC Medicina Interna e Gastroenterologia, Dipartimento di Scienze Mediche e Chirurgiche, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Roma, Italy; (S.B.); (C.R.S.); (S.P.); (G.I.); (G.C.)
- Istituto di Patologia Speciale Medica, Università Cattolica del Sacro Cuore, 00168 Roma, Italy;
| | - Enrico Bocchino
- Istituto di Patologia Speciale Medica, Università Cattolica del Sacro Cuore, 00168 Roma, Italy;
| | - Gianluca Ianiro
- UOC Medicina Interna e Gastroenterologia, Dipartimento di Scienze Mediche e Chirurgiche, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Roma, Italy; (S.B.); (C.R.S.); (S.P.); (G.I.); (G.C.)
- Istituto di Patologia Speciale Medica, Università Cattolica del Sacro Cuore, 00168 Roma, Italy;
| | - Giovanni Cammarota
- UOC Medicina Interna e Gastroenterologia, Dipartimento di Scienze Mediche e Chirurgiche, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Roma, Italy; (S.B.); (C.R.S.); (S.P.); (G.I.); (G.C.)
- Istituto di Patologia Speciale Medica, Università Cattolica del Sacro Cuore, 00168 Roma, Italy;
| | - Antonio Gasbarrini
- UOC Medicina Interna e Gastroenterologia, Dipartimento di Scienze Mediche e Chirurgiche, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Roma, Italy; (S.B.); (C.R.S.); (S.P.); (G.I.); (G.C.)
- Istituto di Patologia Speciale Medica, Università Cattolica del Sacro Cuore, 00168 Roma, Italy;
- Correspondence:
| |
Collapse
|
193
|
Fakhoury HMA, Kvietys PR, AlKattan W, Anouti FA, Elahi MA, Karras SN, Grant WB. Vitamin D and intestinal homeostasis: Barrier, microbiota, and immune modulation. J Steroid Biochem Mol Biol 2020; 200:105663. [PMID: 32194242 DOI: 10.1016/j.jsbmb.2020.105663] [Citation(s) in RCA: 123] [Impact Index Per Article: 24.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2019] [Revised: 02/28/2020] [Accepted: 03/13/2020] [Indexed: 02/07/2023]
Abstract
Vitamin D plays a pivotal role in intestinal homeostasis. Vitamin D can impact the function of virtually every cell in the gut by binding to its intracellular receptor (VDR) and subsequently transcribing relevant genes. In the lumen, the mucus layer and the underlying epithelium serve to keep resident microbiota at bay. Vitamin D ensures an appropriate level of antimicrobial peptides in the mucus and maintains epithelial integrity by reinforcing intercellular junctions. Should bacteria penetrate the epithelial layer and enter the interstitium, immune sentinel cells (e.g. macrophages, dendritic cells, and innate lymphoid cells) elicit inflammation and trigger the adaptive immune response by activating Th1/Th17 cells. Vitamin D/VDR signaling in these cells ensures clearance of the bacteria. Subsequently, vitamin D also quiets the adaptive immune system by suppressing the Th1/Th17 cells and favoring Treg cells. The importance of vitamin D/VDR signaling in intestinal homeostasis is evidenced by the development of a chronic inflammatory state (e.g. IBD) when this signaling system is disrupted.
Collapse
Affiliation(s)
- Hana M A Fakhoury
- Department of Biochemistry and Molecular Medicine, College of Medicine, Alfaisal University, Riyadh, Saudi Arabia.
| | - Peter R Kvietys
- Department of Physiology, College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
| | - Wael AlKattan
- Department of Surgery, College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
| | - Fatme Al Anouti
- College of Natural and Health Sciences, Department of Public Health and Nutrition, Zayed University, Abu Dhabi, United Arab Emirates
| | - Muhammad Affan Elahi
- Department of Biochemistry and Molecular Medicine, College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
| | - Spyridon N Karras
- Division of Endocrinology and Metabolism and Diabetes Center, First Department of Internal Medicine, Medical School, Aristotle University of Thessaloniki, AHEPA University Hospital, Thessaloniki, Greece
| | - William B Grant
- Director, Sunlight, Nutrition, and Health Research Center, San Francisco, CA, USA
| |
Collapse
|
194
|
Philips CA, Ahamed R, Rajesh S, Augustine P. 'You know my name, but not my story' - Deciding on an accurate nomenclature for faecal microbiota transplantation. J Hepatol 2020; 72:1212-1213. [PMID: 32197801 DOI: 10.1016/j.jhep.2020.02.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Accepted: 02/06/2020] [Indexed: 12/23/2022]
Affiliation(s)
- Cyriac Abby Philips
- The Liver Unit and Monarch Liver Lab, Department of Clinical and Experimental Gastroenterology and Hepatology, The Cochin Gastroenterology Group, Ernakulam Medical Centre, Kochi, Kerala, India.
| | - Rizwan Ahamed
- The Liver Unit and Monarch Liver Lab, Department of Clinical and Experimental Gastroenterology and Hepatology, The Cochin Gastroenterology Group, Ernakulam Medical Centre, Kochi, Kerala, India
| | - Sasidharan Rajesh
- The Liver Unit and Monarch Liver Lab, Department of Clinical and Experimental Gastroenterology and Hepatology, The Cochin Gastroenterology Group, Ernakulam Medical Centre, Kochi, Kerala, India
| | - Philip Augustine
- The Liver Unit and Monarch Liver Lab, Department of Clinical and Experimental Gastroenterology and Hepatology, The Cochin Gastroenterology Group, Ernakulam Medical Centre, Kochi, Kerala, India
| |
Collapse
|
195
|
Okahara K, Ishikawa D, Nomura K, Ito S, Haga K, Takahashi M, Shibuya T, Osada T, Nagahara A. Matching between Donors and Ulcerative Colitis Patients Is Important for Long-Term Maintenance after Fecal Microbiota Transplantation. J Clin Med 2020; 9:jcm9061650. [PMID: 32486476 PMCID: PMC7355579 DOI: 10.3390/jcm9061650] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 05/27/2020] [Accepted: 05/28/2020] [Indexed: 12/11/2022] Open
Abstract
We previously demonstrated that fresh fecal microbiota transplantation (FMT) following triple antibiotic therapy (amoxicillin, fosfomycin, metronidazole (AFM); A-FMT) resulted in effective colonization of Bacteroidetes species, leading to short-term clinical response in ulcerative colitis (UC). Its long-term efficacy and criteria for donor selection are unknown. Here, we analyzed the long-term efficacy of A-FMT compared to AFM monotherapy (mono-AFM). AFM was administered to patients with mild to severe UC for 2 weeks until 2 days before fresh FMT. Clinical response and efficacy maintenance were defined by the decrease and no exacerbation in clinical activity index. The population for intention-to-treat analysis comprised 92 patients (A-FMT, n = 55; mono-AFM, n = 37). Clinical response was observed at 4 weeks post-treatment (A-FMT, 56.3%; mono-AFM, 48.6%). Maintenance rate of responders at 24 months post-treatment was significantly higher with A-FMT than mono-AFM (p = 0.034). Significant differences in maintenance rate according to the age difference between donors and patients were observed. Additionally, sibling FMT had a significantly higher maintenance rate than parent–child FMT. Microbial analysis of patients who achieved long-term maintenance showed that some exhibited similarity to their donors, particularly Bacteroidetes species. Thus, A-FMT exhibited long-term efficacy. Therefore, matching between donors and UC patients may be helpful in effectively planning the FMT regimen.
Collapse
Affiliation(s)
| | - Dai Ishikawa
- Correspondence: ; Tel.: +81-(0)3-5802-1060; Fax: +81-(0)3-3813-8862
| | | | | | | | | | | | | | | |
Collapse
|
196
|
Hunter P. A war of attrition against antibiotic resistance: Current strategies try to keep antibiotic resistance at bay and further encourage research to produce genuinely novel antibacterials. EMBO Rep 2020; 21:e50807. [PMID: 32449264 DOI: 10.15252/embr.202050807] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
To fight antibiotic resistance, novel drugs are urgently needed. Regulatory agencies are addressing the economic problems and offer new incentives for developing new antibiotics.
Collapse
|
197
|
Vernocchi P, Del Chierico F, Putignani L. Gut Microbiota Metabolism and Interaction with Food Components. Int J Mol Sci 2020; 21:ijms21103688. [PMID: 32456257 PMCID: PMC7279363 DOI: 10.3390/ijms21103688] [Citation(s) in RCA: 94] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 05/12/2020] [Accepted: 05/14/2020] [Indexed: 12/13/2022] Open
Abstract
The human gut contains trillions of microbes that play a central role in host biology, including the provision of key nutrients from the diet. Food is a major source of precursors for metabolite production; in fact, diet modulates the gut microbiota (GM) as the nutrients, derived from dietary intake, reach the GM, affecting both the ecosystem and microbial metabolic profile. GM metabolic ability has an impact on human nutritional status from childhood. However, there is a wide variability of dietary patterns that exist among individuals. The study of interactions with the host via GM metabolic pathways is an interesting field of research in medicine, as microbiota members produce myriads of molecules with many bioactive properties. Indeed, much evidence has demonstrated the importance of metabolites produced by the bacterial metabolism from foods at the gut level that dynamically participate in various biochemical mechanisms of a cell as a reaction to environmental stimuli. Hence, the GM modulate homeostasis at the gut level, and the alteration in their composition can concur in disease onset or progression, including immunological, inflammatory, and metabolic disorders, as well as cancer. Understanding the gut microbe–nutrient interactions will increase our knowledge of how diet affects host health and disease, thus enabling personalized therapeutics and nutrition.
Collapse
Affiliation(s)
- Pamela Vernocchi
- Unit of Human Microbiome, Bambino Gesù Children’s Hospital, IRCCS, Viale San Paolo 15, 00146 Rome, Italy;
- Correspondence: ; Tel.: +39-0668-594061; Fax: +39-0668-592218
| | - Federica Del Chierico
- Unit of Human Microbiome, Bambino Gesù Children’s Hospital, IRCCS, Viale San Paolo 15, 00146 Rome, Italy;
| | - Lorenza Putignani
- Unit of Parasitology and Unit of Human Microbiome, Bambino Gesù Children’s Hospital, IRCCS, Piazza Sant’ Onofrio 4, 00165 Rome, Italy;
| |
Collapse
|
198
|
Castledine M, Sierocinski P, Padfield D, Buckling A. Community coalescence: an eco-evolutionary perspective. Philos Trans R Soc Lond B Biol Sci 2020; 375:20190252. [PMID: 32200752 PMCID: PMC7133537 DOI: 10.1098/rstb.2019.0252] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/22/2020] [Indexed: 12/17/2022] Open
Abstract
Community coalescence, the mixing of different communities, is widespread throughout microbial ecology. Coalescence can result in approximately equal contributions from the founding communities or dominance of one community over another. These different outcomes have ramifications for community structure and function in natural communities, and the use of microbial communities in biotechnology and medicine. However, we have little understanding of when a particular outcome might be expected. Here, we integrate existing theory and data to speculate on how a crucial characteristic of microbial communities-the type of species interaction that dominates the community-might affect the outcome of microbial community coalescence. Given the often comparable timescales of microbial ecology and microevolution, we explicitly consider ecological and evolutionary dynamics, and their interplay, in determining coalescence outcomes. This article is part of the theme issue 'Conceptual challenges in microbial community ecology'.
Collapse
Affiliation(s)
- Meaghan Castledine
- College of Life and Environmental Sciences, Environment and Sustainability Institute, University of Exeter, Penryn, Cornwall TR10 9EZ, UK
| | | | | | | |
Collapse
|
199
|
Luck B, Engevik MA, Ganesh BP, Lackey EP, Lin T, Balderas M, Major A, Runge J, Luna RA, Sillitoe RV, Versalovic J. Bifidobacteria shape host neural circuits during postnatal development by promoting synapse formation and microglial function. Sci Rep 2020; 10:7737. [PMID: 32385412 PMCID: PMC7210968 DOI: 10.1038/s41598-020-64173-3] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Accepted: 04/12/2020] [Indexed: 12/17/2022] Open
Abstract
We hypothesized that early-life gut microbiota support the functional organization of neural circuitry in the brain via regulation of synaptic gene expression and modulation of microglial functionality. Germ-free mice were colonized as neonates with either a simplified human infant microbiota consortium consisting of four Bifidobacterium species, or with a complex, conventional murine microbiota. We examined the cerebellum, cortex, and hippocampus of both groups of colonized mice in addition to germ-free control mice. At postnatal day 4 (P4), conventionalized mice and Bifidobacterium-colonized mice exhibited decreased expression of synapse-promoting genes and increased markers indicative of reactive microglia in the cerebellum, cortex and hippocampus relative to germ-free mice. By P20, both conventional and Bifidobacterium-treated mice exhibited normal synaptic density and neuronal activity as measured by density of VGLUT2+ puncta and Purkinje cell firing rate respectively, in contrast to the increased synaptic density and decreased firing rate observed in germ-free mice. The conclusions from this study further reveal how bifidobacteria participate in establishing functional neural circuits. Collectively, these data indicate that neonatal microbial colonization of the gut elicits concomitant effects on the host CNS, which promote the homeostatic developmental balance of neural connections during the postnatal time period.
Collapse
Affiliation(s)
- Berkley Luck
- Department of Pathology, Texas Children's Hospital, Houston, Texas, United States of America
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, Texas, United States of America
- Integrative Molecular and Biomedical Sciences (IMBS), Baylor College of Medicine, Houston, Texas, United States of America
| | - Melinda A Engevik
- Department of Pathology, Texas Children's Hospital, Houston, Texas, United States of America.
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, Texas, United States of America.
| | - Bhanu Priya Ganesh
- Department of Neurology, University of Texas Health Science Center, Houston, Texas, United States of America
| | - Elizabeth P Lackey
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, Texas, United States of America
- Department of Neuroscience, Baylor College of Medicine, Houston, Texas, United States of America
| | - Tao Lin
- Department of Pathology, Texas Children's Hospital, Houston, Texas, United States of America
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, Texas, United States of America
| | - Miriam Balderas
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, Texas, United States of America
- Texas Children's Microbiome Center, Texas Children's Hospital, Houston, Texas, United States of America
| | - Angela Major
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, Texas, United States of America
| | - Jessica Runge
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, Texas, United States of America
- Program in Developmental Biology, Baylor College of Medicine, Houston, Texas, United States of America
| | - Ruth Ann Luna
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, Texas, United States of America
- Program in Developmental Biology, Baylor College of Medicine, Houston, Texas, United States of America
| | - Roy V Sillitoe
- Department of Pathology, Texas Children's Hospital, Houston, Texas, United States of America
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, Texas, United States of America
- Department of Neuroscience, Baylor College of Medicine, Houston, Texas, United States of America
- Program in Developmental Biology, Baylor College of Medicine, Houston, Texas, United States of America
| | - James Versalovic
- Department of Pathology, Texas Children's Hospital, Houston, Texas, United States of America
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, Texas, United States of America
- Texas Children's Microbiome Center, Texas Children's Hospital, Houston, Texas, United States of America
| |
Collapse
|
200
|
El-Salhy M, Hatlebakk JG, Gilja OH, Bråthen Kristoffersen A, Hausken T. Efficacy of faecal microbiota transplantation for patients with irritable bowel syndrome in a randomised, double-blind, placebo-controlled study. Gut 2020; 69:859-867. [PMID: 31852769 PMCID: PMC7229896 DOI: 10.1136/gutjnl-2019-319630] [Citation(s) in RCA: 286] [Impact Index Per Article: 57.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2019] [Revised: 11/16/2019] [Accepted: 12/03/2019] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Faecal microbiota transplantation (FMT) from healthy donors to patients with irritable bowel syndrome (IBS) has been attempted in two previous double-blind, placebo-controlled studies. While one of those studies found improvement of the IBS symptoms, the other found no effect. The present study was conducted to clarify these contradictory findings. DESIGN This randomised, double-blind, placebo-controlled study randomised 165 patients with IBS to placebo (own faeces), 30 g FMT or 60 g FMT at a ratio of 1:1:1. The material for FMT was obtained from one healthy, well-characterised donor, frozen and administered via gastroscope. The primary outcome was a reduction in the IBS symptoms at 3 months after FMT (response). A response was defined as a decrease of 50 or more points in the total IBS symptom score. The secondary outcome was a reduction in the dysbiosis index (DI) and a change in the intestinal bacterial profile, analysed by 16S rRNA gene sequencing, at 1 month following FMT. RESULTS Responses occurred in 23.6%, 76.9% (p<0.0001) and 89.1% (p<00.0001) of the patients who received placebo, 30 g FMT and 60 g FMT, respectively. These were accompanied by significant improvements in fatigue and the quality of life in patients who received FMT. The intestinal bacterial profiles changed also significantly in the groups received FMT. The FMT adverse events were mild self-limiting gastrointestinal symptoms. CONCLUSIONS FMT is an effective treatment for patients with IBS. Utilising a well-defined donor with a normal DI and favourable specific microbial signature is essential for successful FMT. The response to FMT increases with the dose. Trial registration www.clinicaltrials.gov (NCT03822299) and www.cristin.no (ID657402).
Collapse
Affiliation(s)
- Magdy El-Salhy
- Stord Hospital, Stord, Norway .,Department of Clinical Medicine, University of Bergen Faculty of Medicine and Dentistry, Bergen, Norway
| | - Jan Gunnar Hatlebakk
- Department of Clinical Medicine, University of Bergen Faculty of Medicine and Dentistry, Bergen, Norway
| | - Odd Helge Gilja
- Department of Clinical Medicine, University of Bergen Faculty of Medicine and Dentistry, Bergen, Norway
| | | | - Trygve Hausken
- Department of Clinical Medicine, University of Bergen Faculty of Medicine and Dentistry, Bergen, Norway
| |
Collapse
|