51
|
Chang CW, Lee HC, Li LH, Chiang Chiau JS, Wang TE, Chuang WH, Chen MJ, Wang HY, Shih SC, Liu CY, Tsai TH, Chen YJ. Fecal Microbiota Transplantation Prevents Intestinal Injury, Upregulation of Toll-Like Receptors, and 5-Fluorouracil/Oxaliplatin-Induced Toxicity in Colorectal Cancer. Int J Mol Sci 2020; 21:386. [PMID: 31936237 PMCID: PMC7013718 DOI: 10.3390/ijms21020386] [Citation(s) in RCA: 136] [Impact Index Per Article: 27.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2019] [Revised: 12/18/2019] [Accepted: 01/06/2020] [Indexed: 02/07/2023] Open
Abstract
FOLFOX (5-fluorouracil, leucovorin, and oxaliplatin), a 5-fluorouracil (5-FU)-based chemotherapy regimen, is one of most common therapeutic regimens for colorectal cancer. However, intestinal mucositis is a common adverse effect for which no effective preventive strategies exist. Moreover, the efficacy and the safety of fecal microbiota transplants (FMT) in cancer patients treated with anti-neoplastic agents are still scant. We investigated the effect of FMT on FOLFOX-induced mucosal injury. BALB/c mice implanted with syngeneic CT26 colorectal adenocarcinoma cells were orally administered FMT daily during and two days after five-day injection of FOLFOX regimen for seven days. Administration of FOLFOX significantly induced marked levels of diarrhea and intestinal injury. FMT reduced the severity of diarrhea and intestinal mucositis. Additionally, the number of goblet cells and zonula occludens-1 decreased, while apoptotic and NF-κB-positive cells increased following FOLFOX treatment. The expression of toll-like receptors (TLRs), MyD88, and serum IL-6 were upregulated following FOLFOX treatment. These responses were attenuated following FMT. The disrupted fecal gut microbiota composition was also restored by FMT after FOLFOX treatment. Importantly, FMT did not cause bacteremia and safely alleviated FOLFOX-induced intestinal mucositis in colorectal cancer-bearing mice. The putative mechanism may involve the gut microbiota TLR-MyD88-NF-κB signaling pathway in mice with implanted colorectal carcinoma cells.
Collapse
Affiliation(s)
- Ching-Wei Chang
- Institute of Traditional Medicine, National Yang-Ming University, Taipei 11221, Taiwan;
- Division of Gastroenterology, Department of Internal Medicine, MacKay Memorial Hospital, Taipei 10449, Taiwan; (T.-E.W.); (M.-J.C.); (H.-Y.W.); (S.-C.S.)
- Department of Medical Research, MacKay Memorial Hospital, New Taipei City 25173, Taiwan; (H.-C.L.); (L.-H.L.); (J.-S.C.C.)
- Department of Medicine, MacKay Medical College, New Taipei City 25245, Taiwan
- MacKay Junior College of Medicine, Nursing, and Management, New Taipei City 11260, Taiwan
| | - Hung-Chang Lee
- Department of Medical Research, MacKay Memorial Hospital, New Taipei City 25173, Taiwan; (H.-C.L.); (L.-H.L.); (J.-S.C.C.)
- Department of Medicine, MacKay Medical College, New Taipei City 25245, Taiwan
- MacKay Junior College of Medicine, Nursing, and Management, New Taipei City 11260, Taiwan
- MacKay Children’s Hospital, Taipei 10449, Taiwan
| | - Li-Hui Li
- Department of Medical Research, MacKay Memorial Hospital, New Taipei City 25173, Taiwan; (H.-C.L.); (L.-H.L.); (J.-S.C.C.)
| | - Jen-Shiu Chiang Chiau
- Department of Medical Research, MacKay Memorial Hospital, New Taipei City 25173, Taiwan; (H.-C.L.); (L.-H.L.); (J.-S.C.C.)
| | - Tsang-En Wang
- Division of Gastroenterology, Department of Internal Medicine, MacKay Memorial Hospital, Taipei 10449, Taiwan; (T.-E.W.); (M.-J.C.); (H.-Y.W.); (S.-C.S.)
- Department of Medicine, MacKay Medical College, New Taipei City 25245, Taiwan
- MacKay Junior College of Medicine, Nursing, and Management, New Taipei City 11260, Taiwan
| | - Wei-Hung Chuang
- Institute of Biomedical Informatics, Center for Systems and Synthetic Biology, National Yang-Ming University, Taipei 11221, Taiwan;
| | - Ming-Jen Chen
- Division of Gastroenterology, Department of Internal Medicine, MacKay Memorial Hospital, Taipei 10449, Taiwan; (T.-E.W.); (M.-J.C.); (H.-Y.W.); (S.-C.S.)
- Department of Medicine, MacKay Medical College, New Taipei City 25245, Taiwan
- MacKay Junior College of Medicine, Nursing, and Management, New Taipei City 11260, Taiwan
| | - Horng-Yuan Wang
- Division of Gastroenterology, Department of Internal Medicine, MacKay Memorial Hospital, Taipei 10449, Taiwan; (T.-E.W.); (M.-J.C.); (H.-Y.W.); (S.-C.S.)
- Department of Medicine, MacKay Medical College, New Taipei City 25245, Taiwan
- MacKay Junior College of Medicine, Nursing, and Management, New Taipei City 11260, Taiwan
| | - Shou-Chuan Shih
- Division of Gastroenterology, Department of Internal Medicine, MacKay Memorial Hospital, Taipei 10449, Taiwan; (T.-E.W.); (M.-J.C.); (H.-Y.W.); (S.-C.S.)
- Department of Medicine, MacKay Medical College, New Taipei City 25245, Taiwan
- MacKay Junior College of Medicine, Nursing, and Management, New Taipei City 11260, Taiwan
| | - Chia-Yuan Liu
- Institute of Traditional Medicine, National Yang-Ming University, Taipei 11221, Taiwan;
- Division of Gastroenterology, Department of Internal Medicine, MacKay Memorial Hospital, Taipei 10449, Taiwan; (T.-E.W.); (M.-J.C.); (H.-Y.W.); (S.-C.S.)
- Department of Medical Research, MacKay Memorial Hospital, New Taipei City 25173, Taiwan; (H.-C.L.); (L.-H.L.); (J.-S.C.C.)
- Department of Medicine, MacKay Medical College, New Taipei City 25245, Taiwan
- MacKay Junior College of Medicine, Nursing, and Management, New Taipei City 11260, Taiwan
| | - Tung-Hu Tsai
- Institute of Traditional Medicine, National Yang-Ming University, Taipei 11221, Taiwan;
- Department of Chemical Engineering, National United University, Miaoli 36063, Taiwan
| | - Yu-Jen Chen
- Institute of Traditional Medicine, National Yang-Ming University, Taipei 11221, Taiwan;
- Department of Medical Research, MacKay Memorial Hospital, New Taipei City 25173, Taiwan; (H.-C.L.); (L.-H.L.); (J.-S.C.C.)
- Department of Medicine, MacKay Medical College, New Taipei City 25245, Taiwan
- MacKay Junior College of Medicine, Nursing, and Management, New Taipei City 11260, Taiwan
- Department of Radiation Oncology, MacKay Memorial Hospital, Taipei 10449, Taiwan
| |
Collapse
|
52
|
Akhtar M, Haleem A, Anvari S, Nazli A, Sager M. An analysis of gut dysbiosis in obesity, diabetes, and chronic gut conditions. IBNOSINA JOURNAL OF MEDICINE AND BIOMEDICAL SCIENCES 2020. [DOI: 10.4103/ijmbs.ijmbs_102_20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
|
53
|
Goloshchapov OV, Olekhnovich EI, Sidorenko SV, Moiseev IS, Kucher MA, Fedorov DE, Pavlenko AV, Manolov AI, Gostev VV, Veselovsky VA, Klimina KM, Kostryukova ES, Bakin EA, Shvetcov AN, Gumbatova ED, Klementeva RV, Shcherbakov AA, Gorchakova MV, Egozcue JJ, Pawlowsky-Glahn V, Suvorova MA, Chukhlovin AB, Govorun VM, Ilina EN, Afanasyev BV. Long-term impact of fecal transplantation in healthy volunteers. BMC Microbiol 2019; 19:312. [PMID: 31888470 PMCID: PMC6938016 DOI: 10.1186/s12866-019-1689-y] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Accepted: 12/17/2019] [Indexed: 02/10/2023] Open
Abstract
BACKGROUND Fecal microbiota transplantation (FMT) has been recently approved by FDA for the treatment of refractory recurrent clostridial colitis (rCDI). Success of FTM in treatment of rCDI led to a number of studies investigating the effectiveness of its application in the other gastrointestinal diseases. However, in the majority of studies the effects of FMT were evaluated on the patients with initially altered microbiota. The aim of our study was to estimate effects of FMT on the gut microbiota composition in healthy volunteers and to monitor its long-term outcomes. RESULTS We have performed a combined analysis of three healthy volunteers before and after capsule FMT by evaluating their general condition, adverse clinical effects, changes of basic laboratory parameters, and several immune markers. Intestinal microbiota samples were evaluated by 16S rRNA gene and shotgun sequencing. The data analysis demonstrated profound shift towards the donor microbiota taxonomic composition in all volunteers. Following FMT, all the volunteers exhibited gut colonization with donor gut bacteria and persistence of this effect for almost ∼1 year of observation. Transient changes of immune parameters were consistent with suppression of T-cell cytotoxicity. FMT was well tolerated with mild gastrointestinal adverse events, however, one volunteer developed a systemic inflammatory response syndrome. CONCLUSIONS The FMT leads to significant long-term changes of the gut microbiota in healthy volunteers with the shift towards donor microbiota composition and represents a relatively safe procedure to the recipients without long-term adverse events.
Collapse
Affiliation(s)
- Oleg V. Goloshchapov
- R.M.Gorbacheva Memorial Institute of Oncology, Hematology and Transplantation, Pavlov First Saint Petersburg State Medical University, St. Petersburg, Russian Federation
| | - Evgenii I. Olekhnovich
- Federal Research and Clinical Centre of Physical and Chemical Medicine of Federal Medical and Biological Agency of Russia, Moscow, Russian Federation
| | - Sergey V. Sidorenko
- Pediatric Research and Clinical Center for Infectious Diseases, St. Petersburg, Russia
- Mechnikov North-West State Medical University, St. Petersburg, Russia
| | - Ivan S. Moiseev
- R.M.Gorbacheva Memorial Institute of Oncology, Hematology and Transplantation, Pavlov First Saint Petersburg State Medical University, St. Petersburg, Russian Federation
| | - Maxim A. Kucher
- R.M.Gorbacheva Memorial Institute of Oncology, Hematology and Transplantation, Pavlov First Saint Petersburg State Medical University, St. Petersburg, Russian Federation
| | - Dmitry E. Fedorov
- Federal Research and Clinical Centre of Physical and Chemical Medicine of Federal Medical and Biological Agency of Russia, Moscow, Russian Federation
| | - Alexander V. Pavlenko
- Federal Research and Clinical Centre of Physical and Chemical Medicine of Federal Medical and Biological Agency of Russia, Moscow, Russian Federation
| | - Alexander I. Manolov
- Federal Research and Clinical Centre of Physical and Chemical Medicine of Federal Medical and Biological Agency of Russia, Moscow, Russian Federation
| | - Vladimir V. Gostev
- Pediatric Research and Clinical Center for Infectious Diseases, St. Petersburg, Russia
- Mechnikov North-West State Medical University, St. Petersburg, Russia
| | - Vladimir A. Veselovsky
- Federal Research and Clinical Centre of Physical and Chemical Medicine of Federal Medical and Biological Agency of Russia, Moscow, Russian Federation
| | - Ksenia M. Klimina
- Federal Research and Clinical Centre of Physical and Chemical Medicine of Federal Medical and Biological Agency of Russia, Moscow, Russian Federation
| | - Elena S. Kostryukova
- Federal Research and Clinical Centre of Physical and Chemical Medicine of Federal Medical and Biological Agency of Russia, Moscow, Russian Federation
| | - Evgeny A. Bakin
- R.M.Gorbacheva Memorial Institute of Oncology, Hematology and Transplantation, Pavlov First Saint Petersburg State Medical University, St. Petersburg, Russian Federation
| | - Alexander N. Shvetcov
- R.M.Gorbacheva Memorial Institute of Oncology, Hematology and Transplantation, Pavlov First Saint Petersburg State Medical University, St. Petersburg, Russian Federation
| | - Elvira D. Gumbatova
- R.M.Gorbacheva Memorial Institute of Oncology, Hematology and Transplantation, Pavlov First Saint Petersburg State Medical University, St. Petersburg, Russian Federation
| | - Ruslana V. Klementeva
- R.M.Gorbacheva Memorial Institute of Oncology, Hematology and Transplantation, Pavlov First Saint Petersburg State Medical University, St. Petersburg, Russian Federation
| | - Alexander A. Shcherbakov
- R.M.Gorbacheva Memorial Institute of Oncology, Hematology and Transplantation, Pavlov First Saint Petersburg State Medical University, St. Petersburg, Russian Federation
| | - Margarita V. Gorchakova
- R.M.Gorbacheva Memorial Institute of Oncology, Hematology and Transplantation, Pavlov First Saint Petersburg State Medical University, St. Petersburg, Russian Federation
| | | | | | | | - Alexey B. Chukhlovin
- R.M.Gorbacheva Memorial Institute of Oncology, Hematology and Transplantation, Pavlov First Saint Petersburg State Medical University, St. Petersburg, Russian Federation
| | - Vadim M. Govorun
- Federal Research and Clinical Centre of Physical and Chemical Medicine of Federal Medical and Biological Agency of Russia, Moscow, Russian Federation
| | - Elena N. Ilina
- Federal Research and Clinical Centre of Physical and Chemical Medicine of Federal Medical and Biological Agency of Russia, Moscow, Russian Federation
| | - Boris V. Afanasyev
- R.M.Gorbacheva Memorial Institute of Oncology, Hematology and Transplantation, Pavlov First Saint Petersburg State Medical University, St. Petersburg, Russian Federation
| |
Collapse
|
54
|
Sood A, Mahajan R, Singh A, Midha V, Mehta V, Narang V, Singh T, Singh Pannu A. Role of Faecal Microbiota Transplantation for Maintenance of Remission in Patients With Ulcerative Colitis: A Pilot Study. J Crohns Colitis 2019; 13:1311-1317. [PMID: 30873549 DOI: 10.1093/ecco-jcc/jjz060] [Citation(s) in RCA: 114] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
OBJECTIVES To study the role of faecal microbiota transplantation [FMT] in maintenance of remission in ulcerative colitis [UC]. METHODS In this pilot study, patients with UC in clinical remission achieved after multi-session FMT were randomly allocated to either maintenance FMT or placebo colonoscopic infusion every 8 weeks, for 48 weeks. The standard of care [SOC] therapy was continued in all patients. The primary endpoint was maintenance of steroid-free clinical remission [Mayo score ≤2, all subscores ≤1] at Week 48. Secondary endpoints were achievement of endoscopic remission [endoscopic Mayo score 0] and histological remission [Nancy grade 0, 1] at Week 48. RESULTS In all, 61 patients in clinical remission were randomised to receive either FMT [n = 31] or placebo [n = 30]. The primary outcome was achieved in 27/31 [87.1%] patients allocated FMT versus 20/30 [66.7%] patients assigned placebo [p = 0.111]. Secondary endpoints of endoscopic remission (FMT: 18/31 [58.1%] versus placebo: 8/30 [26.7%], p = 0.026) and histological remission (FMT: 14/31 [45.2%] versus placebo: 5/30 [16.7%], p = 0. 033) were achieved in a significantly higher number of patients with FMT. Three patients receiving FMT [9.7%] and 8 patients on placebo [26.7%] relapsed. There were no serious adverse events necessitating discontinuation in patients on FMT; one patient who relapsed on placebo required colectomy. CONCLUSIONS Maintenance FMT in patients who are in clinical remission may help sustain clinical, endoscopic and histological remission in patients with UC.
Collapse
Affiliation(s)
- Ajit Sood
- Department of Gastroenterology, Dayanand Medical College & Hospital, Ludhiana, Punjab, India
| | - Ramit Mahajan
- Department of Gastroenterology, Dayanand Medical College & Hospital, Ludhiana, Punjab, India
| | - Arshdeep Singh
- Department of Gastroenterology, Dayanand Medical College & Hospital, Ludhiana, Punjab, India
| | - Vandana Midha
- Department of Internal Medicine, Dayanand Medical College & Hospital, Ludhiana, India
| | - Varun Mehta
- Department of Internal Medicine, Dayanand Medical College & Hospital, Ludhiana, India
| | - Vikram Narang
- Department of Pathology, Dayanand Medical College & Hospital, Ludhiana, Punjab, India
| | - Tarundeep Singh
- Department of Community Medicine and School of Public Health, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Anmol Singh Pannu
- Department of Gastroenterology, Dayanand Medical College & Hospital, Ludhiana, Punjab, India
| |
Collapse
|
55
|
Tian Y, Zhou Y, Huang S, Li J, Zhao K, Li X, Wen X, Li XA. Fecal microbiota transplantation for ulcerative colitis: a prospective clinical study. BMC Gastroenterol 2019; 19:116. [PMID: 31272391 PMCID: PMC6610864 DOI: 10.1186/s12876-019-1010-4] [Citation(s) in RCA: 41] [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] [Received: 11/30/2018] [Accepted: 06/04/2019] [Indexed: 12/27/2022] Open
Abstract
Background Fecal microbiota transplantation may contribute to disease remission in ulcerative colitis; however, the factors that determine the effects of treatment remain unknown. The aim of the present study was to prospectively investigate the clinical efficacy of fecal microbiota transplantation in patients with ulcerative colitis and identify the bacterial signatures associated with clinical remission. Methods A total of 20 patients with ulcerative colitis were included in this prospective and uncontrolled study. All patients underwent gastroscopy five times, once every 3 weeks. Clinical indices were used to assess the efficacy of fecal microbiota transplantation, as well as the Mayo score, a score used to evaluate the extent of intestinal mucosal lesions in patients with ulcerative colitis. The changes in intestinal flora were detected by 16S ribosomal RNA-sequencing, and the relationship between ulcerative colitis and intestinal flora was analyzed. Results After treatment, clinical index scores for diarrhea, abdominal pain, and blood stool decreased significantly (p < 0.05). Erythrocyte sedimentation rate and C-reactive protein levels had not changed significantly; however, the clinical index score for intestinal mucosal lesions and the Mayo score decreased significantly. In addition, 16S ribosomal RNA-sequencing revealed that the intestinal flora in patients diagnosed with ulcerative colitis was different from that of donors. Conclusion Fecal microbiota transplantation has a potential therapeutic value for the treatment of ulcerative colitis as it changes the abundance of bacterial flora and improves the scores for diarrhea, abdominal pain, and mucous membrane lesions in patients with this disease. Trial registration The clinical trial was retrospectively registered with ClinicalTrials.gov (NCT03016780) on January 11th, 2017.
Collapse
Affiliation(s)
- Yan Tian
- Department Of Gastroenterology, The Gastroenterology Tumor and Microenvironment Laboratory, The First Affiliated Hospital of Chengdu Medical College, Chengdu Medical College, Baoguang Road 4, Xindu district, Chengdu, 610041, Sichuan, People's Republic of China
| | - Yan Zhou
- Department Of Gastroenterology, The Gastroenterology Tumor and Microenvironment Laboratory, The First Affiliated Hospital of Chengdu Medical College, Chengdu Medical College, Baoguang Road 4, Xindu district, Chengdu, 610041, Sichuan, People's Republic of China
| | - Sisi Huang
- Department Of Gastroenterology, The Gastroenterology Tumor and Microenvironment Laboratory, The First Affiliated Hospital of Chengdu Medical College, Chengdu Medical College, Baoguang Road 4, Xindu district, Chengdu, 610041, Sichuan, People's Republic of China
| | - Jun Li
- Department Of Gastroenterology, The Gastroenterology Tumor and Microenvironment Laboratory, The First Affiliated Hospital of Chengdu Medical College, Chengdu Medical College, Baoguang Road 4, Xindu district, Chengdu, 610041, Sichuan, People's Republic of China
| | - Kui Zhao
- Department Of Gastroenterology, The Gastroenterology Tumor and Microenvironment Laboratory, The First Affiliated Hospital of Chengdu Medical College, Chengdu Medical College, Baoguang Road 4, Xindu district, Chengdu, 610041, Sichuan, People's Republic of China
| | - Xiaohui Li
- Department Of Gastroenterology, The Gastroenterology Tumor and Microenvironment Laboratory, The First Affiliated Hospital of Chengdu Medical College, Chengdu Medical College, Baoguang Road 4, Xindu district, Chengdu, 610041, Sichuan, People's Republic of China
| | - Xiangchen Wen
- Department Of Gastroenterology, The Gastroenterology Tumor and Microenvironment Laboratory, The First Affiliated Hospital of Chengdu Medical College, Chengdu Medical College, Baoguang Road 4, Xindu district, Chengdu, 610041, Sichuan, People's Republic of China
| | - Xiao-An Li
- Department Of Gastroenterology, The Gastroenterology Tumor and Microenvironment Laboratory, The First Affiliated Hospital of Chengdu Medical College, Chengdu Medical College, Baoguang Road 4, Xindu district, Chengdu, 610041, Sichuan, People's Republic of China.
| |
Collapse
|
56
|
Gutin L, Piceno Y, Fadrosh D, Lynch K, Zydek M, Kassam Z, LaMere B, Terdiman J, Ma A, Somsouk M, Lynch S, El-Nachef N. Fecal microbiota transplant for Crohn disease: A study evaluating safety, efficacy, and microbiome profile. United European Gastroenterol J 2019; 7:807-814. [PMID: 31316785 PMCID: PMC6620877 DOI: 10.1177/2050640619845986] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Accepted: 03/29/2019] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND Emerging trials suggest fecal microbiota transplantation (FMT) is a promising treatment for ulcerative colitis; however, there is a paucity of data in Crohn disease (CD). OBJECTIVE The objectives of this article are to determine whether single-dose FMT improves clinical and endoscopic outcomes in CD patients and to identify meaningful changes in the microbiome in response to FMT. METHODS We performed a prospective, open-label, single-center study. Ten CD patients underwent FMT and were evaluated for clinical response (defined as decrease in Harvey-Bradshaw Index score ≥3 at one month post-FMT) and microbiome profile (16S ribosomal RNA sequencing) at one month post-FMT. RESULTS Three of 10 patients responded to FMT. Two of 10 patients had significant adverse events requiring escalation of therapy. On microbiome analysis, bacterial communities of responders had increased relative abundance of bacteria commonly found in donor gut microbiota. CONCLUSIONS Single-dose FMT in this cohort of CD patients showed modest effect and potential for harm. Responders tended to have lower baseline alpha diversity, suggesting baseline perturbation of microbiota may be an indicator of potential responders to FMT in this patient population. Controlled trials are needed to further assess the efficacy and safety of FMT in CD and determine whether FMT is a viable option in this patient population.Clinicaltrials.gov number: NCT02460705.
Collapse
Affiliation(s)
- Liat Gutin
- Department of Medicine, University of
California San Francisco, San Francisco, CA, USA
| | - Yvette Piceno
- Division of Gastroenterology, University
of California San Francisco, San Francisco, CA, USA
| | - Douglas Fadrosh
- Division of Gastroenterology, University
of California San Francisco, San Francisco, CA, USA
| | - Kole Lynch
- Division of Gastroenterology, University
of California San Francisco, San Francisco, CA, USA
| | - Martin Zydek
- Division of Gastroenterology, University
of California San Francisco, San Francisco, CA, USA
| | | | - Brandon LaMere
- Division of Gastroenterology, University
of California San Francisco, San Francisco, CA, USA
| | - Jonathan Terdiman
- Division of Gastroenterology, University
of California San Francisco, San Francisco, CA, USA
| | - Averil Ma
- Division of Gastroenterology, University
of California San Francisco, San Francisco, CA, USA
| | - Ma Somsouk
- Division of Gastroenterology, Zuckerberg
San Francisco General Hospital, San Francisco, CA, USA
| | - Susan Lynch
- Division of Gastroenterology, University
of California San Francisco, San Francisco, CA, USA
| | - Najwa El-Nachef
- Division of Gastroenterology, University
of California San Francisco, San Francisco, CA, USA,Najwa El-Nachef, University of California
San Francisco, Division of Gastroenterology, 505 Parnassus Ave, Box 0119, M1479,
San Francisco, CA 94143-0119.
| |
Collapse
|
57
|
Gibson KM, Nguyen BN, Neumann LM, Miller M, Buss P, Daniels S, Ahn MJ, Crandall KA, Pukazhenthi B. Gut microbiome differences between wild and captive black rhinoceros - implications for rhino health. Sci Rep 2019; 9:7570. [PMID: 31138833 PMCID: PMC6538756 DOI: 10.1038/s41598-019-43875-3] [Citation(s) in RCA: 84] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2019] [Accepted: 04/24/2019] [Indexed: 12/20/2022] Open
Abstract
A number of recent studies have shown the importance of the mammalian gut microbiome in host health. In the context of endangered species, a few studies have examined the relationship between the gut microbiome in wild versus captive populations due to digestive and other health issues. Unfortunately, the results seem to vary across taxa in terms of captive animals having higher, lower, or equivalent microbiome diversity relative to their wild counterparts. Here, we focus on the black rhinoceros as captive animals suffer from a number of potentially dietary related health effects. We compared gut microbiomes of wild and captive black rhinos to test for differences in taxonomic diversity (alpha and beta) and in functional diversity of the microbiome. We incorporated a more powerful metagenomic shotgun sequencing approach rather than a targeted amplification of the 16S gene for taxonomic assignment of the microbiome. Our results showed no significant differences in the alpha diversity levels between wild and captive black rhinos, but significant differences in beta diversity. We found that bacterial taxa traditionally associated with ruminant guts of domesticated animals had higher relative abundances in captive rhinos. Our metagenomic sequencing results suggest that unknown gut microbes of wild rhinos are being replaced by those found in conventional human-domesticated livestock. Wild rhinos have significantly different functional bacterial communities compared to their captive counterparts. Functional profiling results showed greater abundance of glycolysis and amino acid synthesis pathways in captive rhino microbiomes, representing an animal receiving sub-optimal nutrition with a readily available source of glucose but possibly an imbalance of necessary macro and micronutrients. Given the differences observed between wild and captive rhino gut microbiomes, we make a number of recommendations for potentially modifying captive gut microbiome to better reflect their wild counterparts and thereby hopefully improve overall rhino health in captivity.
Collapse
Affiliation(s)
- Keylie M Gibson
- Computational Biology Institute, The Milken Institute School of Public Health, The George Washington University, Washington, DC, USA
- Department of Biological Sciences, The George Washington University, Washington, DC, USA
| | - Bryan N Nguyen
- Computational Biology Institute, The Milken Institute School of Public Health, The George Washington University, Washington, DC, USA
- Department of Biological Sciences, The George Washington University, Washington, DC, USA
| | - Laura M Neumann
- Department of Environmental and Occupational Health, The Milken Institute School of Public Health, The George Washington University, Washington, DC, USA
| | - Michele Miller
- DST-NRF Centre of Excellence for Biomedical Tuberculosis Research; South African Medical Research Council Centre for Tuberculosis Research; Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Peter Buss
- South African National Parks, Veterinary Wildlife Services, Kruger National Park, Skukuza, South Africa
| | - Savel Daniels
- Department of Botany and Zoology, University of Stellenbosch, Private Bag X1, 7602, Matieland, South Africa
| | - Michelle J Ahn
- Computational Biology Institute, The Milken Institute School of Public Health, The George Washington University, Washington, DC, USA
- Department of Biological Sciences, The George Washington University, Washington, DC, USA
| | - Keith A Crandall
- Computational Biology Institute, The Milken Institute School of Public Health, The George Washington University, Washington, DC, USA
- Department of Epidemiology and Biostatistics, The Milken Institute School of Public Health, The George Washington University, Washington, DC, USA
| | - Budhan Pukazhenthi
- Smithsonian's National Zoo and Conservation Biology Institute, Front Royal, VA, USA.
| |
Collapse
|
58
|
Relationship between the microbiome and ocular health. Ocul Surf 2019; 17:384-392. [PMID: 31125783 DOI: 10.1016/j.jtos.2019.05.006] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Revised: 04/08/2019] [Accepted: 05/20/2019] [Indexed: 02/07/2023]
Abstract
The microbiome is important to the host as a whole, both in maintenance of health and in the pathophysiology of disease. The purpose of this review is to explore the relationship between the gut, ocular microbiome, and ocular disease states. We will also discuss how the microbiome can serve as a potential target for treatment, by methods such as modulation of diet, probiotics and fecal microbiota transplantation. The information discussed in the review has been gathered using literature published from 2004 to November 2018, as indexed in PubMed.
Collapse
|
59
|
Ben Y, Fu C, Hu M, Liu L, Wong MH, Zheng C. Human health risk assessment of antibiotic resistance associated with antibiotic residues in the environment: A review. ENVIRONMENTAL RESEARCH 2019; 169:483-493. [PMID: 30530088 DOI: 10.1016/j.envres.2018.11.040] [Citation(s) in RCA: 571] [Impact Index Per Article: 95.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Revised: 11/11/2018] [Accepted: 11/24/2018] [Indexed: 05/28/2023]
Abstract
The extensive use of antibiotics leading to the rapid spread of antibiotic resistance poses high health risks to humans, but to date there is still lack of a quantitative model to properly assess the risks. Concerns over the health risk of antibiotic residues in the environment are mainly (1) the potential hazard of ingested antibiotic residues in the environment altering the human microbiome and promoting emergence and selection for bacteria resistance inhabiting the human body, and (2) the potential hazard of creating a selection pressure on environmental microbiome and leading to reservoirs of antibiotic resistance in the environment. We provide a holistic view of health risk assessment of antibiotic resistance associated with antibiotic residues in the environment in contrast with that of the antibiotic resistant bacteria and discuss the main knowledge gaps and the future research that should be prioritized to achieve the quantitative risk assessment. We examined and summarized the available data and information on the four core elements of antibiotic resistance associated with antibiotic residues in the environment: hazard identification, exposure assessment, dose-response assessment, and risk characterization. The data required to characterize the risks of antibiotic residues in the environment is severely limited. The main future research needs have been identified to enable better assessments of antibiotic resistance associated with antibiotic residues in the environment: (1) establishment of a standardized monitoring guide of antibiotic residues and antibiotic resistance in the environment, (2) derivation of the relationship between antibiotic levels and pathogenic antibiotic-resistance development in different settings, and (3) establishment of the dose-response relationship between pathogenic antibiotic resistant bacteria and various infection diseases. After identification of key risk determinant parameters, we propose a conceptual framework of human health risk assessments of antibiotic residues in the environment. CAPSULE: A holistic view of human health risk assessment of antibiotic residues in the environment was provided.
Collapse
Affiliation(s)
- Yujie Ben
- Guangdong Provincial Key Laboratory of Soil and Groundwater Pollution Control, School of Environmental Science and Engineering, Southern University of Science and Technology, Shenzhen 518055, China; College of Resources and Environment, University of Chinese Academy of Sciences, Beijing 100190, China; State Environmental Protection Key Laboratory of Integrated Surface Water-Groundwater Pollution Control, School of Environmental Science and Engineering, Southern University of Science and Technology, Shenzhen 518055, China
| | - Caixia Fu
- Guangdong Provincial Key Laboratory of Soil and Groundwater Pollution Control, School of Environmental Science and Engineering, Southern University of Science and Technology, Shenzhen 518055, China; State Environmental Protection Key Laboratory of Integrated Surface Water-Groundwater Pollution Control, School of Environmental Science and Engineering, Southern University of Science and Technology, Shenzhen 518055, China
| | - Min Hu
- Guangdong Provincial Key Laboratory of Soil and Groundwater Pollution Control, School of Environmental Science and Engineering, Southern University of Science and Technology, Shenzhen 518055, China; State Environmental Protection Key Laboratory of Integrated Surface Water-Groundwater Pollution Control, School of Environmental Science and Engineering, Southern University of Science and Technology, Shenzhen 518055, China; School of Civil Engineering, Lanzhou University of Technology, Lanzhou 730050, China
| | - Lei Liu
- Guangdong Provincial Key Laboratory of Soil and Groundwater Pollution Control, School of Environmental Science and Engineering, Southern University of Science and Technology, Shenzhen 518055, China; State Environmental Protection Key Laboratory of Integrated Surface Water-Groundwater Pollution Control, School of Environmental Science and Engineering, Southern University of Science and Technology, Shenzhen 518055, China; Environmental Biotechnology Laboratory, The University of Hong Kong, Hong Kong, China
| | - Ming Hung Wong
- Guangdong Provincial Key Laboratory of Soil and Groundwater Pollution Control, School of Environmental Science and Engineering, Southern University of Science and Technology, Shenzhen 518055, China; State Environmental Protection Key Laboratory of Integrated Surface Water-Groundwater Pollution Control, School of Environmental Science and Engineering, Southern University of Science and Technology, Shenzhen 518055, China; Consortium on Health, Environment, Education and Research (CHEER), Department of Science and Environmental Studies, The Education University of Hong Kong, Tai Po, Hong Kong, China
| | - Chunmiao Zheng
- Guangdong Provincial Key Laboratory of Soil and Groundwater Pollution Control, School of Environmental Science and Engineering, Southern University of Science and Technology, Shenzhen 518055, China; State Environmental Protection Key Laboratory of Integrated Surface Water-Groundwater Pollution Control, School of Environmental Science and Engineering, Southern University of Science and Technology, Shenzhen 518055, China.
| |
Collapse
|
60
|
Sood A, Mahajan R, Juyal G, Midha V, Grewal CS, Mehta V, Singh A, Joshi MC, Narang V, Kaur K, Sidhu H. Efficacy of fecal microbiota therapy in steroid dependent ulcerative colitis: a real world intention-to-treat analysis. Intest Res 2018; 17:78-86. [PMID: 30449078 PMCID: PMC6361022 DOI: 10.5217/ir.2018.00089] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Accepted: 08/30/2018] [Indexed: 12/26/2022] Open
Abstract
Background/Aims Four high-quality randomized controlled trials have proven the efficacy of fecal microbiota transplantation (FMT) in active ulcerative colitis (UC). We assessed the efficacy of FMT in a real-world setting involving steroid-dependent patients with UC. Methods This was a single-center prospective analysis of data from steroid-dependent patients with UC treated with FMT from September 2015 to September 2017 at the Dayanand Medical College, a tertiary care center in India. Fecal samples from random unrelated donors were administered through colonoscopy at weeks 0, 2, 6, 10, 14, 18, and 22. The primary outcome was achievement of steroid-free clinical remission, and the secondary outcomes were clinical response and endoscopic remission at 24 weeks. Modified intention-to-treat analysis was performed, which included subjects who underwent at least 1 FMT. Results Of 345 patients with UC treated during the study period, 49 (14.2%) had steroid-dependent UC. Of these 49 patients, 41 underwent FMT: 33 completed 7 sessions over 22 weeks according to the protocol, and 8 discontinued treatment (non-response, 5; lost to follow-up, 2; and fear of adverse effects, 1). At week 24, steroid-free clinical remission was achieved in 19 out of 41 (46.3%) patients, whereas clinical response and endoscopic remission were achieved in 31 out of 41 (75.6%) and 26 out of 41 (63.4%) patients, respectively. All patients with clinical response were able to withdraw steroids. There were no serious adverse events necessitating discontinuation. Conclusions A multisession FMT via the colonoscopic route is a promising therapeutic option for patients with steroid-dependent UC, as it can induce clinical remission and aid in steroid withdrawal.
Collapse
Affiliation(s)
- Ajit Sood
- Department of Gastroenterology, Dayanand Medical College & Hospital, Ludhiana, India
| | - Ramit Mahajan
- Department of Gastroenterology, Dayanand Medical College & Hospital, Ludhiana, India
| | - Garima Juyal
- School of Biotechnology, Jawaharlal Nehru University, New Delhi, India
| | - Vandana Midha
- Department of Internal Medicine, Dayanand Medical College & Hospital, Ludhiana, India
| | | | - Varun Mehta
- Department of Gastroenterology, Dayanand Medical College & Hospital, Ludhiana, India
| | - Arshdeep Singh
- Department of Gastroenterology, Dayanand Medical College & Hospital, Ludhiana, India
| | - Mohan C Joshi
- Multidisciplinary Centre for Advanced Research and Studies (MCARS), Jamia Millia Islamia, New Delhi, India
| | - Vikram Narang
- Department of Pathology, Dayanand Medical College & Hospital, Ludhiana, India
| | - Kirandeep Kaur
- Department of Pharmacology, Dayanand Medical College & Hospital, Ludhiana, India
| | - Hasrat Sidhu
- Department of Internal Medicine, Dayanand Medical College & Hospital, Ludhiana, India
| |
Collapse
|
61
|
Dias AM, Pereira MS, Padrão NA, Alves I, Marcos-Pinto R, Lago P, Pinho SS. Glycans as critical regulators of gut immunity in homeostasis and disease. Cell Immunol 2018; 333:9-18. [DOI: 10.1016/j.cellimm.2018.07.007] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Revised: 07/04/2018] [Accepted: 07/17/2018] [Indexed: 12/19/2022]
|
62
|
Li J, Tang M, Xue Y. Review of the effects of silver nanoparticle exposure on gut bacteria. J Appl Toxicol 2018; 39:27-37. [PMID: 30247756 DOI: 10.1002/jat.3729] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Revised: 08/17/2018] [Accepted: 08/18/2018] [Indexed: 12/11/2022]
Abstract
Gut bacteria are involved in regulating several important physiological functions in the host, and intestinal dysbacteriosis plays an important role in several human diseases, including intestinal, metabolic and autoimmune disorders. Although silver nanoparticles (AgNPs) are increasingly being incorporated into medical and consumer products due to their unique physicochemical properties, studies have indicated their potential to affect adversely the gut bacteria. In this review, we focus on the biotoxicological effects of AgNPs entering the gastrointestinal tract and the relationship of these effects with important nanoscale properties. We discuss in detail the mechanisms underlying the bactericidal toxicity effects of AgNPs and explore the relationships between AgNPs, gut bacteria and disease. Finally, we highlight the need to focus on the negative effects of AgNPs usage to facilitate appropriate development of these particles.
Collapse
Affiliation(s)
- Jiangyan Li
- Key Laboratory of Environmental Medicine and Engineering, Ministry of Education, School of Public Health & Collaborative Innovation Center of Suzhou Nano Science and Technology, Southeast University, Nanjing, 210009, China.,Jiangsu Key Laboratory for Biomaterials and Devices, Southeast University, Nanjing, 210009, China
| | - Meng Tang
- Key Laboratory of Environmental Medicine and Engineering, Ministry of Education, School of Public Health & Collaborative Innovation Center of Suzhou Nano Science and Technology, Southeast University, Nanjing, 210009, China.,Jiangsu Key Laboratory for Biomaterials and Devices, Southeast University, Nanjing, 210009, China
| | - Yuying Xue
- Key Laboratory of Environmental Medicine and Engineering, Ministry of Education, School of Public Health & Collaborative Innovation Center of Suzhou Nano Science and Technology, Southeast University, Nanjing, 210009, China.,Jiangsu Key Laboratory for Biomaterials and Devices, Southeast University, Nanjing, 210009, China
| |
Collapse
|
63
|
Skowron KB, Shogan BD, Rubin DT, Hyman NH. The New Frontier: the Intestinal Microbiome and Surgery. J Gastrointest Surg 2018; 22:1277-1285. [PMID: 29633119 DOI: 10.1007/s11605-018-3744-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2018] [Accepted: 03/12/2018] [Indexed: 01/31/2023]
Abstract
The microbiome exerts a remarkable effect on human physiology. The study of the human-microbiome relationship is a burgeoning field with great potential to improve our understanding of health and disease. In this review, we address common surgical problems influenced by the human microbiome and explore what is thus far known about this relationship. These include inflammatory bowel disease, colorectal neoplasms, and diverticular disease. We will also discuss the effect of the microbiome on surgical complications, specifically anastomotic leak. We hope that further research in this field will enlighten our management of these and other surgical problems.
Collapse
Affiliation(s)
- Kinga B Skowron
- Department of Surgery, University of Chicago Medicine, 5841 S. Maryland Avenue, MC 5095, Chicago, IL, 60637, USA
| | - Benjamin D Shogan
- Department of Surgery, University of Chicago Medicine, 5841 S. Maryland Avenue, MC 5095, Chicago, IL, 60637, USA.
| | - David T Rubin
- Department of Medicine, Section of Gastroenterology, Hepatology and Nutrition, University of Chicago Medicine, Chicago, IL, USA
| | - Neil H Hyman
- Department of Surgery, University of Chicago Medicine, 5841 S. Maryland Avenue, MC 5095, Chicago, IL, 60637, USA
| |
Collapse
|
64
|
Hu J, Chen L, Tang Y, Xie C, Xu B, Shi M, Zheng W, Zhou S, Wang X, Liu L, Yan Y, Yang T, Niu Y, Hou Q, Xu X, Yan X. Standardized Preparation for Fecal Microbiota Transplantation in Pigs. Front Microbiol 2018; 9:1328. [PMID: 29971061 PMCID: PMC6018536 DOI: 10.3389/fmicb.2018.01328] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Accepted: 05/30/2018] [Indexed: 12/26/2022] Open
Abstract
The intestine of pigs harbors a mass of microorganisms which are essential for intestinal homeostasis and host health. Intestinal microbial disorders induce enteric inflammation and metabolic dysfunction, thereby causing adverse effects on the growth and health of pigs. In the human medicine, fecal microbiota transplantation (FMT), which engrafts the fecal microbiota from a healthy donor into a patient recipient, has shown efficacy in intestinal microbiota restoration. In addition, it has been used widely in therapy for human gastrointestinal diseases, including Clostridium difficile infection, inflammatory bowel diseases, and irritable bowel syndrome. Given that pigs share many similarities with humans, in terms of anatomy, nutritional physiology, and intestinal microbial compositions, FMT may also be used to restore the normal intestinal microbiota of pigs. However, feasible procedures for performing FMT in pigs remains unclear. Here, we summarize a standardized preparation for FMT in pigs by combining the standard methodology for human FMT with pig production. The key issues include the donor selection, fecal material preparation, fecal material transfer, stool bank establishment, and the safety for porcine FMT. Optimal donors should be selected to ensure the efficacy of porcine FMT and reduce the risks of transmitting infectious diseases to recipients during FMT. Preparing for fresh fecal material is highly recommended. Alternatively, frozen fecal suspension can also be prepared as an optimal choice because it is convenient and has similar efficacy. Oral administration of fecal suspension could be an optimal method for porcine fecal material transfer. Furthermore, the dilution ratio of fecal materials and the frequency of fecal material transfer could be adjusted according to practical situations in the pig industry. To meet the potential large-scale requirement in the pig industry, it is important to establish a stool bank to make porcine FMT readily available. Future studies should also focus on providing more robust safety data on FMT to improve the safety and tolerability of the recipient pigs. This standardized preparation for porcine FMT can facilitate the development of microbial targeted therapies and improve the intestinal health of pigs.
Collapse
Affiliation(s)
- Jun Hu
- State Key Laboratory of Agricultural Microbiology, College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan, China.,The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China.,Hubei Provincial Engineering Laboratory for Pig Precision Feeding and Feed Safety Technology, Wuhan, China
| | - Lingli Chen
- State Key Laboratory of Agricultural Microbiology, College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan, China.,The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China.,Hubei Provincial Engineering Laboratory for Pig Precision Feeding and Feed Safety Technology, Wuhan, China
| | - Yimei Tang
- State Key Laboratory of Agricultural Microbiology, College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan, China.,The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China.,Hubei Provincial Engineering Laboratory for Pig Precision Feeding and Feed Safety Technology, Wuhan, China
| | - Chunlin Xie
- State Key Laboratory of Agricultural Microbiology, College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan, China.,The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China.,Hubei Provincial Engineering Laboratory for Pig Precision Feeding and Feed Safety Technology, Wuhan, China
| | - Baoyang Xu
- State Key Laboratory of Agricultural Microbiology, College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan, China.,The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China.,Hubei Provincial Engineering Laboratory for Pig Precision Feeding and Feed Safety Technology, Wuhan, China
| | - Min Shi
- State Key Laboratory of Agricultural Microbiology, College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan, China.,The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China.,Hubei Provincial Engineering Laboratory for Pig Precision Feeding and Feed Safety Technology, Wuhan, China
| | - Wenyong Zheng
- State Key Laboratory of Agricultural Microbiology, College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan, China.,The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China.,Hubei Provincial Engineering Laboratory for Pig Precision Feeding and Feed Safety Technology, Wuhan, China
| | - Shuyi Zhou
- State Key Laboratory of Agricultural Microbiology, College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan, China.,The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China.,Hubei Provincial Engineering Laboratory for Pig Precision Feeding and Feed Safety Technology, Wuhan, China
| | - Xinkai Wang
- State Key Laboratory of Agricultural Microbiology, College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan, China.,The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China.,Hubei Provincial Engineering Laboratory for Pig Precision Feeding and Feed Safety Technology, Wuhan, China
| | - Liu Liu
- State Key Laboratory of Agricultural Microbiology, College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan, China.,The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China.,Hubei Provincial Engineering Laboratory for Pig Precision Feeding and Feed Safety Technology, Wuhan, China
| | - Yiqin Yan
- State Key Laboratory of Agricultural Microbiology, College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan, China.,The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China.,Hubei Provincial Engineering Laboratory for Pig Precision Feeding and Feed Safety Technology, Wuhan, China
| | - Tao Yang
- State Key Laboratory of Agricultural Microbiology, College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan, China.,The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China.,Hubei Provincial Engineering Laboratory for Pig Precision Feeding and Feed Safety Technology, Wuhan, China
| | - Yaorong Niu
- State Key Laboratory of Agricultural Microbiology, College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan, China.,The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China.,Hubei Provincial Engineering Laboratory for Pig Precision Feeding and Feed Safety Technology, Wuhan, China
| | - Qiliang Hou
- State Key Laboratory of Agricultural Microbiology, College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan, China.,The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China.,Hubei Provincial Engineering Laboratory for Pig Precision Feeding and Feed Safety Technology, Wuhan, China
| | - Xiaofan Xu
- State Key Laboratory of Agricultural Microbiology, College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan, China.,The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China.,Hubei Provincial Engineering Laboratory for Pig Precision Feeding and Feed Safety Technology, Wuhan, China
| | - Xianghua Yan
- State Key Laboratory of Agricultural Microbiology, College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan, China.,The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China.,Hubei Provincial Engineering Laboratory for Pig Precision Feeding and Feed Safety Technology, Wuhan, China
| |
Collapse
|
65
|
D'Odorico I, Di Bella S, Monticelli J, Giacobbe DR, Boldock E, Luzzati R. Role of fecal microbiota transplantation in inflammatory bowel disease. J Dig Dis 2018; 19:322-334. [PMID: 29696802 DOI: 10.1111/1751-2980.12603] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Revised: 01/09/2018] [Accepted: 04/22/2018] [Indexed: 12/11/2022]
Abstract
There is increasing evidence of the key role played by altered intestinal microbiota in the pathogenesis of inflammatory bowel disease (IBD). Management strategies involving immune modulation are effective and widely used, but treatment failures and side effects occur. Fecal microbiota transplantation (FMT) provides a novel, perhaps complementary, strategy to restore the normal gut microbiota in patients with IBD. This review summarizes the available efficacy and safety data on the use of FMT in patients with IBD. Several aspects remain to be clarified about the clinical predictors of the response to FMT, its most appropriate route of administration, and the most appropriate quantity and quality of microbiota to be transplanted. Further studies focusing on long-term outcomes and safety are also warranted.
Collapse
Affiliation(s)
| | - Stefano Di Bella
- Infectious Diseases Division, University Hospital of Trieste, Trieste, Italy
| | - Jacopo Monticelli
- Infectious Diseases Division, University Hospital of Trieste, Trieste, Italy
| | - Daniele R Giacobbe
- Infectious Diseases Unit, OspedalePoliclinico San Martino-IRCCS per l'Oncologia and University of Genoa, Genoa, Italy
| | - Emma Boldock
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK
| | - Roberto Luzzati
- Infectious Diseases Division, University Hospital of Trieste, Trieste, Italy
| |
Collapse
|
66
|
Taylor JM, Clarke EL, Baker K, Lauder A, Kim D, Bailey A, Wu GD, Collman RG, Doyle-Meyers L, Russell-Lodrigue K, Blanchard J, Bushman FD, Bohm R. Evaluation of a therapy for Idiopathic Chronic Enterocolitis in rhesus macaques ( Macaca mulatta) and linked microbial community correlates. PeerJ 2018; 6:e4612. [PMID: 29666764 PMCID: PMC5899420 DOI: 10.7717/peerj.4612] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2017] [Accepted: 03/22/2018] [Indexed: 01/01/2023] Open
Abstract
Idiopathic chronic enterocolitis (ICE) is one of the most commonly encountered and difficult to manage diseases of captive rhesus macaques (Macaca mulatta). The etiology is not well understood, but perturbations in gut microbial communities have been implicated. Here we evaluated the effects of a 14-day course of vancomycin, neomycin, and fluconazole on animals affected with ICE, comparing treated, untreated, and healthy animals. We performed microbiome analysis on duodenal and colonic mucosal samples and feces in order to probe bacterial and/or fungal taxa potentially associated with ICE. All treated animals showed a significant and long-lasting improvement in stool consistency over time when compared to untreated and healthy controls. Microbiome analysis revealed trends associating bacterial community composition with ICE, particularly lineages of the Lactobacillaceae family. Sequencing of DNA from macaque food biscuits revealed that fungal sequences recovered from stool were dominated by yeast-derived food additives; in contrast, bacteria in stool appeared to be authentic gut residents. In conclusion, while validation in larger cohorts is needed, the treatment described here was associated with significantly improved clinical signs; results suggested possible correlates of microbiome structure with disease, though no strong associations were detected between single microbes and ICE.
Collapse
Affiliation(s)
- Joshua M Taylor
- Division of Veterinary Medicine, Tulane National Primate Research Center, Covington, LA, United States of America
| | - Erik L Clarke
- Department of Microbiology, University of Pennsylvania School of Medicine, Philadelphia, PA, United States of America
| | - Kate Baker
- Division of Veterinary Medicine, Tulane National Primate Research Center, Covington, LA, United States of America
| | - Abigail Lauder
- Department of Microbiology, University of Pennsylvania School of Medicine, Philadelphia, PA, United States of America
| | - Dorothy Kim
- Division of Gastroenterology, Hepatology, and Nutrition, The Children's Hospital of Philadelphia, University of Pennsylvania School of Medicine, Philadelphia, PA, United States of America
| | - Aubrey Bailey
- Department of Microbiology, University of Pennsylvania School of Medicine, Philadelphia, PA, United States of America.,Department of Microbiology and Immunology, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
| | - Gary D Wu
- Division of Gastroenterology, Hepatology, and Nutrition, The Children's Hospital of Philadelphia, University of Pennsylvania School of Medicine, Philadelphia, PA, United States of America
| | - Ronald G Collman
- Division of Pulmonary, Allergy and Critical Care, Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA, United States of America
| | - Lara Doyle-Meyers
- Division of Veterinary Medicine, Tulane National Primate Research Center, Covington, LA, United States of America
| | - Kasi Russell-Lodrigue
- Division of Veterinary Medicine, Tulane National Primate Research Center, Covington, LA, United States of America
| | - James Blanchard
- Division of Veterinary Medicine, Tulane National Primate Research Center, Covington, LA, United States of America
| | - Frederic D Bushman
- Department of Microbiology, University of Pennsylvania School of Medicine, Philadelphia, PA, United States of America
| | - Rudolf Bohm
- Division of Veterinary Medicine, Tulane National Primate Research Center, Covington, LA, United States of America
| |
Collapse
|
67
|
Heath RD, Cockerell C, Mankoo R, Ibdah JA, Tahan V. Fecal microbiota transplantation and its potential therapeutic uses in gastrointestinal disorders. North Clin Istanb 2018; 5:79-88. [PMID: 29607440 PMCID: PMC5864716 DOI: 10.14744/nci.2017.10692] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Accepted: 11/09/2017] [Indexed: 02/08/2023] Open
Abstract
Typical human gut flora has been well characterized in previous studies and has been noted to have significant differences when compared with the typical microbiome of various disease states involving the gastrointestinal tract. Such diseases include Clostridium difficile colitis, inflammatory bowel disease, functional bowel syndromes, and various states of liver disease. A growing number of studies have investigated the use of a fecal microbiota transplant as a potential therapy for these disease states.
Collapse
Affiliation(s)
- Ryan D. Heath
- Division of Gastroenterology and Hepatology, University of Missouri, Columbia, Missouri, USA
| | - Courtney Cockerell
- Division of Gastroenterology and Hepatology, University of Missouri, Columbia, Missouri, USA
| | - Ravinder Mankoo
- Department of Internal Medicine, University of Missouri-Columbia 1 Hospital Drive, Columbia, Missouri, USA
| | - Jamal A. Ibdah
- Division of Gastroenterology and Hepatology, University of Missouri, Columbia, Missouri, USA
| | - Veysel Tahan
- Division of Gastroenterology and Hepatology, University of Missouri, Columbia, Missouri, USA
| |
Collapse
|
68
|
Obesogenic diet-induced gut barrier dysfunction and pathobiont expansion aggravate experimental colitis. PLoS One 2017; 12:e0187515. [PMID: 29107964 PMCID: PMC5673181 DOI: 10.1371/journal.pone.0187515] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Accepted: 10/20/2017] [Indexed: 01/01/2023] Open
Abstract
Consumption of a typical Western diet is a risk factor for several disorders. Metabolic syndrome is the most common disease associated with intake of excess fat. However, the incidence of inflammatory bowel disease is also greater in subjects consuming a Western diet, although the mechanism of this phenomenon is not clearly understood. We examined the morphological and functional changes of the intestine, the first site contacting dietary fat, in mice fed a high-fat diet (HFD) inducing obesity. Paneth cell area and production of antimicrobial peptides by Paneth cells were decreased in HFD-fed mice. Goblet cell number and secretion of mucin by goblet cells were also decreased, while intestinal permeability was increased in HFD-fed mice. HFD-fed mice were more susceptible to experimental colitis, and exhibited severe colonic inflammation, accompanied by the expansion of selected pathobionts such as Atopobium sp. and Proteobacteria. Fecal microbiota transplantation transferred the susceptibility to DSS-colitis, and antibiotic treatment abrogated colitis progression. These data suggest that an experimental HFD-induced Paneth cell dysfunction and subsequent intestinal dysbiosis characterized by pathobiont expansion can be predisposing factors to the development of inflammatory bowel disease.
Collapse
|
69
|
Khajah MA. The potential role of fecal microbiota transplantation in the treatment of inflammatory Bowel disease. Scand J Gastroenterol 2017; 52:1172-1184. [PMID: 28685630 DOI: 10.1080/00365521.2017.1347812] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Inflammatory bowel disease (IBD) is a chronic inflammatory disorder of an unknown etiology. Its pathogenesis involves an interplay of infectious, genetic, environmental, and immunological factors. The current therapeutic options have various limitations in terms of cost, side effect profile, and the development of drug resistance and dependence. Therefore, there is a need to develop future therapeutic options which are safe and effective to control the inflammatory process. This review focuses in a method for the administration of fecal matters (which contains a mixture of various commensals) from a healthy donor to the inflamed colon called fecal microbiota transplantation (FMT) aiming to correct the underlying dysbiosis in the gut as one of the major driving force for the inflammatory process. IBD patients have reduced number of protective (e.g., clostridia and bacteroids) and increased number of pathogenic (e.g., adhesive invasive E. coli and mycobacterium avium paratuberculosis) commensals, and this method is aimed to shift these changes in the gut. Recent studies from animal models and clinical trials suggest promising effects of this method in treating patients with IBD, but more studies are urgently needed to confirm its efficacy and safety, since the etiology of this chronic inflammatory disease is not fully understood and caution should be taken when transplanting fecal matters between individuals which might transfer other infectious organisms and diseases.
Collapse
Affiliation(s)
- Maitham Abbas Khajah
- a Pharmacology & Therapeutics, Faculty of Pharmacy , Kuwait University , Kuwait , Kuwait
| |
Collapse
|
70
|
Estrogen–gut microbiome axis: Physiological and clinical implications. Maturitas 2017; 103:45-53. [DOI: 10.1016/j.maturitas.2017.06.025] [Citation(s) in RCA: 301] [Impact Index Per Article: 37.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Revised: 06/20/2017] [Accepted: 06/22/2017] [Indexed: 12/16/2022]
|
71
|
Staley C, Kaiser T, Beura LK, Hamilton MJ, Weingarden AR, Bobr A, Kang J, Masopust D, Sadowsky MJ, Khoruts A. Stable engraftment of human microbiota into mice with a single oral gavage following antibiotic conditioning. MICROBIOME 2017; 5:87. [PMID: 28760163 PMCID: PMC5537947 DOI: 10.1186/s40168-017-0306-2] [Citation(s) in RCA: 132] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Accepted: 07/12/2017] [Indexed: 05/03/2023]
Abstract
BACKGROUND Human microbiota-associated (HMA) animal models relying on germ-free recipient mice are being used to study the relationship between intestinal microbiota and human disease. However, transfer of microbiota into germ-free animals also triggers global developmental changes in the recipient intestine, which can mask disease-specific attributes of the donor material. Therefore, a simple model of replacing microbiota into a developmentally mature intestinal environment remains highly desirable. RESULTS Here we report on the development of a sequential, three-course antibiotic conditioning regimen that allows sustained engraftment of intestinal microorganisms following a single oral gavage with human donor microbiota. SourceTracker, a Bayesian, OTU-based algorithm, indicated that 59.3 ± 3.0% of the fecal bacterial communities in treated mice were attributable to the donor source. This overall degree of microbiota engraftment was similar in mice conditioned with antibiotics and germ-free mice. Limited surveys of systemic and mucosal immune sites did not show evidence of immune activation following introduction of human microbiota. CONCLUSIONS The antibiotic treatment protocol described here followed by a single gavage of human microbiota may provide a useful, complimentary HMA model to that established in germ-free facilities. The model has the potential for further in-depth translational investigations of microbiota in a variety of human disease states.
Collapse
Affiliation(s)
| | - Thomas Kaiser
- BioTechnology Institute, University of Minnesota, St. Paul, MN, USA
| | - Lalit K Beura
- Center for Immunology, University of Minnesota, 2101 6th St. S.E., Room 3-184, Wallin Medical Biosciences Building, Minneapolis, MN, 55414, USA
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN, USA
| | | | | | - Aleh Bobr
- Center for Immunology, University of Minnesota, 2101 6th St. S.E., Room 3-184, Wallin Medical Biosciences Building, Minneapolis, MN, 55414, USA
- Division of Gastroenterology, Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Johnthomas Kang
- Center for Immunology, University of Minnesota, 2101 6th St. S.E., Room 3-184, Wallin Medical Biosciences Building, Minneapolis, MN, 55414, USA
- Division of Gastroenterology, Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - David Masopust
- Center for Immunology, University of Minnesota, 2101 6th St. S.E., Room 3-184, Wallin Medical Biosciences Building, Minneapolis, MN, 55414, USA
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN, USA
| | - Michael J Sadowsky
- BioTechnology Institute, University of Minnesota, St. Paul, MN, USA
- Department of Soil, Water, and Climate, University of Minnesota, St. Paul, MN, USA
| | - Alexander Khoruts
- BioTechnology Institute, University of Minnesota, St. Paul, MN, USA.
- Center for Immunology, University of Minnesota, 2101 6th St. S.E., Room 3-184, Wallin Medical Biosciences Building, Minneapolis, MN, 55414, USA.
- Division of Gastroenterology, Department of Medicine, University of Minnesota, Minneapolis, MN, USA.
| |
Collapse
|
72
|
Lan N, Ashburn J, Shen B. Fecal microbiota transplantation for Clostridium difficile infection in patients with ileal pouches. Gastroenterol Rep (Oxf) 2017; 5:200-207. [PMID: 28852524 PMCID: PMC5554389 DOI: 10.1093/gastro/gox018] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2016] [Revised: 02/02/2017] [Accepted: 02/20/2017] [Indexed: 12/30/2022] Open
Abstract
Background:Clostridium difficile infection (CDI) in patients with ileal pouch-anal anastomosis (IPAA) has been increasingly recognized. The aim of this study was to evaluate the outcome of fecal microbiota transplantation (FMT) in patients with pouch and CDI. Methods: All consecutive patients that underwent FMT for CDI from 2012 to 2016 were extracted from our IRB-approved, prospectively maintained Registry of Pouch Disorders. The primary outcome was negative stool tests for Clostridium difficile after FMT and the secondary outcomes were symptomatic and endoscopic responses. Results: A total of 13 patients were included in this study, with 10 being Caucasian males (76.9%). All patients had underlying ulcerative colitis for J pouch surgery. After a mean of 2.8±0.8 courses of antibiotic treatments was given and failed, 22 sessions of FMT were administered with an average of 1.7±1.1 sessions each. Within the 22 sessions, 16 were given via pouchoscopy, 4 via esophagogastroduodenoscopy and 2 via enemas. All patients tested negative on C. difficile polymerase chain reaction (PCR) after the initial FMT with a total of 7/12 (58.3%) documented patients showed symptomatic improvements and 3/11 (27.3%) patients showed endoscopic improvement according to the modified Pouchitis Disease Activity Index. During the follow-up of 1.2±1.1 years, there were a total of five patients (38.5%) that had recurrence after the successful initial treatment and four of them were successfully treated again with FMT. Conclusions: FMT appeared to be effective in eradication of CDI in patients with ileal pouches. However, FMT had a modest impact on endoscopic inflammation and recurrence after FMT and recurrence was common.
Collapse
Affiliation(s)
- Nan Lan
- Center for Inflammatory Bowel Diseases, Digestive Disease Institute, The Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Jean Ashburn
- Center for Inflammatory Bowel Diseases, Digestive Disease Institute, The Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Bo Shen
- Center for Inflammatory Bowel Diseases, Digestive Disease Institute, The Cleveland Clinic Foundation, Cleveland, OH, USA
- Corresponding author. The Interventional Inflammatory Bowel Disease (i-IBD) Unit, Digestive Disease and Surgery Institute-A31, The Cleveland Clinic Foundation, 9500 Euclid Ave, Cleveland, OH 44195, USA. Tel: +1–216–444–9252; Fax: +1–216–444–6305;
| |
Collapse
|
73
|
Stedtfeld RD, Stedtfeld TM, Fader KA, Williams MR, Bhaduri P, Quensen J, Zacharewski TR, Tiedje JM, Hashsham SA. TCDD influences reservoir of antibiotic resistance genes in murine gut microbiome. FEMS Microbiol Ecol 2017; 93:3798199. [PMID: 28475713 PMCID: PMC5458050 DOI: 10.1093/femsec/fix058] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2016] [Accepted: 05/02/2017] [Indexed: 02/06/2023] Open
Abstract
Dysbiosis of the gut microbiome via antibiotics, changes in diet and infection can select for bacterial groups that more frequently harbor antimicrobial resistance genes (ARGs) and mobile genetic elements (MGEs). However, the impact of environmental toxicants on the reservoir of ARGs in the gut microbiome has received less attention. 2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD) is a potent aryl hydrocarbon receptor (AhR) agonist with multiple toxic health effects including immune dysfunction. The selective pressure of TCDD on the abundance of ARG and MGE-harboring gut populations was examined using C57BL/6 mice exposed to 0-30 μg/kg TCDD for 28 and 92 days with the latter having a 30-day recovery period. DNA extracted from temporally collected fecal pellets was characterized using a qPCR array with 384 assays targeting ARGs and MGEs. Fourteen genes, typically observed in Enterobacteriaceae, increased significantly within 8 days of initial dosing, persisted throughout the treatment period, and remained induced 30 days post dosing. A qPCR primer set targeting Enterobacteriaceae also showed 10- to 100-fold higher abundance in TCDD-treated groups, which was further verified using metagenomics. Results show a bloom of ARG-harboring bacterial groups in the gut due to a xenobiotic compound that is not a metal, biocide or antimicrobial.
Collapse
Affiliation(s)
- Robert D. Stedtfeld
- Department of Civil and Environmental Engineering, East Lansing, MI 48824, USA
| | | | - Kelly A. Fader
- Institute for Integrative Toxicology, Michigan State University, East Lansing, MI 48824, USA
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI 48824, USA
| | - Maggie R. Williams
- Department of Civil and Environmental Engineering, East Lansing, MI 48824, USA
| | - Prianca Bhaduri
- Department of Civil and Environmental Engineering, East Lansing, MI 48824, USA
| | - John Quensen
- Center for Microbial Ecology, Michigan State University, East Lansing, MI 48824, USA
| | - Timothy R. Zacharewski
- Institute for Integrative Toxicology, Michigan State University, East Lansing, MI 48824, USA
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI 48824, USA
| | - James M. Tiedje
- Center for Microbial Ecology, Michigan State University, East Lansing, MI 48824, USA
| | - Syed A. Hashsham
- Department of Civil and Environmental Engineering, East Lansing, MI 48824, USA
- Center for Microbial Ecology, Michigan State University, East Lansing, MI 48824, USA
| |
Collapse
|
74
|
Advanced acuity in microbial biofilm genesis, development, associated clinical infections and control. ACTA ACUST UNITED AC 2017. [DOI: 10.1016/j.antinf.2017.01.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
|
75
|
Abstract
Fecal microbiota transplantation (FMT) is the transfer of stool from a healthy donor into the colon of a patient whose disease is a result of an altered microbiome, with the goal of restoring the normal microbiota and thus curing the disease. The most effective and well-studied indication for FMT is recurrent Clostridium difficile infection. At this time, there is insufficient evidence to recommend FMT for other gastrointestinal diseases, but studies are under way. There is also insufficient evidence to recommend FMT for nongastrointestinal diseases at this time. The field is rapidly emerging.
Collapse
Affiliation(s)
- Stephen M Vindigni
- Division of Gastroenterology, Department of Medicine, School of Medicine, University of Washington, 1959 Northeast Pacific Street, Box 356424, Seattle, WA 98195-6424, USA.
| | - Christina M Surawicz
- Division of Gastroenterology, Department of Medicine, University of Washington, 325 9th Avenue, Box 359773, Seattle, WA 98104, USA
| |
Collapse
|
76
|
Weingarden AR, Vaughn BP. Intestinal microbiota, fecal microbiota transplantation, and inflammatory bowel disease. Gut Microbes 2017; 8:238-252. [PMID: 28609251 PMCID: PMC5479396 DOI: 10.1080/19490976.2017.1290757] [Citation(s) in RCA: 322] [Impact Index Per Article: 40.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Inflammatory bowel disease (IBD) is a complex set of diseases that lead to chronic inflammation in the gastrointestinal tract. Although the etiology of IBD is not fully understood, it is well-known that the intestinal microbiota is associated with the development and maintenance of IBD. Manipulation of the gut microbiota, therefore, may represent a target for IBD therapy. Fecal microbiota transplantation (FMT), where fecal microbiota from a healthy donor is transplanted into a patient's GI tract, is already a successful therapy for Clostridium difficile infection. FMT is currently being explored as a potential therapy for IBD as well. In this review, the associations between the gut microbiota and IBD and the emerging data on FMT for IBD will be discussed.
Collapse
Affiliation(s)
- Alexa R. Weingarden
- Division of Gastroenterology, Hepatology and Nutrition, University of Minnesota, Minneapolis, MN, USA
| | - Byron P. Vaughn
- Division of Gastroenterology, Hepatology and Nutrition, University of Minnesota, Minneapolis, MN, USA,CONTACT Byron P. Vaughn 420 Delaware street SE, MMC36, Minneapolis, MN 55455
| |
Collapse
|
77
|
Corfield A. Eukaryotic protein glycosylation: a primer for histochemists and cell biologists. Histochem Cell Biol 2017; 147:119-147. [PMID: 28012131 PMCID: PMC5306191 DOI: 10.1007/s00418-016-1526-4] [Citation(s) in RCA: 90] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/25/2016] [Indexed: 12/21/2022]
Abstract
Proteins undergo co- and posttranslational modifications, and their glycosylation is the most frequent and structurally variegated type. Histochemically, the detection of glycan presence has first been performed by stains. The availability of carbohydrate-specific tools (lectins, monoclonal antibodies) has revolutionized glycophenotyping, allowing monitoring of distinct structures. The different types of protein glycosylation in Eukaryotes are described. Following this educational survey, examples where known biological function is related to the glycan structures carried by proteins are given. In particular, mucins and their glycosylation patterns are considered as instructive proof-of-principle case. The tissue and cellular location of glycoprotein biosynthesis and metabolism is reviewed, with attention to new findings in goblet cells. Finally, protein glycosylation in disease is documented, with selected examples, where aberrant glycan expression impacts on normal function to let disease pathology become manifest. The histological applications adopted in these studies are emphasized throughout the text.
Collapse
Affiliation(s)
- Anthony Corfield
- Mucin Research Group, School of Clinical Sciences, Bristol Royal Infirmary, University of Bristol, Bristol, BS2 8HW, UK.
| |
Collapse
|
78
|
Berlec A, Perše M, Ravnikar M, Lunder M, Erman A, Cerar A, Štrukelj B. Dextran sulphate sodium colitis in C57BL/6J mice is alleviated by Lactococcus lactis and worsened by the neutralization of Tumor necrosis Factor α. Int Immunopharmacol 2017; 43:219-226. [PMID: 28039805 DOI: 10.1016/j.intimp.2016.12.027] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Revised: 12/08/2016] [Accepted: 12/21/2016] [Indexed: 02/07/2023]
Abstract
TNFα has a well-established role in inflammatory bowel disease that affects the gastrointestinal tract and is usually manifested as Crohn's disease or ulcerative colitis. We have compared Lactococcus lactis NZ9000 displaying TNFα-binding affibody with control Lactococcus lactis and with anti-TNFα antibody infliximab for the treatment of mice with dextran sulphate sodium (DSS)-induced colitis. L. lactis NZ9000 alleviated the colitis severity one week after colitis induction with DSS, more effectively when administered in preventive fashion prior to, during and after DSS administration. TNFα-binding L. lactis was less effective than control L. lactis, particularly when TNFα-binding L. lactis was administered in preventive fashion. Similarly, an apparently detrimental effect of TNFα neutralization was observed in mice that were intraperitoneally administered anti-TNFα monoclonal antibody infliximab prior to colitis induction. The highest concentrations of tissue TNFα were observed in groups without DSS colitis that were treated either with TNFα-binding L. lactis or infliximab. To conclude, we have confirmed that L. lactis exerts a protective effect on DSS-induced colitis in mice. Contrary to expectations, but in line with some reports, the neutralization of TNFα aggravated disease symptoms in the acute phase of colitis and increased TNFα concentration in colon tissue of healthy mice. Nevertheless, we have demonstrated that oral administration of bacteria with surface displayed TNFα-binding affibody can interfere significantly with TNFα signaling and mimic the infliximab response in the given animal model of colitis.
Collapse
Affiliation(s)
- Aleš Berlec
- Department of Biotechnology, Jožef Stefan Institute, Jamova 39, SI-1000 Ljubljana, Slovenia.
| | - Martina Perše
- Institute of Pathology, Medical Experimental Centre, Faculty of Medicine, University of Ljubljana, Zaloška 4, 1000 Ljubljana, Slovenia
| | - Matjaž Ravnikar
- Faculty of Pharmacy, University of Ljubljana, Aškerčeva 7, 1000 Ljubljana, Slovenia
| | - Mojca Lunder
- Faculty of Pharmacy, University of Ljubljana, Aškerčeva 7, 1000 Ljubljana, Slovenia
| | - Andreja Erman
- Institute of Cell Biology, Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Anton Cerar
- Institute of Pathology, Medical Experimental Centre, Faculty of Medicine, University of Ljubljana, Zaloška 4, 1000 Ljubljana, Slovenia
| | - Borut Štrukelj
- Department of Biotechnology, Jožef Stefan Institute, Jamova 39, SI-1000 Ljubljana, Slovenia; Faculty of Pharmacy, University of Ljubljana, Aškerčeva 7, 1000 Ljubljana, Slovenia
| |
Collapse
|
79
|
Mizuno S, Nanki K, Matsuoka K, Saigusa K, Ono K, Arai M, Sugimoto S, Kiyohara H, Nakashima M, Takeshita K, Naganuma M, Suda W, Hattori M, Kanai T. Single fecal microbiota transplantation failed to change intestinal microbiota and had limited effectiveness against ulcerative colitis in Japanese patients. Intest Res 2017; 15:68-74. [PMID: 28239315 PMCID: PMC5323309 DOI: 10.5217/ir.2017.15.1.68] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Revised: 10/03/2016] [Accepted: 10/14/2016] [Indexed: 02/07/2023] Open
Abstract
Background/Aims Recent developments in analytical techniques including next-generation sequencing have clarified the correlation between intestinal microbiota and inflammatory bowel disease. Fecal microbiota transplantation (FMT) for patients with ulcerative colitis (UC) is proposed as a potential approach to resolving their dysbiosis; however, its safety and efficacy have not been confirmed. This single-arm, open-label, non-randomized study aimed to evaluate the safety and efficacy of FMT for Japanese patients with UC as the first registered clinical trial in Japan. Methods We enrolled 10 patients with active UC despite medical therapy. The donors were the patients' relatives and were carefully screened for infectious diseases. Fecal material was administered via colonoscopy, and the primary endpoint was the presence or absence of serious adverse events related to FMT. The secondary endpoint was a change in partial Mayo score at 12 weeks post-FMT. Scores ≤2 were considered a clinical response. Fecal samples were collected to follow changes in gut microbiota, while extracted complementary DNA were analyzed by a next-generation sequencer. We obtained written informed consent from all patients and donors. This study was approved by our Institutional Review Board and is registered in the University hospital Medical Information Network (UMIN) Clinical Trials Registry (UMIN 000012814). Results Five patients with moderate disease and five with severe disease were enrolled. No severe adverse effects were observed. One patient achieved clinical response; however, none of the patients' microbiota diversity recovered to the donor levels. Conclusions The use of single FMT for UC was safe; however, we failed to show its clinical efficacy and potential to change the intestinal microbiota.
Collapse
Affiliation(s)
- Shinta Mizuno
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Kosaku Nanki
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Katsuyoshi Matsuoka
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Keiichiro Saigusa
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Keiko Ono
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Mari Arai
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Shinya Sugimoto
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Hiroki Kiyohara
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Moeko Nakashima
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Kozue Takeshita
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan.; Department of Immunology, Keio University School of Medicine, Tokyo, Japan
| | - Makoto Naganuma
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Wataru Suda
- Department of Immunology, Keio University School of Medicine, Tokyo, Japan.; Laboratory of Metagenomics, Graduate School of Frontier Sciences, The University of Tokyo, Chiba, Japan
| | - Masahira Hattori
- Laboratory of Metagenomics, Graduate School of Frontier Sciences, The University of Tokyo, Chiba, Japan.; Graduate School of Advanced Science and Engineering, Waseda University, Tokyo, Japan
| | - Takanori Kanai
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
80
|
Zhang JZ, Bao CH, Shi Z, Weng ZJ, Wang XM, Wu LY, Wu HG. Role of intestinal microbiota in pathogenesis of inflammatory bowel disease. Shijie Huaren Xiaohua Zazhi 2016; 24:4505-4513. [DOI: 10.11569/wcjd.v24.i33.4505] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Inflammatory bowel disease (IBD) is characterized by chronic non-specific inflammation of the intestinal tract, and it easily relapses. The pathogenesis of IBD is not fully clear up to now. Intestinal microbiota has been confirmed to play an important role in the development and activation of the intestinal immune system. The changes of intestinal microbiota may induce or aggravate IBD, and regulating intestinal microbiota may have positive effects on the treatment of IBD. This article will discuss the relationship between intestinal microbiota and IBD.
Collapse
|
81
|
Lower Level of Bacteroides in the Gut Microbiota Is Associated with Inflammatory Bowel Disease: A Meta-Analysis. BIOMED RESEARCH INTERNATIONAL 2016; 2016:5828959. [PMID: 27999802 PMCID: PMC5143693 DOI: 10.1155/2016/5828959] [Citation(s) in RCA: 186] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/17/2016] [Accepted: 08/09/2016] [Indexed: 12/15/2022]
Abstract
Background and Aims. Multiple studies have reported associations between inflammatory bowel disease (IBD) and the flora disequilibrium of Bacteroides. We performed a meta-analysis of the available data to provide a more precise estimate of the association between Bacteroides level in the gut and IBD. Methods. We searched PubMed/MEDLINE, EMBASE, Cochrane Library, Wiley Library, BIOSIS previews, Web of Science, CNKI, and ScienceDirect databases for published literature on IBD and gut microbiota from 1990 to 2016. Quality of all eligible studies was assessed using the Newcastle-Ottawa Quality Assessment Scale (NOS). We compared the level of Bacteroides in IBD patients with that in a control group without IBD, different types of IBD patients, and IBD patients with active phase and in remission. Results. We identified 63 articles, 9 of which contained sufficient data for evaluation. The mean level of Bacteroides was significantly lower in Crohn's disease (CD) and ulcerative colitis (UC) patients in active phase than in normal controls. The level of Bacteroides in remission CD and UC patients was much lower than patients in the control group. Bacteroides level was even lower in patients with CD and UC in active phase than in remission. Conclusions. This analysis suggests that lower levels of Bacteroides are associated with IBD, especially in active phase.
Collapse
|
82
|
Vaughn BP, Vatanen T, Allegretti JR, Bai A, Xavier RJ, Korzenik J, Gevers D, Ting A, Robson SC, Moss AC. Increased Intestinal Microbial Diversity Following Fecal Microbiota Transplant for Active Crohn's Disease. Inflamm Bowel Dis 2016; 22:2182-90. [PMID: 27542133 PMCID: PMC4995064 DOI: 10.1097/mib.0000000000000893] [Citation(s) in RCA: 165] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
BACKGROUND The microbiota in the lumen of patients with Crohn's disease (CD) is characterized by reduced diversity, particularly Firmicutes and Bacteroidetes. It is unknown whether the introduction of the intestinal microbiota from healthy individuals could correct this dysbiosis and reverse mucosal inflammation. We investigated the response to fecal microbial transplantation (FMT) from healthy individuals to subjects with active CD. METHODS We performed a prospective open-label study (uncontrolled) of FMT from healthy donors to subjects with active CD. A single FMT was performed by colonoscopy. Recipients' microbial diversity, mucosal T-cell phenotypes, and clinical and inflammatory parameters were measured over 12 weeks, and safety over 26 weeks. RESULTS Nineteen subjects were treated with FMT and completed the study follow-up. Fifty-eight percent (11/19) demonstrated a clinical response (Harvey-Bradshaw Index decrease >3) following FMT. Fifteen subjects had sufficient pre/postfecal samples for analysis. A significant increase in microbial diversity occurred after FMT (P = 0.02). This was greater in clinical responders than nonresponders. Patients who experienced a clinical response demonstrated a significant shift in fecal microbial composition toward their donor's profile as assessed by the Bray-Curtis index at 4 weeks (P = 0.003). An increase in regulatory T cells (CD4CD25CD127lo) was also noted in recipients' lamina propria following FMT. No serious adverse events were noted over the 26-week study period. CONCLUSIONS In this open-label study, FMT led to an expansion in microbial bacterial diversity in patients with active CD. FMT was overall safe, although the clinical response was variable. Determining donor microbial factors that influence clinical response is needed before randomized clinical trials of FMT in CD.
Collapse
Affiliation(s)
- Byron P Vaughn
- Beth-Israel Deaconess Medical Center, Division of Gastroenterology, Inflammatory Bowel Disease Center, and Harvard Medical School, Boston, MA
| | - Tommi Vatanen
- Broad Institute of MIT and Harvard, Cambridge, MA
- Department of Computer Science, Aalto University School of Science, 02150 Espoo, Finland
| | - Jessica R Allegretti
- Brigham and Women’s Hospital, Division of Gastroenterology, Crohn’s and Colitis Center, and Harvard Medical School, Boston, MA
| | - Aiping Bai
- Beth-Israel Deaconess Medical Center, Division of Gastroenterology, Inflammatory Bowel Disease Center, and Harvard Medical School, Boston, MA
| | - Ramnik J Xavier
- Broad Institute of MIT and Harvard, Cambridge, MA
- Gastrointestinal Unit and Center for the Study of Inflammatory Bowel Disease and Center for Computational and Integrative Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | - Joshua Korzenik
- Brigham and Women’s Hospital, Division of Gastroenterology, Crohn’s and Colitis Center, and Harvard Medical School, Boston, MA
| | - Dirk Gevers
- Broad Institute of MIT and Harvard, Cambridge, MA
- Janssen Human Microbiome Institute, Janssen R&D, Cambridge, MA
| | - Amanda Ting
- Beth-Israel Deaconess Medical Center, Division of Gastroenterology, Inflammatory Bowel Disease Center, and Harvard Medical School, Boston, MA
| | - Simon C Robson
- Beth-Israel Deaconess Medical Center, Division of Gastroenterology, Inflammatory Bowel Disease Center, and Harvard Medical School, Boston, MA
| | - Alan C Moss
- Beth-Israel Deaconess Medical Center, Division of Gastroenterology, Inflammatory Bowel Disease Center, and Harvard Medical School, Boston, MA
| |
Collapse
|
83
|
Tian Z, Liu J, Liao M, Li W, Zou J, Han X, Kuang M, Shen W, Li H. Beneficial Effects of Fecal Microbiota Transplantation on Ulcerative Colitis in Mice. Dig Dis Sci 2016; 61:2262-2271. [PMID: 26846120 DOI: 10.1007/s10620-016-4060-2] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Accepted: 01/26/2016] [Indexed: 12/12/2022]
Abstract
BACKGROUND Ulcerative colitis (UC) is a chronic condition and the most common form of inflammatory bowel disease. The goal of standard treatment is mainly to induce and maintain remission with anti-inflammatory, immunosuppressive agents, and/or colectomy. Fecal microbiota transplantation (FMT) has been used successfully to treat relapsing or refractory Clostridium difficile infection. The alteration of microbiota in mouse models of UC as well as in patients suggested the possibility of treating UC with FMT. AIMS To study the effects of FMT on dextran sodium sulfate (DSS)-induced UC model in mice. METHODS Littermates of BALB/c and C57BL/6J were randomized into four groups: normal control , treatment with DSS for 7 days (DSS - FMT), treatment with DSS followed by FMT for another 8 days (DSS + FMT), and treatment with DSS and FMT followed by another 5 days for recovery (remission). Body weight, survival rate, and DAI scores of mice in each group were recorded. Changes in distal colon were studied by histopathology. Alterations of spleen and lamina propria regulatory lymphocytes, major bacterial species in feces and inflammatory cytokines in colon were also studied. RESULTS C57BL/6J mice experienced more significant weight loss than BALB/c mice after DSS treatment, regardless of whether the two strains of mice were co-housed or not. FMT caused reversal of DAI scores in BALB/c but not in C57BL/6J mice. In BALB/c mice, FMT also reduced colon inflammation that was paralleled by decreased inflammatory cytokine levels, altered bacterial microbiota, and regulatory lymphocyte proportions. CONCLUSIONS FMT is effective in a mouse model of UC through its modulation on gut microbiota and the host immune system.
Collapse
MESH Headings
- Animals
- B-Lymphocytes, Regulatory/immunology
- B-Lymphocytes, Regulatory/pathology
- CD4-Positive T-Lymphocytes
- Colitis, Ulcerative/chemically induced
- Colitis, Ulcerative/microbiology
- Colitis, Ulcerative/pathology
- Colitis, Ulcerative/therapy
- Colon/immunology
- Colon/microbiology
- Colon/pathology
- Cytokines/immunology
- DNA, Bacterial/analysis
- Dextran Sulfate/toxicity
- Disease Models, Animal
- Fecal Microbiota Transplantation/methods
- Feces/microbiology
- Female
- Gastrointestinal Microbiome/genetics
- Intestinal Mucosa/immunology
- Intestinal Mucosa/microbiology
- Intestinal Mucosa/pathology
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Spleen/immunology
- Spleen/pathology
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/pathology
Collapse
Affiliation(s)
- Zhihui Tian
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Jie Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Mengyu Liao
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Wenjuan Li
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Jiaqi Zou
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Xinxin Han
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Mingjie Kuang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Wanqiu Shen
- Department of Chemical Biology, School of Pharmacy, Tianjin Medical University, Tianjin, 300070, China
| | - Haidong Li
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China.
| |
Collapse
|
84
|
Brittnacher MJ, Heltshe SL, Hayden HS, Radey MC, Weiss EJ, Damman CJ, Zisman TL, Suskind DL, Miller SI. GUTSS: An Alignment-Free Sequence Comparison Method for Use in Human Intestinal Microbiome and Fecal Microbiota Transplantation Analysis. PLoS One 2016; 11:e0158897. [PMID: 27391011 PMCID: PMC4938407 DOI: 10.1371/journal.pone.0158897] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Accepted: 06/23/2016] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Comparative analysis of gut microbiomes in clinical studies of human diseases typically rely on identification and quantification of species or genes. In addition to exploring specific functional characteristics of the microbiome and potential significance of species diversity or expansion, microbiome similarity is also calculated to study change in response to therapies directed at altering the microbiome. Established ecological measures of similarity can be constructed from species abundances, however methods for calculating these commonly used ecological measures of similarity directly from whole genome shotgun (WGS) metagenomic sequence are lacking. RESULTS We present an alignment-free method for calculating similarity of WGS metagenomic sequences that is analogous to the Bray-Curtis index for species, implemented by the General Utility for Testing Sequence Similarity (GUTSS) software application. This method was applied to intestinal microbiomes of healthy young children to measure developmental changes toward an adult microbiome during the first 3 years of life. We also calculate similarity of donor and recipient microbiomes to measure establishment, or engraftment, of donor microbiota in fecal microbiota transplantation (FMT) studies focused on mild to moderate Crohn's disease. We show how a relative index of similarity to donor can be calculated as a measure of change in a patient's microbiome toward that of the donor in response to FMT. CONCLUSION Because clinical efficacy of the transplant procedure cannot be fully evaluated without analysis methods to quantify actual FMT engraftment, we developed a method for detecting change in the gut microbiome that is independent of species identification and database bias, sensitive to changes in relative abundance of the microbial constituents, and can be formulated as an index for correlating engraftment success with clinical measures of disease. More generally, this method may be applied to clinical evaluation of human microbiomes and provide potential diagnostic determination of individuals who may be candidates for specific therapies directed at alteration of the microbiome.
Collapse
Affiliation(s)
- Mitchell J. Brittnacher
- Department of Microbiology, University of Washington, Seattle, Washington, United States of America
| | - Sonya L. Heltshe
- Department of Pediatrics, University of Washington, Seattle, Washington, United States of America
- Seattle Children's Research Institute, Seattle, Washington, United States of America
| | - Hillary S. Hayden
- Department of Microbiology, University of Washington, Seattle, Washington, United States of America
| | - Matthew C. Radey
- Department of Microbiology, University of Washington, Seattle, Washington, United States of America
| | - Eli J. Weiss
- Department of Microbiology, University of Washington, Seattle, Washington, United States of America
| | - Christopher J. Damman
- Division of Gastroenterology, University of Washington, Seattle, Washington, United States of America
| | - Timothy L. Zisman
- Division of Gastroenterology, University of Washington, Seattle, Washington, United States of America
| | - David L. Suskind
- Department of Pediatrics, University of Washington, Seattle, Washington, United States of America
- Seattle Children’s Hospital, Seattle, Washington, United States of America
| | - Samuel I. Miller
- Department of Microbiology, University of Washington, Seattle, Washington, United States of America
- Department of Medicine, University of Washington, Seattle, Washington, United States of America
- Department of Immunology, University of Washington, Seattle, Washington, United States of America
- Department of Genome Sciences, University of Washington, Seattle, Washington, United States of America
| |
Collapse
|
85
|
Vindigni SM, Zisman TL, Suskind DL, Damman CJ. The intestinal microbiome, barrier function, and immune system in inflammatory bowel disease: a tripartite pathophysiological circuit with implications for new therapeutic directions. Therap Adv Gastroenterol 2016; 9:606-25. [PMID: 27366227 PMCID: PMC4913337 DOI: 10.1177/1756283x16644242] [Citation(s) in RCA: 140] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
We discuss the tripartite pathophysiological circuit of inflammatory bowel disease (IBD), involving the intestinal microbiota, barrier function, and immune system. Dysfunction in each of these physiological components (dysbiosis, leaky gut, and inflammation) contributes in a mutually interdependent manner to IBD onset and exacerbation. Genetic and environmental risk factors lead to disruption of gut homeostasis: genetic risks predominantly affect the immune system, environmental risks predominantly affect the microbiota, and both affect barrier function. Multiple genetic and environmental 'hits' are likely necessary to establish and exacerbate disease. Most conventional IBD therapies currently target only one component of the pathophysiological circuit, inflammation; however, many patients with IBD do not respond to immune-modulating therapies. Hope lies in new classes of therapies that target the microbiota and barrier function.
Collapse
Affiliation(s)
- Stephen M. Vindigni
- Division of Gastroenterology, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Timothy L. Zisman
- Division of Gastroenterology, Department of Medicine, University of Washington, Seattle, WA, USA
| | - David L. Suskind
- Department of Pediatrics, Seattle Children’s Hospital and University of Washington, Seattle, WA, USA
| | | |
Collapse
|
86
|
Shi Y, Dong Y, Huang W, Zhu D, Mao H, Su P. Fecal Microbiota Transplantation for Ulcerative Colitis: A Systematic Review and Meta-Analysis. PLoS One 2016; 11:e0157259. [PMID: 27295210 PMCID: PMC4905678 DOI: 10.1371/journal.pone.0157259] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Accepted: 05/26/2016] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Fecal microbiota transplantation (FMT) has been recognized as a novel treatment for ulcerative colitis (UC). However, its efficacy and safety remain unclear. OBJECTIVE We conducted this systematic review to assess the efficacy and safety of FMT in UC. DATA SOURCES PubMed, EMBASE, Cochrane Central, Web of Science Core Collection, and three other Chinese databases were searched for reports of FMT in UC with clear outcomes. DATA EXTRACTION AND SYNTHESIS We estimated pooled rates [with 95% confidence interval (CI)] of clinical remission among 15 cohort studies and clinical response among 16 cohort studies. RESULTS Twenty five studies (2 randomized controlled trials, 15 cohort studies, and 8 case studies) with 234 UC patients were included. Overall, 41.58% (84/202) patients achieved clinical remission (CR) and 65.28% (126/193) achieved clinical response. Among the cohort studies, the pooled estimate of patients who achieved CR and clinical response were 40.5% (95% CI 24.7%-58.7%), and 66.1% (95% CI 43.7%-83.0%). Most adverse events were slight and self-resolving. The analyses of gut microbiota in 7 studies showed that FMT could increase microbiota diversity and richness, similarity, and certain change of bacterial composition. CONCLUSION FMT provides a promising effect for UC with few adverse events. Successful FMT may be associated with an increase in microbiota diversity and richness, similarity, and certain change of bacterial composition.
Collapse
Affiliation(s)
- Yanqiang Shi
- The Second Clinical Medical School, Southern Medical University, 510280, Guangzhou City, Guangdong Province, China
| | - Yiwei Dong
- The Second Clinical Medical School, Southern Medical University, 510280, Guangzhou City, Guangdong Province, China
| | - Wenhui Huang
- The Second Clinical Medical School, Southern Medical University, 510280, Guangzhou City, Guangdong Province, China
| | - Decong Zhu
- The Second Clinical Medical School, Southern Medical University, 510280, Guangzhou City, Guangdong Province, China
| | - Hua Mao
- Department of Gastroenterology, Zhujiang Hospital, Southern Medical University, 510280, Guangzhou City, Guangdong Province, China
| | - Peizhu Su
- Department of Gastroenterology, Zhujiang Hospital, Southern Medical University, 510280, Guangzhou City, Guangdong Province, China
| |
Collapse
|
87
|
Peng Z, Xiang J, He Z, Zhang T, Xu L, Cui B, Li P, Huang G, Ji G, Nie Y, Wu K, Fan D, Zhang F. Colonic transendoscopic enteral tubing: A novel way of transplanting fecal microbiota. Endosc Int Open 2016; 4:E610-3. [PMID: 27556065 PMCID: PMC4993903 DOI: 10.1055/s-0042-105205] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Accepted: 02/26/2016] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND AND STUDY AIMS Placement of a tube through the anus into the cecum has not yet been established as a method of administering whole-colonic treatment. The aim of this study was to evaluate the safety, feasibility, and value of transendoscopic enteral tubing (TET) for fecal microbiota transplantation (FMT) through the colon. PATIENTS AND METHODS A prospective observational study was performed of FMT using a new colonic TET technique. Under endoscopic guidance, a TET tube was affixed to the cecum with clips. The safety, value, and satisfaction with the FMT by TET were evaluated. RESULTS A total of 54 patients underwent TET. The success rate of the TET procedure was 100 % (54/54). Duration of the TET procedures was 14.8 ± 5.8 min. During the TET tube retention period, 98.1 % (53/54) of patients were satisfied with TET. The retention time for whole-colon delivery of the fecal microbiota suspension was 12.4 ± 2.3 days. In 88.4 % (49/54) of cases, no discomfort was reported during injection through the TET tube of the microbiota suspension. No adverse events were see in patients who required tube extubation after FMT. CONCLUSIONS Colonic TET is a novel, safe, convenient, and reliable procedure for FMT that results in a high degree of patient satisfaction.
Collapse
Affiliation(s)
- Zhaoyuan Peng
- Medical Center for Digestive Diseases, the Second Affiliated Hospital of Nanjing Medical University, Nanjing 210011, Jiangsu Province, China.,Key Lab of Holistic Integrative Enterology, Nanjing Medical University, 121 Jiangjiayuan, Nanjing 210011, Jiangsu Province, China.
| | - Jie Xiang
- Medical Center for Digestive Diseases, the Second Affiliated Hospital of Nanjing Medical University, Nanjing 210011, Jiangsu Province, China.,Key Lab of Holistic Integrative Enterology, Nanjing Medical University, 121 Jiangjiayuan, Nanjing 210011, Jiangsu Province, China.
| | - Zhi He
- Medical Center for Digestive Diseases, the Second Affiliated Hospital of Nanjing Medical University, Nanjing 210011, Jiangsu Province, China.,Key Lab of Holistic Integrative Enterology, Nanjing Medical University, 121 Jiangjiayuan, Nanjing 210011, Jiangsu Province, China.
| | - Ting Zhang
- Medical Center for Digestive Diseases, the Second Affiliated Hospital of Nanjing Medical University, Nanjing 210011, Jiangsu Province, China.,Key Lab of Holistic Integrative Enterology, Nanjing Medical University, 121 Jiangjiayuan, Nanjing 210011, Jiangsu Province, China.
| | - Lijuan Xu
- Medical Center for Digestive Diseases, the Second Affiliated Hospital of Nanjing Medical University, Nanjing 210011, Jiangsu Province, China.,Key Lab of Holistic Integrative Enterology, Nanjing Medical University, 121 Jiangjiayuan, Nanjing 210011, Jiangsu Province, China.
| | - Bota Cui
- Medical Center for Digestive Diseases, the Second Affiliated Hospital of Nanjing Medical University, Nanjing 210011, Jiangsu Province, China.,Key Lab of Holistic Integrative Enterology, Nanjing Medical University, 121 Jiangjiayuan, Nanjing 210011, Jiangsu Province, China.
| | - Pan Li
- Medical Center for Digestive Diseases, the Second Affiliated Hospital of Nanjing Medical University, Nanjing 210011, Jiangsu Province, China.,Key Lab of Holistic Integrative Enterology, Nanjing Medical University, 121 Jiangjiayuan, Nanjing 210011, Jiangsu Province, China.
| | - Guangming Huang
- Medical Center for Digestive Diseases, the Second Affiliated Hospital of Nanjing Medical University, Nanjing 210011, Jiangsu Province, China.,Key Lab of Holistic Integrative Enterology, Nanjing Medical University, 121 Jiangjiayuan, Nanjing 210011, Jiangsu Province, China.
| | - Guozhong Ji
- Medical Center for Digestive Diseases, the Second Affiliated Hospital of Nanjing Medical University, Nanjing 210011, Jiangsu Province, China.,Key Lab of Holistic Integrative Enterology, Nanjing Medical University, 121 Jiangjiayuan, Nanjing 210011, Jiangsu Province, China.
| | - Yongzhan Nie
- State Key Laboratory of Cancer Biology & Xijing Hospital of Digestive Diseases, the Fourth Military Medical University, Xi’an 710032, China.
| | - Kaichun Wu
- State Key Laboratory of Cancer Biology & Xijing Hospital of Digestive Diseases, the Fourth Military Medical University, Xi’an 710032, China.
| | - Daiming Fan
- State Key Laboratory of Cancer Biology & Xijing Hospital of Digestive Diseases, the Fourth Military Medical University, Xi’an 710032, China.
| | - Faming Zhang
- Medical Center for Digestive Diseases, the Second Affiliated Hospital of Nanjing Medical University, Nanjing 210011, Jiangsu Province, China.,Key Lab of Holistic Integrative Enterology, Nanjing Medical University, 121 Jiangjiayuan, Nanjing 210011, Jiangsu Province, China.,Corresponding author Faming Zhang, MD, PhD Medical Center for Digestive DiseasesThe Second Affiliated Hospital of Nanjing Medical University121 Jiang JiayuanNanjing 210011China+86-25-58509883
| |
Collapse
|
88
|
Abstract
Inflammatory bowel disease is a complex, chronic, multifactorial inflammatory disorder of the digestive tract. Standard therapies include immunosuppressive and biological treatments, but there is increasing interest in the potential benefit of complementary and alternative medicine for the treatment of inflammatory bowel disease. Given the high prevalence of use of complementary and alternative medicine among inflammatory bowel disease patients, gastroenterologists must remain knowledgeable regarding the risks and benefits of these treatment options. This article reviews the updated scientific data on the use of biologically based complementary and alternative therapies for the treatment of inflammatory bowel disease.
Collapse
|
89
|
Abstract
Although genes contribute to colorectal cancer, the gut microbiota are an important player. Accumulating evidence suggests that chronic infection and the ensuing inflammation contributes to tumor initiation and tumor progression. A variety of bacterial species and tumor-promoting virulence mechanisms have been investigated. Significant advances have been made in understanding the composition and functional capabilities of the gut microbiota and its roles in cancer. In the current review, we discuss the novel roles of microbiota in the progression of colon cancer. Although microbiota technically include organisms other than bacteria e.g., viruses and fungi, this review will primarily focus on bacteria. We summarize epidemiological studies of human microbiome and colon cancer. We discuss the progress in the scientific understanding of the interplay between the gut microbiota, barrier function, and host responses in experimental models. Further, we discuss the potential application in prevention, diagnosis, and therapy of colon cancer by targeting microbiota. We discuss the challenges lie ahead and the future direction in studying gut microbiome in colon cancer to close the gap between the basic sciences and clinical application.
Collapse
Affiliation(s)
- Jun Sun
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Ikuko Kato
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI 48201, USA
- Department of Pathology, Wayne State University School of Medicine, Detroit, MI 48201, USA
| |
Collapse
|
90
|
Anti-infective activities of lactobacillus strains in the human intestinal microbiota: from probiotics to gastrointestinal anti-infectious biotherapeutic agents. Clin Microbiol Rev 2016; 27:167-99. [PMID: 24696432 DOI: 10.1128/cmr.00080-13] [Citation(s) in RCA: 228] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
A vast and diverse array of microbial species displaying great phylogenic, genomic, and metabolic diversity have colonized the gastrointestinal tract. Resident microbes play a beneficial role by regulating the intestinal immune system, stimulating the maturation of host tissues, and playing a variety of roles in nutrition and in host resistance to gastric and enteric bacterial pathogens. The mechanisms by which the resident microbial species combat gastrointestinal pathogens are complex and include competitive metabolic interactions and the production of antimicrobial molecules. The human intestinal microbiota is a source from which Lactobacillus probiotic strains have often been isolated. Only six probiotic Lactobacillus strains isolated from human intestinal microbiota, i.e., L. rhamnosus GG, L. casei Shirota YIT9029, L. casei DN-114 001, L. johnsonii NCC 533, L. acidophilus LB, and L. reuteri DSM 17938, have been well characterized with regard to their potential antimicrobial effects against the major gastric and enteric bacterial pathogens and rotavirus. In this review, we describe the current knowledge concerning the experimental antibacterial activities, including antibiotic-like and cell-regulating activities, and therapeutic effects demonstrated in well-conducted, placebo-controlled, randomized clinical trials of these probiotic Lactobacillus strains. What is known about the antimicrobial activities supported by the molecules secreted by such probiotic Lactobacillus strains suggests that they constitute a promising new source for the development of innovative anti-infectious agents that act luminally and intracellularly in the gastrointestinal tract.
Collapse
|
91
|
Abstract
Fungal biofilms have become an increasingly important clinical problem. The widespread use of antibiotics, frequent use of indwelling medical devices, and a trend toward increased patient immunosuppression have resulted in a creation of opportunity for clinically important yeasts and molds to form biofilms. This review will discuss the diversity and importance of fungal biofilms in the context of clinical medicine, provide novel insights into the clinical management of fungal biofilm infection, present evidence why these structures are recalcitrant to antifungal therapy, and discuss how our knowledge and understanding may lead to novel therapeutic intervention.
Collapse
|
92
|
Wen HZ, Hao WW, Liu YT, Shao LJ. Fecal microbiota transplantation for inflammatory bowel disease. Shijie Huaren Xiaohua Zazhi 2016; 24:1545-1551. [DOI: 10.11569/wcjd.v24.i10.1545] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Fecal microbiota transplantation (FMT) for treatment of inflammatory bowel disease (IBD), a common digestive disease, has attracted great interest in recent years, mainly because of the high prevalence and recurrence of IBD. IBD is difficult to treat, and standard therapy does not always induce remission. FMT is an alternative approach that can induce remission in some patients with active IBD. However, many unanswered questions on the clinical application of FMT in IBD remain. This review discusses the efficacy, adverse events, and optional route of administration of FMT in IBD.
Collapse
|
93
|
Fröhlich EE, Fröhlich E. Cytotoxicity of Nanoparticles Contained in Food on Intestinal Cells and the Gut Microbiota. Int J Mol Sci 2016; 17:509. [PMID: 27058534 PMCID: PMC4848965 DOI: 10.3390/ijms17040509] [Citation(s) in RCA: 133] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Revised: 03/28/2016] [Accepted: 03/31/2016] [Indexed: 01/15/2023] Open
Abstract
Toxicity of nanoparticles (NPs) upon oral exposure has been studied in animals using physiological changes, behavior, histology, and blood analysis for evaluation. The effects recorded include the combination of the action on cells of the exposed animal and the reaction of the microorganisms that populate the external and internal surfaces of the body. The importance of these microorganisms, collectively termed as microbiota, for the health of the host has been widely recognized. They may also influence toxicity of NPs but these effects are difficult to differentiate from toxicity on cells of the gastrointestinal tract. To estimate the likelihood of preferential damage of the microbiota by NPs the relative sensitivity of enterocytes and bacteria was compared. For this comparison NPs with antimicrobial action present in consumer products were chosen. The comparison of cytotoxicity with Escherichia coli as representative for intestinal bacteria and on gastrointestinal cells revealed that silver NPs damaged bacteria at lower concentrations than enterocytes, while the opposite was true for zinc oxide NPs. These results indicate that silver NPs may cause adverse effects by selectively affecting the gut microbiota. Fecal transplantation from NP-exposed animals to unexposed ones offers the possibility to verify this hypothesis.
Collapse
Affiliation(s)
- Esther E Fröhlich
- Institute of Experimental and Clinical Pharmacology, Medical University of Graz, Universitätsplatz 4, Graz A-8010, Austria.
| | - Eleonore Fröhlich
- Center for Medical Research, Medical University of Graz, Stiftingtalstr. 24, Graz A-8010, Austria.
| |
Collapse
|
94
|
Leszczyszyn JJ, Radomski M, Leszczyszyn AM. Intestinal microbiota transplant - current state of knowledge. Reumatologia 2016; 54:24-8. [PMID: 27407273 PMCID: PMC4847330 DOI: 10.5114/reum.2016.58758] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Accepted: 02/26/2016] [Indexed: 12/21/2022] Open
Abstract
Faecal microbiota transplantation (FMT) has induced a lot scientific interest and hopes for the last couple of years. FMT has been approved as a treatment of recurrent Clostridium difficile colitis. Highly sophisticated molecular DNA identification methods have been used to assess the healthy human microbiome as well as its disturbances in several diseases. The metabolic and immunologic functions of the microbiome have become more clear and understandable. A lot of pathological changes, such as production of short-chain fatty acids or components of the inflammatory cascade, caused by changes in microbiome diversity, variability and richness have been observed among patients suffering from inflammatory bowel diseases, irritable bowel syndrome, type 2 diabetes or rheumatoid arthritis. The published clinical results are encouraging, but still there is huge demand for FMT controlled clinical trials.
Collapse
Affiliation(s)
- Jarosław Jerzy Leszczyszyn
- National Institute of Geriatrics, Rheumatology and Rehabilitation, Warsaw, Poland
- Biotransplant Research, Wroclaw, Poland
| | - Marek Radomski
- National Institute of Geriatrics, Rheumatology and Rehabilitation, Warsaw, Poland
- School of Pharmacy and Pharmaceutical Sciences, University of Dublin, Trinity College, Dublin, Ireland
| | | |
Collapse
|
95
|
Trueba AF, Ritz T, Trueba G. The Role of the Microbiome in the Relationship of Asthma and Affective Disorders. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 874:263-88. [PMID: 26589224 DOI: 10.1007/978-3-319-20215-0_13] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The effect of stress, anxiety and other affective states on inflammatory conditions such as asthma is well documented. Although several immune pathway mechanisms have been proposed and studied, they cannot fully explain the relationship. In this chapter we present a new perspective on asthma development and exacerbation that integrates findings on the role of psychological factors in asthma with the microbiome and the hygiene hypothesis in asthma development.
Collapse
Affiliation(s)
- Ana F Trueba
- Universidad San Francisco de Quito, Quito, Ecuador.
| | - Thomas Ritz
- Southern Methodist University, Dallas, TX, USA.
| | | |
Collapse
|
96
|
Abstract
There has been increasing interest in understanding the role of the human gut microbiome to elucidate the therapeutic potential of its manipulation. Fecal microbiota transplantation (FMT) is the administration of a solution of fecal matter from a donor into the intestinal tract of a recipient in order to directly change the recipient's gut microbial composition and confer a health benefit. FMT has been used to successfully treat recurrent Clostridium difficile infection. There are preliminary indications to suggest that it may also carry therapeutic potential for other conditions such as inflammatory bowel disease, obesity, metabolic syndrome, and functional gastrointestinal disorders.
Collapse
Affiliation(s)
- Shaan Gupta
- Department of Medicine, Gastrointestinal Diseases Research Unit, Queen’s University, Kingston, ON, Canada
| | - Emma Allen-Vercoe
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, ON, Canada
| | | |
Collapse
|
97
|
Alfano M, Canducci F, Nebuloni M, Clementi M, Montorsi F, Salonia A. The interplay of extracellular matrix and microbiome in urothelial bladder cancer. Nat Rev Urol 2016; 13:77-90. [PMID: 26666363 PMCID: PMC7097604 DOI: 10.1038/nrurol.2015.292] [Citation(s) in RCA: 92] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Many pathological changes in solid tumours are caused by the accumulation of genetic mutations and epigenetic molecular alterations. In addition, tumour progression is profoundly influenced by the environment surrounding the transformed cells. The interplay between tumour cells and their microenvironment has been recognized as one of the key determinants of cancer development and is being extensively investigated. Data suggest that both the extracellular matrix and the microbiota represent microenvironments that contribute to the onset and progression of tumours. Through the introduction of omics technologies and pyrosequencing analyses, a detailed investigation of these two microenvironments is now possible. In urological research, assessment of their dysregulation has become increasingly important to provide diagnostic, prognostic and predictive biomarkers for urothelial bladder cancer. Understanding the roles of the extracellular matrix and microbiota, two key components of the urothelial mucosa, in the sequelae of pathogenic events that occur in the development and progression of urothelial carcinomas will be important to overcome the shortcomings in current bladder cancer treatment strategies.
Collapse
Affiliation(s)
- Massimo Alfano
- Division of Experimental Oncology/Unit of Urology, URI, IRCCS Ospedale San Raffaele, Via Olgettina 60, 20132 Milan Italy
| | - Filippo Canducci
- Department of Biotechnology and Life Sciences, Università degli Studi dell'Insubria, Via Dunant 3, 21100 Varese Italy
| | - Manuela Nebuloni
- Department of Clinical Sciences, Pathology Unit, L. Sacco Hospital, Università degli Studi di Milano, Via Giovanni Battista Grassi 74, 20157 Milan Italy
| | - Massimo Clementi
- Università Vita-Salute San Raffaele, Via Olgettina 60, 20132 Milan Italy
| | - Francesco Montorsi
- Università Vita-Salute San Raffaele, Via Olgettina 60, 20132 Milan Italy
| | - Andrea Salonia
- Università Vita-Salute San Raffaele, Via Olgettina 60, 20132 Milan Italy
| |
Collapse
|
98
|
Moloney RD, Johnson AC, O'Mahony SM, Dinan TG, Greenwood‐Van Meerveld B, Cryan JF. Stress and the Microbiota-Gut-Brain Axis in Visceral Pain: Relevance to Irritable Bowel Syndrome. CNS Neurosci Ther 2016; 22:102-17. [PMID: 26662472 PMCID: PMC6492884 DOI: 10.1111/cns.12490] [Citation(s) in RCA: 253] [Impact Index Per Article: 28.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Revised: 11/05/2015] [Accepted: 11/05/2015] [Indexed: 02/06/2023] Open
Abstract
Visceral pain is a global term used to describe pain originating from the internal organs of the body, which affects a significant proportion of the population and is a common feature of functional gastrointestinal disorders (FGIDs) such as irritable bowel syndrome (IBS). While IBS is multifactorial, with no single etiology to completely explain the disorder, many patients also experience comorbid behavioral disorders, such as anxiety or depression; thus, IBS is described as a disorder of the gut-brain axis. Stress is implicated in the development and exacerbation of visceral pain disorders. Chronic stress can modify central pain circuitry, as well as change motility and permeability throughout the gastrointestinal (GI) tract. More recently, the role of the gut microbiota in the bidirectional communication along the gut-brain axis, and subsequent changes in behavior, has emerged. Thus, stress and the gut microbiota can interact through complementary or opposing factors to influence visceral nociceptive behaviors. This review will highlight the evidence by which stress and the gut microbiota interact in the regulation of visceral nociception. We will focus on the influence of stress on the microbiota and the mechanisms by which microbiota can affect the stress response and behavioral outcomes with an emphasis on visceral pain.
Collapse
Affiliation(s)
- Rachel D. Moloney
- Laboratory of NeurogastroenterologyAPC Microbiome InstituteUniversity College CorkCorkIreland
- Present address:
Oklahoma Center for NeuroscienceUniversity of Oklahoma Health Science CenterOklahoma CityOKUSA
| | - Anthony C. Johnson
- Oklahoma Center for NeuroscienceUniversity of Oklahoma Health Science CenterOklahoma CityOKUSA
| | - Siobhain M. O'Mahony
- Laboratory of NeurogastroenterologyAPC Microbiome InstituteUniversity College CorkCorkIreland
- Department of Anatomy and NeuroscienceUniversity College CorkCorkIreland
| | - Timothy G. Dinan
- Laboratory of NeurogastroenterologyAPC Microbiome InstituteUniversity College CorkCorkIreland
- Department of Psychiatry and Neurobehavioural ScienceUniversity College CorkCorkIreland
| | - Beverley Greenwood‐Van Meerveld
- Oklahoma Center for NeuroscienceUniversity of Oklahoma Health Science CenterOklahoma CityOKUSA
- V.A. Medical CenterOklahoma CityOKUSA
| | - John F. Cryan
- Laboratory of NeurogastroenterologyAPC Microbiome InstituteUniversity College CorkCorkIreland
- Department of Anatomy and NeuroscienceUniversity College CorkCorkIreland
| |
Collapse
|
99
|
Meeker S, Seamons A, Maggio-Price L, Paik J. Protective links between vitamin D, inflammatory bowel disease and colon cancer. World J Gastroenterol 2016; 22:933-48. [PMID: 26811638 PMCID: PMC4716046 DOI: 10.3748/wjg.v22.i3.933] [Citation(s) in RCA: 94] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Revised: 09/28/2015] [Accepted: 11/24/2015] [Indexed: 02/06/2023] Open
Abstract
Vitamin D deficiency has been associated with a wide range of diseases and multiple forms of cancer including breast, colon, and prostate cancers. Relatively recent work has demonstrated vitamin D to be critical in immune function and therefore important in inflammatory diseases such as inflammatory bowel disease (IBD). Because vitamin D deficiency or insufficiency is increasingly prevalent around the world, with an estimated 30%-50% of children and adults at risk for vitamin D deficiency worldwide, it could have a significant impact on IBD. Epidemiologic studies suggest that low serum vitamin D levels are a risk factor for IBD and colon cancer, and vitamin D supplementation is associated with decreased colitis disease activity and/or alleviated symptoms. Patients diagnosed with IBD have a higher incidence of colorectal cancer than the general population, which supports the notion that inflammation plays a key role in cancer development and underscores the importance of understanding how vitamin D influences inflammation and its cancer-promoting effects. In addition to human epidemiological data, studies utilizing mouse models of colitis have shown that vitamin D is beneficial in preventing or ameliorating inflammation and clinical disease. The precise role of vitamin D on colitis is unknown; however, vitamin D regulates immune cell trafficking and differentiation, gut barrier function and antimicrobial peptide synthesis, all of which may be protective from IBD and colon cancer. Here we focus on effects of vitamin D on inflammation and inflammation-associated colon cancer and discuss the potential use of vitamin D for protection and treatment of IBD and colon cancer.
Collapse
|
100
|
Bowman KA, Broussard EK, Surawicz CM. Fecal microbiota transplantation: current clinical efficacy and future prospects. Clin Exp Gastroenterol 2015; 8:285-91. [PMID: 26566371 PMCID: PMC4627401 DOI: 10.2147/ceg.s61305] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Fecal microbiota transplantation (FMT) has gained mainstream attention with its remarkable efficacy in treating recurrent Clostridium difficile infection (RCDI) when there are no other effective therapies. Methods of selecting donors and routes of administration vary among studies, but there are now randomized controlled trials showing efficacy of FMT in treating RCDI. Ongoing trials of FMT for other disease such as inflammatory bowel disease are underway; this therapy should not be used for these conditions unless there is strong evidence for efficacy. Long-term safety data are sorely needed, as well as clarification of regulatory concerns.
Collapse
Affiliation(s)
- Kathryn A Bowman
- Department of Medicine, University of Washington School of Medicine, Seattle, WA, USA
| | - Elizabeth K Broussard
- Division of Gastroenterology, University of Washington School of Medicine, Seattle, WA, USA
| | - Christina M Surawicz
- Division of Gastroenterology, University of Washington School of Medicine, Seattle, WA, USA
| |
Collapse
|