401
|
Romick-Rosendale LE, Haslam DB, Lane A, Denson L, Lake K, Wilkey A, Watanabe M, Bauer S, Litts B, Luebbering N, Dandoy CE, Davies SM. Antibiotic Exposure and Reduced Short Chain Fatty Acid Production after Hematopoietic Stem Cell Transplant. Biol Blood Marrow Transplant 2018; 24:2418-2424. [PMID: 30055351 DOI: 10.1016/j.bbmt.2018.07.030] [Citation(s) in RCA: 83] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Accepted: 07/18/2018] [Indexed: 01/17/2023]
Abstract
Human studies have shown loss of diversity of the gut microbiome following hematopoietic stem cell transplantation (HSCT) in association with significant gut injury caused by the preparative regimen. Prolonged antibiotic use worsens loss of microbiome diversity and increases risk of complications such as graft-versus-host disease (GVHD). Our data support the hypothesis that loss of intestinal commensals that produce short-chain fatty acids (SCFAs) may increase dysbiosis. Here, we report an extensive longitudinal examination of changes in the luminal SCFAs in children undergoing allogeneic HSCT, and the relationship of those changes to the microbiota and antibiotic exposure. We found significant and progressive alterations in butyrate, and in additional SCFAs in stool in the first 14 days after transplant, a finding not observed in published mouse studies. SCFA levels were lower in children receiving antibiotics with activity against anaerobic organisms. Moreover, day 14 post-HSCT butyrate and propionate levels are lower in children who went on to develop GVHD, although our disease population was small. These data provide insight into the mechanism of prior observations that loss of diversity and increased antibiotic use are associated with GVHD following HSCT. Our findings offer potential modifiable targets to reduce risk of GVHD and improve survival after HSCT.
Collapse
Affiliation(s)
- Lindsey E Romick-Rosendale
- Division of Pathology and Laboratory Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio.
| | - David B Haslam
- Division of Infectious Diseases, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Adam Lane
- Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Lee Denson
- Division of Pediatric Gastroenterology, Hepatology, and Nutrition, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Kelly Lake
- Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Alyss Wilkey
- Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Miki Watanabe
- Division of Pathology and Laboratory Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Stuart Bauer
- Division of Pathology and Laboratory Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Bridget Litts
- Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Nathan Luebbering
- Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Christopher E Dandoy
- Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Stella M Davies
- Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| |
Collapse
|
402
|
Rashidi A, Shanley R, Yohe SL, Thyagarajan B, Curtsinger J, Anasetti C, Waller EK, Scott BL, Blazar BR, Weisdorf DJ. Recipient single nucleotide polymorphisms in Paneth cell antimicrobial peptide genes and acute graft-versus-host disease: analysis of BMT CTN-0201 and -0901 samples. Br J Haematol 2018; 182:887-894. [PMID: 30004111 DOI: 10.1111/bjh.15492] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2018] [Accepted: 05/14/2018] [Indexed: 01/06/2023]
Abstract
Host genetics shape the gut microbiota, and gut dysbiosis increases the risk of acute graft-versus-host disease (aGVHD). Paneth cells and microbiota have interactions that contribute to immune regulation. α-defensin-5 (HD5) and regenerating islet-derived protein 3 alpha (Reg3A) are the most abundant Paneth cell antimicrobial peptides (AMPs). We hypothesized that single nucleotide polymorphisms (SNPs) in the genes for HD5 (DEFA5) and Reg3A (REG3A) predict aGVHD risk. We analysed pre-transplant recipient peripheral blood mononuclear cell samples from randomized Blood and Marrow Transplant Clinical Trials Network (BMT CTN) studies 0201 (94 patients with bone marrow and 93 with peripheral blood grafts) and 0901 (86 patients with myeloablative and 77 with reduced-intensity conditioning; all using peripheral blood grafts). In multivariable analysis (with a SNP × graft source interaction term in CTN-0201 and a SNP × conditioning intensity term in CTN-0901), DEFA5 rs4415345 and rs4610776 were associated with altered incidence of aGVHD grade II-IV [rs4415345 G vs. C: hazard ratio (HR) 0·58, 95% confidence interval (95% CI) 0·37-0·92, P = 0·02; rs4610776 T vs. A: HR 1·53, 95% CI 1·01-2·32, P = 0·05] in CTN-0201, but not CTN-0901, suggesting a stronger effect in bone marrow allografts. REG3A SNP was not associated with aGVHD. Host genetics may influence aGVHD risk by modulating Paneth cell function.
Collapse
Affiliation(s)
- Armin Rashidi
- Division of Hematology, Oncology, and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Ryan Shanley
- Masonic Cancer Center Biostatistics Core, University of Minnesota, Minneapolis, MN, USA
| | - Sophia L Yohe
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, USA
| | - Bharat Thyagarajan
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, USA
| | - Julie Curtsinger
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
| | | | - Edmund K Waller
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, USA
| | - Bart L Scott
- Fred Hutchinson Cancer Research Center, University of Washington Medical Center/Seattle Cancer Care Alliance, Seattle, WA, USA
| | - Bruce R Blazar
- Division of Blood and Marrow Transplantation, Department of Pediatrics, University of Minnesota, Minneapolis, MN, USA
| | - Daniel J Weisdorf
- Division of Hematology, Oncology, and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
403
|
Kim DS, Kwon JE, Lee SH, Kim EK, Ryu JG, Jung KA, Choi JW, Park MJ, Moon YM, Park SH, Cho ML, Kwok SK. Attenuation of Rheumatoid Inflammation by Sodium Butyrate Through Reciprocal Targeting of HDAC2 in Osteoclasts and HDAC8 in T Cells. Front Immunol 2018; 9:1525. [PMID: 30034392 PMCID: PMC6043689 DOI: 10.3389/fimmu.2018.01525] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Accepted: 06/20/2018] [Indexed: 01/14/2023] Open
Abstract
Rheumatoid arthritis (RA) is a systemic autoimmune disease caused by both genetic and environmental factors. Recently, investigators have focused on the gut microbiota, which is thought to be an environmental factor that affects the development of RA. Metabolites secreted by the gut microbiota maintain homeostasis in the gut through various mechanisms [e.g., butyrate, which is one of the major metabolites of gut microbiota, exerts an anti-inflammatory effect by activating G-protein-coupled receptors and inhibiting histone deacetylases (HDACs)]. Here, we focused on the inhibition of the HDACs by butyrate in RA. To this end, we evaluated the therapeutic effects of butyrate in an animal model of autoimmune arthritis. The arthritis score and incidence were lower in the butyrate-treated group compared to the control group. Also, butyrate inhibited HDAC2 in osteoclasts and HDAC8 in T cells, leading to the acetylation of glucocorticoid receptors and estrogen-related receptors α, respectively. Additionally, control of the TH17/Treg cell balance and inhibition of osteoclastogenesis were confirmed by the changes in target gene expression. Interleukin-10 (IL-10) produced by butyrate-induced expanded Treg cells was critical, as treatment with butyrate did not affect inflammatory arthritis in IL-10-knockout mice. This immune-cell regulation of butyrate was also detected in humans. These findings suggest that butyrate is a candidate agent for the treatment of RA.
Collapse
Affiliation(s)
- Da Som Kim
- The Rheumatism Research Center, College of Medicine, Catholic Research Institute of Medical Science, The Catholic University of Korea, Seoul, South Korea
| | - Jeong-Eun Kwon
- The Rheumatism Research Center, College of Medicine, Catholic Research Institute of Medical Science, The Catholic University of Korea, Seoul, South Korea
| | - Seung Hoon Lee
- The Rheumatism Research Center, College of Medicine, Catholic Research Institute of Medical Science, The Catholic University of Korea, Seoul, South Korea.,Division of Immunology, Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA, United States
| | - Eun Kyung Kim
- The Rheumatism Research Center, College of Medicine, Catholic Research Institute of Medical Science, The Catholic University of Korea, Seoul, South Korea
| | - Jun-Geol Ryu
- The Rheumatism Research Center, College of Medicine, Catholic Research Institute of Medical Science, The Catholic University of Korea, Seoul, South Korea
| | | | - Jeong-Won Choi
- The Rheumatism Research Center, College of Medicine, Catholic Research Institute of Medical Science, The Catholic University of Korea, Seoul, South Korea
| | - Min-Jung Park
- The Rheumatism Research Center, College of Medicine, Catholic Research Institute of Medical Science, The Catholic University of Korea, Seoul, South Korea
| | - Young-Mee Moon
- The Rheumatism Research Center, College of Medicine, Catholic Research Institute of Medical Science, The Catholic University of Korea, Seoul, South Korea
| | - Sung-Hwan Park
- Division of Rheumatology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Mi-La Cho
- The Rheumatism Research Center, College of Medicine, Catholic Research Institute of Medical Science, The Catholic University of Korea, Seoul, South Korea.,Laboratory of Immune Network, Conversant Research Consortium in Immunologic Disease, College of Medicine, The Catholic University of Korea, Seoul, South Korea.,College of Medicine, The Institute for Aging and Metabolic Diseases, The Catholic University of Korea, Seoul, South Korea
| | - Seung-Ki Kwok
- Division of Rheumatology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| |
Collapse
|
404
|
Gill PA, van Zelm MC, Muir JG, Gibson PR. Review article: short chain fatty acids as potential therapeutic agents in human gastrointestinal and inflammatory disorders. Aliment Pharmacol Ther 2018; 48:15-34. [PMID: 29722430 DOI: 10.1111/apt.14689] [Citation(s) in RCA: 341] [Impact Index Per Article: 48.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Revised: 02/09/2018] [Accepted: 04/06/2018] [Indexed: 12/12/2022]
Abstract
BACKGROUND Butyrate, propionate and acetate are short chain fatty acids (SCFA), important for maintaining a healthy colon and are considered as protective in colorectal carcinogenesis. However, they may also regulate immune responses and the composition of the intestinal microbiota. Consequently, their importance in a variety of chronic inflammatory diseases is emerging. AIMS To review the physiology and metabolism of SCFA in humans, cellular and molecular mechanisms by which SCFA may act in health and disease, and approaches for therapeutic delivery of SCFA. METHODS A PubMed literature search was conducted for clinical and pre-clinical studies using search terms: 'dietary fibre', short-chain fatty acids', 'acetate', 'propionate', 'butyrate', 'inflammation', 'immune', 'gastrointestinal', 'metabolism'. RESULTS A wide range of pre-clinical evidence supports roles for SCFA as modulators of not only colonic function, but also multiple inflammatory and metabolic processes. SCFA are implicated in many autoimmune, allergic and metabolic diseases. However, translating effects of SCFA from animal studies to human disease is limited by physiological and dietary differences and by the challenge of delivering sufficient amounts of SCFA to the target sites that include the colon and the systemic circulation. Development of novel targeted approaches for colonic delivery, combined with postbiotic supplementation, may represent desirable strategies to achieve adequate targeted SCFA delivery. CONCLUSIONS There is a large array of potential disease-modulating effects of SCFA. Adequate targeted delivery to the sites of action is the main limitation of such application. The ongoing development and evaluation of novel delivery techniques offer potential for translating promise to therapeutic benefit.
Collapse
Affiliation(s)
- P A Gill
- Department of Gastroenterology, Central Clinical School, Monash University and Alfred Hospital, Melbourne, Vic., Australia.,Department of Immunology and Pathology, Central Clinical School, Monash University and Alfred Hospital, Melbourne, Vic, Australia
| | - M C van Zelm
- Department of Immunology and Pathology, Central Clinical School, Monash University and Alfred Hospital, Melbourne, Vic, Australia
| | - J G Muir
- Department of Gastroenterology, Central Clinical School, Monash University and Alfred Hospital, Melbourne, Vic., Australia
| | - P R Gibson
- Department of Gastroenterology, Central Clinical School, Monash University and Alfred Hospital, Melbourne, Vic., Australia
| |
Collapse
|
405
|
Haak BW, Littmann ER, Chaubard JL, Pickard AJ, Fontana E, Adhi F, Gyaltshen Y, Ling L, Morjaria SM, Peled JU, van den Brink MR, Geyer AI, Cross JR, Pamer EG, Taur Y. Impact of gut colonization with butyrate-producing microbiota on respiratory viral infection following allo-HCT. Blood 2018; 131:2978-2986. [PMID: 29674425 PMCID: PMC6024637 DOI: 10.1182/blood-2018-01-828996] [Citation(s) in RCA: 122] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Accepted: 03/24/2018] [Indexed: 01/01/2023] Open
Abstract
Respiratory viral infections are frequent in patients undergoing allogeneic hematopoietic stem cell transplantation (allo-HCT) and can potentially progress to lower respiratory tract infection (LRTI). The intestinal microbiota contributes to resistance against viral and bacterial pathogens in the lung. However, whether intestinal microbiota composition and associated changes in microbe-derived metabolites contribute to the risk of LRTI following upper respiratory tract viral infection remains unexplored in the setting of allo-HCT. Fecal samples from 360 allo-HCT patients were collected at the time of stem cell engraftment and subjected to deep, 16S ribosomal RNA gene sequencing to determine microbiota composition, and short-chain fatty acid levels were determined in a nested subset of fecal samples. The development of respiratory viral infections and LRTI was determined for 180 days following allo-HCT. Clinical and microbiota risk factors for LRTI were subsequently evaluated using survival analysis. Respiratory viral infection occurred in 149 (41.4%) patients. Of those, 47 (31.5%) developed LRTI. Patients with higher abundances of butyrate-producing bacteria were fivefold less likely to develop viral LRTI, independent of other factors (adjusted hazard ratio = 0.22, 95% confidence interval 0.04-0.69). Higher representation of butyrate-producing bacteria in the fecal microbiota is associated with increased resistance against respiratory viral infection with LRTI in allo-HCT patients.
Collapse
Affiliation(s)
- Bastiaan W Haak
- Infectious Disease Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
- Center for Experimental and Molecular Medicine, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Eric R Littmann
- Infectious Disease Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
- Center for Microbes, Inflammation, and Cancer and
| | - Jean-Luc Chaubard
- Donald B. and Catherine C. Marron Cancer Metabolism Center, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Amanda J Pickard
- Donald B. and Catherine C. Marron Cancer Metabolism Center, Memorial Sloan Kettering Cancer Center, New York, NY
| | | | - Fatima Adhi
- Infectious Diseases Service, New York University School of Medicine, New York, NY
| | | | - Lilan Ling
- Center for Microbes, Inflammation, and Cancer and
| | - Sejal M Morjaria
- Infectious Disease Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
- Center for Microbes, Inflammation, and Cancer and
- Weill Cornell Medical College, New York, NY; and
| | - Jonathan U Peled
- Weill Cornell Medical College, New York, NY; and
- Adult Bone Marrow Transplant Service and
| | - Marcel R van den Brink
- Weill Cornell Medical College, New York, NY; and
- Adult Bone Marrow Transplant Service and
| | - Alexander I Geyer
- Weill Cornell Medical College, New York, NY; and
- Pulmonary Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Justin R Cross
- Donald B. and Catherine C. Marron Cancer Metabolism Center, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Eric G Pamer
- Infectious Disease Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
- Center for Microbes, Inflammation, and Cancer and
- Weill Cornell Medical College, New York, NY; and
| | - Ying Taur
- Infectious Disease Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
- Center for Microbes, Inflammation, and Cancer and
- Weill Cornell Medical College, New York, NY; and
| |
Collapse
|
406
|
Advances in the understanding and management of mucositis during stem cell transplantation. Curr Opin Support Palliat Care 2018; 11:341-346. [PMID: 28984677 DOI: 10.1097/spc.0000000000000310] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
PURPOSE OF REVIEW Mucositis is a severe and common side effect of anticancer treatments, with an incidence of between 40 and 80% depending on the cytotoxic regimen used. The most profound mucositis burden is experienced during conditioning regimens for hematopoietic stem cell transplant (HSCT), where the use of highly mucotoxic agents with or without total body irradiation leads to serious damage throughout the alimentary tract. Currently, the assessment and management of both oral and gastrointestinal mucositis lack authoritative guideline, with recommendations only achieved in narrow clinical scenarios. This review provides a brief overview of current management guidelines for mucositis in both adult and pediatric patients receiving HSCT, highlights recent advances in mucositis prevention and discusses future research avenues. RECENT FINDINGS The Multinational Association of Supportive Care in Cancer and International Society for Oral Oncology (MASCC/ISOO) guidelines for the prevention of mucositis in HSCT are scarce, with low level laser therapy (photobiomodulation) and palifermin only recommended for oral mucositis. Loperamide and octreotide remain gold-standard for the treatment of diarrhea, despite poor efficacy. Although several interventions have been trialled in pediatric cohorts, no recommendations currently exist for children receiving high-dose chemotherapy or total body irradiation for HSCT. SUMMARY HSCT continues to be associated with mucositis, which impacts on patients' ability and willingness to receive engraftment, and worsens clinical outcome. Research into the prevention and treatment of mucositis in this setting remains limited, with an overwhelming amount of small, single-center studies that fail to achieve a sufficient level of evidence that warrant recommendation(s). As such, our ability to manage mucotoxic side effects of high-dose chemotherapy and irradiation is limited, particularly in children.
Collapse
|
407
|
Wu X, Wu Y, He L, Wu L, Wang X, Liu Z. Effects of the intestinal microbial metabolite butyrate on the development of colorectal cancer. J Cancer 2018; 9:2510-2517. [PMID: 30026849 PMCID: PMC6036887 DOI: 10.7150/jca.25324] [Citation(s) in RCA: 136] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2018] [Accepted: 05/02/2018] [Indexed: 12/15/2022] Open
Abstract
Colorectal cancer (CRC) is one of the major health threats in developed countries. Changes in dietary components, such as more protein and lipid intake, can increase the risk of CRC. Diet affects CRC in many ways. They regulate the composition and function of gut microbiota, which have an amazing metabolic capacity and can produce short chain fatty acids (SCFAs), such as propionate, acetate, and butyrate. Butyrate is a principal energy source for colonic epithelial cells and plays an important role in maintaining the stability of gut microbiota and the integrity of intestinal epithelium. However, there are few studies reviewing the anti-CRC potentials of butyrate. This review summarizes the recent research progresses in the effect of gut microbiota imbalance and the decrease in intestinal microbial metabolite butyrate caused by unbalanced diet on CRC development, and discusses the mechanisms of butyrate-induced anti-CRC activities, which may guide people to prevent CRC by improving diet structures.
Collapse
Affiliation(s)
- Xinqiang Wu
- Gannan Medical University, Ganzhou, Jiangxi, China
| | - Yuanbing Wu
- The First People's Hospital of Jiashan County, Jiaxing, Zhejiang, China
| | - Liangmei He
- The First Affiliated Hospital, Gannan Medical University, Ganzhou, Jiangxi, China
| | - Longhuo Wu
- College of Pharmacy, Gannan Medical University, Ganzhou, Jiangxi, China
| | - Xiangcai Wang
- The First Affiliated Hospital, Gannan Medical University, Ganzhou, Jiangxi, China
| | - Zhiping Liu
- School of Basic Medicine, Gannan Medical University, Ganzhou, Jiangxi, China
- Ganzhou Cancer Precision Engineering Research Center, Ganzhou, Jiangxi, China
| |
Collapse
|
408
|
Triff K, McLean MW, Callaway E, Goldsby J, Ivanov I, Chapkin RS. Dietary fat and fiber interact to uniquely modify global histone post-translational epigenetic programming in a rat colon cancer progression model. Int J Cancer 2018; 143:1402-1415. [PMID: 29659013 DOI: 10.1002/ijc.31525] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2017] [Revised: 02/21/2018] [Accepted: 03/22/2018] [Indexed: 12/15/2022]
Abstract
Dietary fermentable fiber generates short-chain fatty acids (SCFA), for example, butyrate, in the colonic lumen which serves as a chemoprotective histone deacetylase inhibitor and/or as an acetylation substrate for histone acetylases. In addition, n-3 polyunsaturated fatty acids (n-3 PUFA) in fish oil can affect the chromatin landscape by acting as ligands for tumor suppressive nuclear receptors. In an effort to gain insight into the global dimension of post-translational modification of histones (including H3K4me3 and H3K9ac) and clarify the chemoprotective impact of dietary bioactive compounds on transcriptional control in a preclinical model of colon cancer, we generated high-resolution genome-wide RNA (RNA-Seq) and "chromatin-state" (H3K4me3-seq and H3K9ac-seq) maps for intestinal (epithelial colonocytes) crypts in rats treated with a colon carcinogen and fed diets containing bioactive (i) fish oil, (ii) fermentable fiber (a rich source of SCFA), (iii) a combination of fish oil plus pectin, or (iv) control, devoid of fish oil or pectin. In general, poor correlation was observed between differentially transcribed (DE) and enriched genes (DERs) at multiple epigenetic levels. The combinatorial diet (fish oil + pectin) uniquely affected transcriptional profiles in the intestinal epithelium, for example, upregulating lipid catabolism and beta-oxidation associated genes. These genes were linked to activated ligand-dependent nuclear receptors associated with n-3 PUFA and were also correlated with the mitochondrial L-carnitine shuttle and the inhibition of lipogenesis. These findings demonstrate that the chemoprotective fish oil + pectin combination diet uniquely induces global histone state modifications linked to the expression of chemoprotective genes.
Collapse
Affiliation(s)
- Karen Triff
- Department of Nutrition and Food Science - Program in Integrative Nutrition and Complex Diseases, Texas A&M University, College Station, TX
- Department of Biology, Texas A&M University, College Station, TX
| | - Mathew W McLean
- Department of Statistics, Texas A&M University, College Station, TX
| | - Evelyn Callaway
- Department of Nutrition and Food Science - Program in Integrative Nutrition and Complex Diseases, Texas A&M University, College Station, TX
| | - Jennifer Goldsby
- Department of Nutrition and Food Science - Program in Integrative Nutrition and Complex Diseases, Texas A&M University, College Station, TX
| | - Ivan Ivanov
- Veterinary Physiology & Pharmacology, Texas A&M University, College Station, TX
| | - Robert S Chapkin
- Department of Nutrition and Food Science - Program in Integrative Nutrition and Complex Diseases, Texas A&M University, College Station, TX
| |
Collapse
|
409
|
Santos e Sousa P, Bennett CL, Chakraverty R. Unraveling the Mechanisms of Cutaneous Graft-Versus-Host Disease. Front Immunol 2018; 9:963. [PMID: 29770141 PMCID: PMC5940745 DOI: 10.3389/fimmu.2018.00963] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Accepted: 04/18/2018] [Indexed: 12/20/2022] Open
Abstract
The skin is the most common target organ affected by graft-versus-host disease (GVHD), with severity and response to therapy representing important predictors of patient survival. Although many of the initiating events in GVHD pathogenesis have been defined, less is known about why treatment resistance occurs or why there is often a permanent failure to restore tissue homeostasis. Emerging data suggest that the unique immune microenvironment in the skin is responsible for defining location- and context-specific mechanisms of injury that are distinct from those involved in other target organs. In this review, we address recent advances in our understanding of GVHD biology in the skin and outline the new research themes that will ultimately enable design of precision therapies.
Collapse
Affiliation(s)
- Pedro Santos e Sousa
- UCL Cancer Institute, University College London, London, United Kingdom
- UCL Institute of Immunity and Transplantation, University College London, London, United Kingdom
| | - Clare L. Bennett
- UCL Cancer Institute, University College London, London, United Kingdom
- UCL Institute of Immunity and Transplantation, University College London, London, United Kingdom
| | - Ronjon Chakraverty
- UCL Cancer Institute, University College London, London, United Kingdom
- UCL Institute of Immunity and Transplantation, University College London, London, United Kingdom
| |
Collapse
|
410
|
Abstract
Allogeneic haematopoietic stem cell transplantation (allo-HSCT) is considered to be the strongest curative immunotherapy for various malignancies (primarily, but not limited to, haematologic malignancies). However, application of allo-HSCT is limited owing to its life-threatening major complications, such as graft-versus-host disease (GVHD), relapse and infections. Recent advances in large-scale DNA sequencing technology have facilitated rapid identification of the microorganisms that make up the microbiota and evaluation of their interactions with host immunity in various diseases, including cancer. This has resulted in renewed interest regarding the role of the intestinal flora in patients with haematopoietic malignancies who have received an allo-HSCT and in whether the microbiota affects clinical outcomes, including GVHD, relapse, infections and transplant-related mortality. In this Review, we discuss the potential role of intestinal microbiota in these major complications after allo-HSCT, summarize clinical trials evaluating the microbiota in patients who have received allo-HSCT and discuss how further studies of the microbiota could inform the development of strategies that improve outcomes of allo-HSCT.
Collapse
Affiliation(s)
- Yusuke Shono
- Department of Immunology, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York, USA
- Department of Medicine, State University of New York Downstate Medical Center, Brooklyn, New York, USA
| | - Marcel R. M. van den Brink
- Department of Immunology, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York, USA
- Weill Medical College of Cornell University, New York, New York, USA
- Adult BMT Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| |
Collapse
|
411
|
Han L, Jin H, Zhou L, Zhang X, Fan Z, Dai M, Lin Q, Huang F, Xuan L, Zhang H, Liu Q. Intestinal Microbiota at Engraftment Influence Acute Graft-Versus-Host Disease via the Treg/Th17 Balance in Allo-HSCT Recipients. Front Immunol 2018; 9:669. [PMID: 29740427 PMCID: PMC5928130 DOI: 10.3389/fimmu.2018.00669] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2018] [Accepted: 03/19/2018] [Indexed: 12/20/2022] Open
Abstract
Animal models have indicated that intestinal microbiota influence acute graft-versus-host disease (aGVHD) by modulating immune homeostasis. But, in humans, the mechanism by which the microbiota induces aGVHD remains unclear. In this study, we investigated the relationship between the intestinal microbiota and T cell subsets in patients who undergo allogeneic hematopoietic stem cell transplantation (allo-HSCT) to explore the mechanism by which microbiota induced aGVHD. Based on aGVHD, this study was categorized into two groups: grades II–IV aGVHD (aGVHD group, n = 32) and grade 0–I aGVHD (non-aGVHD group, n = 49). The intestinal microbiota was detected by 16S rRNA gene sequencing, and the T cell subsets and histone 3 (H3) acetylation in CD4+ T cells in the peripheral blood was assayed by flow cytometry at the time of engraftment. The aGVHD group had greater low microbial diversity than the non-aGVHD group (56.3 versus 24.5%, p = 0.004). The bacterial community was depleted of Clostridia (e.g., the Lachnospiraceae and Ruminococcaceae families) and enriched for Gammaproteobacteria (e.g., the Enterobacteriaceae family) in the aGVHD group compared with the non-aGVHD group. The relative abundance of Lachnospiraceae and Ruminococcaceae was positively correlated with the Treg/Th17 ratio counts (r = 0.469 and 0.419; p < 0.001 and <0.001, respectively), whereas Enterobacteriaceae was negatively correlated with the Treg/Th17 ratio (r = −0.277; p = 0.012). The level of acetylated H3 in CD4+ T cells was not only correlated with Lachnospiraceae/Ruminococcaceae, but also with the Treg/Th17 ratio (r = 0.354; p = 0.001). In conclusions, our results suggest that decreased Lachnospiraceae and Ruminococcaceae and increased Enterobacteriaceae, correlate with a Treg/Th17 imbalance, which might be through acetylated H3 in CD4+ T cells. These findings suggest that intestinal microbiota might induce aGVHD by influencing the Treg/Th17 balance.
Collapse
Affiliation(s)
- Lijie Han
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Hua Jin
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Lizhi Zhou
- Department of Biostatistics, Southern Medical University, Guangzhou, China
| | - Xin Zhang
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zhiping Fan
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Min Dai
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Qianyun Lin
- Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Fen Huang
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Li Xuan
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Haiyan Zhang
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Qifa Liu
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
412
|
Phan TL, Carlin K, Ljungman P, Politikos I, Boussiotis V, Boeckh M, Shaffer ML, Zerr DM. Human Herpesvirus-6B Reactivation Is a Risk Factor for Grades II to IV Acute Graft-versus-Host Disease after Hematopoietic Stem Cell Transplantation: A Systematic Review and Meta-Analysis. Biol Blood Marrow Transplant 2018; 24:2324-2336. [PMID: 29684567 PMCID: PMC8934525 DOI: 10.1016/j.bbmt.2018.04.021] [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: 02/01/2018] [Accepted: 04/17/2018] [Indexed: 12/17/2022]
Abstract
Graft-versus-host disease (GVHD) is an important cause of morbidity and mortality after allogeneic hematopoietic cell transplantation (HCT). Many studies have suggested that human herpesvirus-6B (HHV-6B) plays a role in acute GVHD (aGVHD) after HCT. Our objective was to systematically summarize and analyze evidence regarding HHV-6B reactivation and development of aGVHD. PubMed and EMBASE databases were searched using terms for HHV-6, HCT, and aGVHD, yielding 865 unique results. Case reports, reviews, articles focusing on inherited chromosomally integrated HHV-6, poster presentations, and articles not published in English were excluded. The remaining 467 articles were reviewed for the following requirements: a statistical analysis of HHV-6B reactivation and a GVHD was described, HHV-6B reactivation was defined by PCR, and blood (plasma, serum, or peripheral blood mononuclear cells) was used for HHV-6B PCR. Data were abstracted from publications that met these criteria (n = 33). Publications were assigned to 1 of 3 groups: (1) HHV-6B reactivation was analyzed as a time-dependent risk factor for subsequent aGVHD (n = 14), (2) aGVHD was analyzed as a time-dependent risk factor for subsequent HHV-6B reactivation (n = 1), and (3) analysis without temporal specification (n = 18). A statistically significant association (P < .05) between HHV-6B reactivation and aGVHD was observed in 10 of 14 studies (71%) in group 1, 0 of 1 study (0%) in Group 2, and 8 of 18 studies (44.4%) in Group 3. Of the 14 studies that analyzed HHV-6B as a risk factor for subsequent aGVHD, 11 performed a multivariate analysis and reported a hazard ratio, which reached statistical significance in 9 of these s tudies. Meta-analysis of these 11 studies demonstrated a statistically significant association between HHV-6B and subsequent grades II to IV aGVHD (hazard ratio, 2.65; 95% confidence interval, 1.89 to 3.72; P < .001).HHV-6B reactivation is associated with aGVHD, and when studies have a temporal component to their design, HHV-6B reactivation is associated with subsequent aGVHD. Further research is needed to investigate whether antiviral prophylaxis reduces incidence or severity of aGVHD.
Collapse
Affiliation(s)
- Tuan L Phan
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, Louisiana; HHV-6 Foundation, Santa Barbara, California
| | - Kristen Carlin
- Center for Clinical and Translational Research, Seattle Children's Research Institute, Seattle, Washington
| | - Per Ljungman
- Department of Cellular Therapy and Allogeneic Stem Cell Transplantation, Karolinska University Hospital, Stockholm, Sweden; Division of Hematology, Department of Medicine, Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Ioannis Politikos
- Adult Bone Marrow Transplant Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Vicki Boussiotis
- Department of Hematology-Oncology and Cancer Biology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Michael Boeckh
- Department of Medicine, Vaccine and Infectious Disease and Clinical Research Division, Fred Hutchinson Cancer Research Center, University of Washington, Seattle, Washington
| | - Michele L Shaffer
- Center for Clinical and Translational Research, Seattle Children's Research Institute, Seattle, Washington; Department of Statistics, University of Washington, Seattle, Washington
| | - Danielle M Zerr
- Center for Clinical and Translational Research, Seattle Children's Research Institute, Seattle, Washington; Department of Pediatrics, University of Washington, Seattle, Washington.
| |
Collapse
|
413
|
Le Bastard Q, Ward T, Sidiropoulos D, Hillmann BM, Chun CL, Sadowsky MJ, Knights D, Montassier E. Fecal microbiota transplantation reverses antibiotic and chemotherapy-induced gut dysbiosis in mice. Sci Rep 2018; 8:6219. [PMID: 29670191 PMCID: PMC5906603 DOI: 10.1038/s41598-018-24342-x] [Citation(s) in RCA: 95] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Accepted: 03/23/2018] [Indexed: 01/07/2023] Open
Abstract
Fecal microbiota transplantation (FMT) is now widely used to treat recurrent Clostridium difficile infection, but has been less studied as a means to restore microbiome diversity and composition following antibiotic or chemotherapy treatments. The purpose of our study was to assess the efficacy of FMT to reverse antibiotic- and chemotherapy-induced gut dysbiosis in a mouse model. C57BL/6J mice were treated with ampicillin for 1 week and/or received a single intraperitoneal injection of 5-Fluorouracil. Fresh stool was collected and analyzed using shotgun metagenomics and the Illumina sequencing platform. Ampicillin caused a significant and immediate decrease in bacterial species richness and diversity that persisted for one week. In mice that received FMT, disruption of the intestinal microbiota was reversed immediately. Antibiotic and chemotherapy administration caused significant alteration in species distribution, including a decrease in the relative proportions of Clostridium scindens and Faecalibacterium prausnitzii, and an increase in known pathogenic species. In mice receiving FMT, we observed a significant increase in species known to exhibit anti-inflammatory properties. Moreover, chemotherapy led to a critical decrease in key ‘health-promoting’ species and to an altered functional profile, especially when chemotherapy was administered in tandem with antibiotics, and that FMT can ameliorate these effects.
Collapse
Affiliation(s)
- Quentin Le Bastard
- Université de Nantes, Microbiotas Hosts Antibiotics and bacterial Resistances (MiHAR), Nantes, 44000, France
| | - Tonya Ward
- Biotechnology Institute, University of Minnesota, Saint Paul, Minnesota, 55108, USA
| | - Dimitri Sidiropoulos
- Biotechnology Institute, University of Minnesota, Saint Paul, Minnesota, 55108, USA
| | - Benjamin M Hillmann
- Department of Computer Science and Engineering, University of Minnesota, Minneapolis, Minnesota, 55455, USA
| | - Chan Lan Chun
- Biotechnology Institute, University of Minnesota, Saint Paul, Minnesota, 55108, USA.,Department of Civil Engineering and National Resources Research Institute, University of Minnesota Duluth, Duluth, Minnesota, USA
| | - Michael J Sadowsky
- Biotechnology Institute, University of Minnesota, Saint Paul, Minnesota, 55108, USA.,Department of Soil Water & Climate, and Department of Plant and Microbial Biology, University of Minnesota, Saint Paul, Minnesota, 55108, USA
| | - Dan Knights
- Biotechnology Institute, University of Minnesota, Saint Paul, Minnesota, 55108, USA. .,Department of Computer Science and Engineering, University of Minnesota, Minneapolis, Minnesota, 55455, USA.
| | - Emmanuel Montassier
- Université de Nantes, Microbiotas Hosts Antibiotics and bacterial Resistances (MiHAR), Nantes, 44000, France.
| |
Collapse
|
414
|
DeFilipp Z, Peled JU, Li S, Mahabamunuge J, Dagher Z, Slingerland AE, Del Rio C, Valles B, Kempner ME, Smith M, Brown J, Dey BR, El-Jawahri A, McAfee SL, Spitzer TR, Ballen KK, Sung AD, Dalton TE, Messina JA, Dettmer K, Liebisch G, Oefner P, Taur Y, Pamer EG, Holler E, Mansour MK, van den Brink MRM, Hohmann E, Jenq RR, Chen YB. Third-party fecal microbiota transplantation following allo-HCT reconstitutes microbiome diversity. Blood Adv 2018; 2:745-753. [PMID: 29592876 PMCID: PMC5894265 DOI: 10.1182/bloodadvances.2018017731] [Citation(s) in RCA: 157] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Accepted: 02/27/2018] [Indexed: 12/16/2022] Open
Abstract
We hypothesized that third-party fecal microbiota transplantation (FMT) may restore intestinal microbiome diversity after allogeneic hematopoietic cell transplantation (allo-HCT). In this open-label single-group pilot study, 18 subjects were enrolled before allo-HCT and planned to receive third-party FMT capsules. FMT capsules were administered no later than 4 weeks after neutrophil engraftment, and antibiotics were not allowed within 48 hours before FMT. Five patients did not receive FMT because of the development of early acute gastrointestinal (GI) graft-versus-host disease (GVHD) before FMT (n = 3), persistent HCT-associated GI toxicity (n = 1), or patient decision (n = 1). Thirteen patients received FMT at a median of 27 days (range, 19-45 days) after HCT. Participants were able to swallow and tolerate all FMT capsules, meeting the primary study endpoint of feasibility. FMT was tolerated well, with 1 treatment-related significant adverse event (abdominal pain). Two patients subsequently developed acute GI GVHD, with 1 patient also having concurrent bacteremia. No additional cases of bacteremia occurred. Median follow-up for survivors is 15 months (range, 13-20 months). The Kaplan-Meier estimates for 12-month overall survival and progression-free survival after FMT were 85% (95% confidence interval, 51%-96%) and 85% (95% confidence interval, 51%-96%), respectively. There was 1 nonrelapse death resulting from acute GI GVHD (12-month nonrelapse mortality, 8%; 95% confidence interval, 0%-30%). Analysis of stool composition and urine 3-indoxyl sulfate concentration indicated improvement in intestinal microbiome diversity after FMT that was associated with expansion of stool-donor taxa. These results indicate that empiric third-party FMT after allo-HCT appears to be feasible, safe, and associated with expansion of recipient microbiome diversity. This trial was registered at www.clinicaltrials.gov as #NCT02733744.
Collapse
Affiliation(s)
- Zachariah DeFilipp
- Blood and Marrow Transplant Program, Massachusetts General Hospital, Boston, MA
| | - Jonathan U Peled
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
- Department of Medicine, Weill Cornell Medical College, New York, NY
| | - Shuli Li
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston, MA
| | | | - Zeina Dagher
- Division of Infectious Diseases, Massachusetts General Hospital, Boston, MA
| | - Ann E Slingerland
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
- Department of Medicine, Weill Cornell Medical College, New York, NY
| | - Candice Del Rio
- Blood and Marrow Transplant Program, Massachusetts General Hospital, Boston, MA
| | - Betsy Valles
- Blood and Marrow Transplant Program, Massachusetts General Hospital, Boston, MA
| | - Maria E Kempner
- Blood and Marrow Transplant Program, Massachusetts General Hospital, Boston, MA
| | - Melissa Smith
- Blood and Marrow Transplant Program, Massachusetts General Hospital, Boston, MA
| | - Jami Brown
- Blood and Marrow Transplant Program, Massachusetts General Hospital, Boston, MA
| | - Bimalangshu R Dey
- Blood and Marrow Transplant Program, Massachusetts General Hospital, Boston, MA
| | - Areej El-Jawahri
- Blood and Marrow Transplant Program, Massachusetts General Hospital, Boston, MA
| | - Steven L McAfee
- Blood and Marrow Transplant Program, Massachusetts General Hospital, Boston, MA
| | - Thomas R Spitzer
- Blood and Marrow Transplant Program, Massachusetts General Hospital, Boston, MA
| | - Karen K Ballen
- Division of Hematology/Oncology, University of Virginia School of Medicine, Charlottesville, VA
| | - Anthony D Sung
- Division of Hematologic Malignancies and Cellular Therapies and
| | - Tara E Dalton
- Division of Hematologic Malignancies and Cellular Therapies and
| | - Julia A Messina
- Division of Infectious Diseases, Duke University School of Medicine, Durham, NC
| | - Katja Dettmer
- Institute of Functional Genomics, University of Regensburg, Regensburg, Germany
| | - Gerhard Liebisch
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital Regensburg, Regensburg, Germany
| | - Peter Oefner
- Institute of Functional Genomics, University of Regensburg, Regensburg, Germany
| | - Ying Taur
- Department of Medicine, Weill Cornell Medical College, New York, NY
- Infectious Disease Service and Center for Microbes, Inflammation and Cancer, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Eric G Pamer
- Department of Medicine, Weill Cornell Medical College, New York, NY
- Infectious Disease Service and Center for Microbes, Inflammation and Cancer, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Ernst Holler
- Department of Hematology and Oncology, Internal Medicine III, University Medical Center, Regensburg, Germany; and
| | - Michael K Mansour
- Division of Infectious Diseases, Massachusetts General Hospital, Boston, MA
| | - Marcel R M van den Brink
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
- Department of Medicine, Weill Cornell Medical College, New York, NY
| | - Elizabeth Hohmann
- Division of Infectious Diseases, Massachusetts General Hospital, Boston, MA
| | - Robert R Jenq
- Department of Genomic Medicine and
- Department of Stem Cell Transplantation Cellular Therapy, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Yi-Bin Chen
- Blood and Marrow Transplant Program, Massachusetts General Hospital, Boston, MA
| |
Collapse
|
415
|
Varelias A, Bunting MD, Ormerod KL, Koyama M, Olver SD, Straube J, Kuns RD, Robb RJ, Henden AS, Cooper L, Lachner N, Gartlan KH, Lantz O, Kjer-Nielsen L, Mak JY, Fairlie DP, Clouston AD, McCluskey J, Rossjohn J, Lane SW, Hugenholtz P, Hill GR. Recipient mucosal-associated invariant T cells control GVHD within the colon. J Clin Invest 2018; 128:1919-1936. [PMID: 29629900 DOI: 10.1172/jci91646] [Citation(s) in RCA: 78] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Accepted: 02/16/2018] [Indexed: 12/11/2022] Open
Abstract
Mucosal-associated invariant T (MAIT) cells are a unique innate-like T cell subset that responds to a wide array of bacteria and yeast through recognition of riboflavin metabolites presented by the MHC class I-like molecule MR1. Here, we demonstrate using MR1 tetramers that recipient MAIT cells are present in small but definable numbers in graft-versus-host disease (GVHD) target organs and protect from acute GVHD in the colon following bone marrow transplantation (BMT). Consistent with their preferential juxtaposition to microbial signals in the colon, recipient MAIT cells generate large amounts of IL-17A, promote gastrointestinal tract integrity, and limit the donor alloantigen presentation that in turn drives donor Th1 and Th17 expansion specifically in the colon after BMT. Allogeneic BMT recipients deficient in IL-17A also develop accelerated GVHD, suggesting MAIT cells likely regulate GVHD, at least in part, by the generation of this cytokine. Indeed, analysis of stool microbiota and colon tissue from IL-17A-/- and MR1-/- mice identified analogous shifts in microbiome operational taxonomic units (OTU) and mediators of barrier integrity that appear to represent pathways controlled by similar, IL-17A-dependent mechanisms. Thus, MAIT cells act to control barrier function to attenuate pathogenic T cell responses in the colon and, given their very high frequency in humans, likely represent an important population in clinical BMT.
Collapse
Affiliation(s)
- Antiopi Varelias
- Bone Marrow Transplantation Laboratory, QIMR Berghofer Medical Research Institute, Brisbane, Australia.,Faculty of Medicine, and
| | - Mark D Bunting
- Bone Marrow Transplantation Laboratory, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Kate L Ormerod
- Australian Centre for Ecogenomics, The University of Queensland, Brisbane, Australia
| | - Motoko Koyama
- Bone Marrow Transplantation Laboratory, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Stuart D Olver
- Bone Marrow Transplantation Laboratory, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Jasmin Straube
- Gordon and Jessie Gilmour Leukaemia Research Laboratory, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Rachel D Kuns
- Bone Marrow Transplantation Laboratory, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Renee J Robb
- Bone Marrow Transplantation Laboratory, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Andrea S Henden
- Bone Marrow Transplantation Laboratory, QIMR Berghofer Medical Research Institute, Brisbane, Australia.,The Royal Brisbane and Women's Hospital, Brisbane, Australia
| | - Leanne Cooper
- Gordon and Jessie Gilmour Leukaemia Research Laboratory, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Nancy Lachner
- Australian Centre for Ecogenomics, The University of Queensland, Brisbane, Australia
| | - Kate H Gartlan
- Bone Marrow Transplantation Laboratory, QIMR Berghofer Medical Research Institute, Brisbane, Australia.,Faculty of Medicine, and
| | - Olivier Lantz
- INSERM U932 and Department de Biologie des Tumeurs, Institute Curie and Centre d'Investigation Clinique, CICBT507 IGR/Curie, Paris, France
| | - Lars Kjer-Nielsen
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Parkville, Australia
| | - Jeffrey Yw Mak
- Australian Research Council Centre of Excellence in Advanced Molecular Imaging, Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia
| | - David P Fairlie
- Australian Research Council Centre of Excellence in Advanced Molecular Imaging, Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia
| | | | - James McCluskey
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Parkville, Australia
| | - Jamie Rossjohn
- Infection and Immunity Program and The Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute Monash University, Clayton, Australia.,Institute of Infection and Immunity, Cardiff University School of Medicine, Cardiff, United Kingdom.,Australian Research Council Centre of Excellence in Advanced Molecular Imaging, Monash University, Clayton, Australia
| | - Steven W Lane
- Faculty of Medicine, and.,Gordon and Jessie Gilmour Leukaemia Research Laboratory, QIMR Berghofer Medical Research Institute, Brisbane, Australia.,The Royal Brisbane and Women's Hospital, Brisbane, Australia
| | - Philip Hugenholtz
- Australian Centre for Ecogenomics, The University of Queensland, Brisbane, Australia
| | - Geoffrey R Hill
- Bone Marrow Transplantation Laboratory, QIMR Berghofer Medical Research Institute, Brisbane, Australia.,Faculty of Medicine, and.,The Royal Brisbane and Women's Hospital, Brisbane, Australia
| |
Collapse
|
416
|
Ichinohe T, Miyama T, Kawase T, Honjo Y, Kitaura K, Sato H, Shin-I T, Suzuki R. Next-Generation Immune Repertoire Sequencing as a Clue to Elucidate the Landscape of Immune Modulation by Host-Gut Microbiome Interactions. Front Immunol 2018; 9:668. [PMID: 29666626 PMCID: PMC5891584 DOI: 10.3389/fimmu.2018.00668] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2017] [Accepted: 03/19/2018] [Indexed: 12/23/2022] Open
Abstract
The human immune system is a fine network consisted of the innumerable numbers of functional cells that balance the immunity and tolerance against various endogenous and environmental challenges. Although advances in modern immunology have revealed a role of many unique immune cell subsets, technologies that enable us to capture the whole landscape of immune responses against specific antigens have been not available to date. Acquired immunity against various microorganisms including host microbiome is principally founded on T cell and B cell populations, each of which expresses antigen-specific receptors that define a unique clonotype. Over the past several years, high-throughput next-generation sequencing has been developed as a powerful tool to profile T- and B-cell receptor repertoires in a given individual at the single-cell level. Sophisticated immuno-bioinformatic analyses by use of this innovative methodology have been already implemented in clinical development of antibody engineering, vaccine design, and cellular immunotherapy. In this article, we aim to discuss the possible application of high-throughput immune receptor sequencing in the field of nutritional and intestinal immunology. Although there are still unsolved caveats, this emerging technology combined with single-cell transcriptomics/proteomics provides a critical tool to unveil the previously unrecognized principle of host–microbiome immune homeostasis. Accumulation of such knowledge will lead to the development of effective ways for personalized immune modulation through deeper understanding of the mechanisms by which the intestinal environment affects our immune ecosystem.
Collapse
Affiliation(s)
- Tatsuo Ichinohe
- Department of Hematology and Oncology, Research Institute for Radiation Biology and Medicine (RIRBM), Hiroshima University, Hiroshima, Japan
| | - Takahiko Miyama
- Department of Hematology and Oncology, Research Institute for Radiation Biology and Medicine (RIRBM), Hiroshima University, Hiroshima, Japan
| | - Takakazu Kawase
- Department of Hematology and Oncology, Research Institute for Radiation Biology and Medicine (RIRBM), Hiroshima University, Hiroshima, Japan
| | - Yasuko Honjo
- Department of Hematology and Oncology, Research Institute for Radiation Biology and Medicine (RIRBM), Hiroshima University, Hiroshima, Japan
| | | | | | | | - Ryuji Suzuki
- Repertoire Genesis Incorporation, Ibaraki, Japan.,Department of Rheumatology and Clinical Immunology, Clinical Research Center for Rheumatology and Allergy, National Hospital Organization Sagamihara Hospital, Sagamihara, Japan
| |
Collapse
|
417
|
Abstract
Fermented milk products like yogurt or soft cheese provide calcium, phosphorus, and protein. All these nutrients influence bone growth and bone loss. In addition, fermented milk products may contain prebiotics like inulin which may be added to yogurt, and provide probiotics which are capable of modifying intestinal calcium absorption and/or bone metabolism. On the other hand, yogurt consumption may ensure a more regular ingestion of milk products and higher compliance, because of various flavors and sweetness. Bone mass accrual, bone homeostasis, and attenuation of sex hormone deficiency-induced bone loss seem to benefit from calcium, protein, pre-, or probiotics ingestion, which may modify gut microbiota composition and metabolism. Fermented milk products might also represent a marker of lifestyle promoting healthy bone health.
Collapse
Affiliation(s)
- René Rizzoli
- Service of Bone Diseases, Faculty of Medicine, Geneva University Hospitals, 1211, Geneva 14, Switzerland.
| | - Emmanuel Biver
- Service of Bone Diseases, Faculty of Medicine, Geneva University Hospitals, 1211, Geneva 14, Switzerland
| |
Collapse
|
418
|
Beckerson J, Szydlo RM, Hickson M, Mactier CE, Innes AJ, Gabriel IH, Palanicawandar R, Kanfer EJ, Macdonald DH, Milojkovic D, Rahemtulla A, Chaidos A, Karadimitris A, Olavarria E, Apperley JF, Pavlu J. Impact of route and adequacy of nutritional intake on outcomes of allogeneic haematopoietic cell transplantation for haematologic malignancies. Clin Nutr 2018; 38:738-744. [PMID: 29650256 DOI: 10.1016/j.clnu.2018.03.008] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Accepted: 03/11/2018] [Indexed: 01/08/2023]
Abstract
BACKGROUND Allogeneic haematopoietic cell transplantation (HCT) is often associated with poor oral intake due to painful mucositis and gastrointestinal sequalae that occur following a preparative regimen of intensive chemotherapy and/or total body radiation. Although attractive to assume that optimal nutrition improves HCT outcomes, there are limited data to support this. It is also unclear whether artificial nutrition support should be provided as enteral tube feeding or parenteral nutrition (PN). METHODS We analysed day-100 non-relapse mortality (NRM), incidence of acute graft-versus-host disease (GvHD), acute gastrointestinal GvHD, 5-year survival and GvHD-free/relapse-free survival (GRFS) according to both route and adequacy of nutritional intake prior to neutrophil engraftment, together with other known prognostic factors, in a retrospective cohort of 484 patients who underwent allogeneic HCT for haematologic malignancy between 2000 and 2014. RESULTS Multivariate analyses showed increased NRM with inadequate nutrition (hazard ratio (HR) 4.1; 95% confidence interval (CI) 2.2-7.2) and adequate PN (HR 2.9; 95% CI 1.6-5.4) compared to adequate enteral nutrition (EN) both P < .001. There were increased incidences of gastrointestinal GvHD of any stage and all GvHD ≥ grade 2 in patients who received PN (odds ratio (OR) 2.0; 95% CI 1.2-3.3; P = .006, and OR 1.8; 95% CI 1.1-3.0; P = .018, respectively), compared to adequate EN. Patients who received adequate PN and inadequate nutrition also had reduced probabilities of survival and GRFS at 5 years. CONCLUSION Adequate EN during the early transplantation course is associated with reduced NRM, improved survival and GRFS at 5 years. Furthermore, adequate EN is associated with lower incidence of overall and gut acute GvHD than PN, perhaps because of its ability to maintain mucosal integrity, modulate the immune response to intensive chemo/radiotherapy and support the gastrointestinal tract environment, including gut microflora.
Collapse
Affiliation(s)
- Julie Beckerson
- Nutrition and Dietetics, Hammersmith Hospital, Imperial College Healthcare NHS Trust, London, UK.
| | - Richard M Szydlo
- Centre for Haematology, Imperial College London at Hammersmith Hospital, London, UK
| | - Mary Hickson
- Institute of Health and Community, Plymouth University, Devon, UK
| | - Catriona E Mactier
- Centre for Haematology, Imperial College London at Hammersmith Hospital, London, UK
| | - Andrew J Innes
- Centre for Haematology, Imperial College London at Hammersmith Hospital, London, UK
| | - Ian H Gabriel
- Centre for Haematology, Imperial College London at Hammersmith Hospital, London, UK
| | | | - Edward J Kanfer
- Centre for Haematology, Imperial College London at Hammersmith Hospital, London, UK
| | - Donald H Macdonald
- Centre for Haematology, Imperial College London at Hammersmith Hospital, London, UK
| | - Dragana Milojkovic
- Centre for Haematology, Imperial College London at Hammersmith Hospital, London, UK
| | - Amin Rahemtulla
- Centre for Haematology, Imperial College London at Hammersmith Hospital, London, UK
| | - Aristeidis Chaidos
- Centre for Haematology, Imperial College London at Hammersmith Hospital, London, UK
| | | | - Eduardo Olavarria
- Centre for Haematology, Imperial College London at Hammersmith Hospital, London, UK
| | - Jane F Apperley
- Centre for Haematology, Imperial College London at Hammersmith Hospital, London, UK
| | - Jiri Pavlu
- Centre for Haematology, Imperial College London at Hammersmith Hospital, London, UK
| |
Collapse
|
419
|
Cho JA, Chinnapen DJF. Targeting friend and foe: Emerging therapeutics in the age of gut microbiome and disease. J Microbiol 2018; 56:183-188. [PMID: 29492875 DOI: 10.1007/s12275-018-8037-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Revised: 02/09/2018] [Accepted: 02/11/2018] [Indexed: 12/31/2022]
Abstract
Mucosal surfaces that line our gastrointestinal tract are continuously exposed to trillions of bacteria that form a symbiotic relationship and impact host health and disease. It is only beginning to be understood that the cross-talk between the host and microbiome involve dynamic changes in commensal bacterial population, secretion, and absorption of metabolites between the host and microbiome. As emerging evidence implicates dysbiosis of gut microbiota in the pathology and progression of various diseases such as inflammatory bowel disease, obesity, and allergy, conventional treatments that either overlook the microbiome in the mechanism of action, or eliminate vast populations of microbes via wide-spectrum antibiotics need to be reconsidered. It is also becoming clear the microbiome can influence the body's response to therapeutic treatments for cancers. As such, targeting the microbiome as treatment has garnered much recent attention and excitement from numerous research labs and biotechnology companies. Treatments range from fecal microbial transplantation to precision-guided molecular approaches. Here, we survey recent progress in the development of innovative therapeutics that target the microbiome to treat disease, and highlight key findings in the interplay between host microbes and therapy.
Collapse
Affiliation(s)
- Jin Ah Cho
- Department of Food and Nutrition, Chungnam National University, Daejeon, 34134, Republic of Korea
| | - Daniel J F Chinnapen
- Division of Gastroenterology, Boston Children's Hospital, Boston, 02115, USA.
- Department of Pediatrics, Harvard Medical School, Boston, 02115, USA.
- Harvard Digestive Diseases Center, Boston, 02115, USA.
| |
Collapse
|
420
|
Neutrophils provide cellular communication between ileum and mesenteric lymph nodes at graft-versus-host disease onset. Blood 2018; 131:1858-1869. [PMID: 29463561 DOI: 10.1182/blood-2017-10-812891] [Citation(s) in RCA: 95] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Accepted: 02/09/2018] [Indexed: 12/13/2022] Open
Abstract
Conditioning-induced damage of the intestinal tract plays a critical role during the onset of acute graft-versus-host disease (GVHD). Therapeutic interference with these early events of GVHD is difficult, and currently used immunosuppressive drugs mainly target donor T cells. However, not donor T cells but neutrophils reach the sites of tissue injury first, and therefore could be a potential target for GVHD prevention. A detailed analysis of neutrophil fate during acute GVHD and the effect on T cells is difficult because of the short lifespan of this cell type. By using a novel photoconverter reporter system, we show that neutrophils that had been photoconverted in the ileum postconditioning later migrated to mesenteric lymph nodes (mLN). This neutrophil migration was dependent on the intestinal microflora. In the mLN, neutrophils colocalized with T cells and presented antigen on major histocompatibility complex (MHC)-II, thereby affecting T cell expansion. Pharmacological JAK1/JAK2 inhibition reduced neutrophil influx into the mLN and MHC-II expression, thereby interfering with an early event in acute GVHD pathogenesis. In agreement with this finding, neutrophil depletion reduced acute GVHD. We conclude that neutrophils are attracted to the ileum, where the intestinal barrier is disrupted, and then migrate to the mLN, where they participate in alloantigen presentation. JAK1/JAK2-inhibition can interfere with this process, which provides a potential therapeutic strategy to prevent early events of tissue damage-related innate immune cell activation and, ultimately, GVHD.
Collapse
|
421
|
Andermann TM, Peled JU, Ho C, Reddy P, Riches M, Storb R, Teshima T, van den Brink MRM, Alousi A, Balderman S, Chiusolo P, Clark WB, Holler E, Howard A, Kean LS, Koh AY, McCarthy PL, McCarty JM, Mohty M, Nakamura R, Rezvani K, Segal BH, Shaw BE, Shpall EJ, Sung AD, Weber D, Whangbo J, Wingard JR, Wood WA, Perales MA, Jenq RR, Bhatt AS. The Microbiome and Hematopoietic Cell Transplantation: Past, Present, and Future. Biol Blood Marrow Transplant 2018; 24:1322-1340. [PMID: 29471034 DOI: 10.1016/j.bbmt.2018.02.009] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Accepted: 02/08/2018] [Indexed: 01/07/2023]
Affiliation(s)
- Tessa M Andermann
- Division of Infectious Diseases, Department of Medicine, Stanford University, Stanford, California
| | - Jonathan U Peled
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York; Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Christine Ho
- Blood and Marrow Transplantation, Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, New York
| | - Pavan Reddy
- Department of Medicine, University of Michigan Cancer Center, Ann Arbor, Michigan
| | - Marcie Riches
- Division of Hematology/Oncology, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Rainer Storb
- Fred Hutchinson Cancer Research Center, Seattle, Washington; Department of Medicine, University of Washington School of Medicine, Seattle, Washington
| | - Takanori Teshima
- Department of Hematology, Hokkaido University Faculty of Medicine, Sapporo, Japan
| | - Marcel R M van den Brink
- Immunology Program, Sloan Kettering Institute, New York, New York; Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Amin Alousi
- Multidiscipline GVHD Clinic and Research Program, Department of Stem Cell Transplant and Cellular Therapies, University of Texas, MD Anderson Cancer Center, Houston, Texas
| | - Sophia Balderman
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, New York
| | - Patrizia Chiusolo
- Hematology Department, Fondazione Policlinico Universitario A. Gemelli, Università Cattolica Sacro Cuore, Rome, Italy
| | - William B Clark
- Bone Marrow Transplant Program, Division of Hematology/Oncology and Palliative Care, Department of Internal Medicine, Virginia Commonwealth University, Richmond, Virginia
| | - Ernst Holler
- Department of Internal Medicine 3, University Medical Center, Regensburg, Germany
| | - Alan Howard
- Center for International Blood and Marrow Transplant Research, Minneapolis, Minnesota
| | - Leslie S Kean
- Fred Hutchinson Cancer Research Center, Seattle, Washington; Department of Pediatrics, University of Washington School of Medicine, Seattle, Washington; Ben Towne Center for Childhood Cancer Research, Seattle Children's Research Institute, Seattle, Washington
| | - Andrew Y Koh
- Divisions of Hematology/Oncology and Infectious Diseases, Departments of Pediatrics and Microbiology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Philip L McCarthy
- Blood and Marrow Transplantation, Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, New York
| | - John M McCarty
- Bone Marrow Transplantation Program, Virginia Commonwealth University Massey Cancer, Richmond, Virginia
| | - Mohamad Mohty
- Clinical Hematology and Cellular Therapy Department, Hôpital Saint-Antoine, AP-HP, Paris, France; Sorbonne Université, Paris, France; INSERM UMRs U938, Paris, France
| | - Ryotaro Nakamura
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope, Duarte, California
| | - Katy Rezvani
- Section of Cellular Therapy, Good Manufacturing Practices Facility, Department of Stem Cell Transplant and Cellular Therapy, University of Texas MD Anderson Cancer Center, Houston, Texas; Department of Medicine, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Brahm H Segal
- Department of Medicine, University at Buffalo Jacobs School of Medicine and Biomedical Sciences, Buffalo, New York; Division of Infectious Diseases, Roswell Park Comprehensive Cancer Center, Buffalo, New York; Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, New York
| | - Bronwen E Shaw
- Center for International Blood and Bone Marrow Transplant Research, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Elizabeth J Shpall
- Cell Therapy Laboratory and Cord Blood Bank, Department of Stem Cell Transplantation and Cellular Therapy, University of Texas, MD Anderson Cancer Center, Houston, Texas
| | - Anthony D Sung
- Division of Hematologic Malignancies and Cellular Therapy, Duke University School of Medicine, Duke Cancer Institute, Durham, North Carolina
| | - Daniela Weber
- Department of Internal Medicine 3, University Medical Center, Regensburg, Germany
| | - Jennifer Whangbo
- Dana-Farber Cancer Institute, Boston Children's Hospital, Boston, Massachusetts
| | - John R Wingard
- Department of Medicine, University of Florida Health Cancer Center, Gainesville, Florida; Bone Marrow Transplant Program, Division of Hematology/Oncology, University of Florida College of Medicine, Florida
| | - William A Wood
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Miguel-Angel Perales
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York; Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Robert R Jenq
- Departments of Genomic Medicine and Stem Cell Transplantation Cellular Therapy, Division of Cancer Medicine, University of Texas MD Anderson Cancer Center, Houston, Texas.
| | - Ami S Bhatt
- Department of Genetics and Division of Hematology, Department of Medicine, Stanford University, Stanford, California.
| | | |
Collapse
|
422
|
Gonçalves P, Araújo JR, Di Santo JP. A Cross-Talk Between Microbiota-Derived Short-Chain Fatty Acids and the Host Mucosal Immune System Regulates Intestinal Homeostasis and Inflammatory Bowel Disease. Inflamm Bowel Dis 2018; 24:558-572. [PMID: 29462379 DOI: 10.1093/ibd/izx029] [Citation(s) in RCA: 274] [Impact Index Per Article: 39.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Indexed: 12/22/2022]
Abstract
Gut microbiota has a fundamental role in the energy homeostasis of the host and is essential for proper "education" of the immune system. Intestinal microbial communities are able to ferment dietary fiber releasing short-chain fatty acids (SCFAs). The SCFAs, particularly butyrate (BT), regulate innate and adaptive immune cell generation, trafficing, and function. For example, BT has an anti-inflammatory effect by inhibiting the recruitment and proinflammatory activity of neutrophils, macrophages, dendritic cells, and effector T cells and by increasing the number and activity of regulatory T cells. Gut microbial dysbiosis, ie, a microbial community imbalance, has been suggested to play a role in the development of inflammatory bowel disease (IBD). The relationship between dysbiosis and IBD has been difficult to prove, especially in humans, and is probably complex and dynamic, rather than one of a simple cause and effect relationship. However, IBD patients have dysbiosis with reduced numbers of SCFAs-producing bacteria and reduced BT concentration that is linked to a marked increase in the number of proinflammatory immune cells in the gut mucosa of these patients. Thus, microbial dysbiosis and reduced BT concentration may be a factor in the emergence and severity of IBD. Understanding the relationship between microbial dysbiosis and reduced BT concentration to IBD may lead to novel therapeutic interventions.
Collapse
Affiliation(s)
- Pedro Gonçalves
- Innate Immunity Unit, Institut Pasteur, Paris, France.,Institut National de la Santé et de la Recherche Médicale (INSERM) U1223, Paris, France
| | - João Ricardo Araújo
- Molecular Microbial Pathogenesis Unit, Institut Pasteur, Paris, France.,Institut National de la Santé et de la Recherche Médicale (INSERM) U1202, Paris, France
| | - James P Di Santo
- Institut National de la Santé et de la Recherche Médicale (INSERM) U1223, Paris, France
| |
Collapse
|
423
|
Nguyen HD, Kuril S, Bastian D, Yu XZ. T-Cell Metabolism in Hematopoietic Cell Transplantation. Front Immunol 2018; 9:176. [PMID: 29479351 PMCID: PMC5811499 DOI: 10.3389/fimmu.2018.00176] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Accepted: 01/19/2018] [Indexed: 12/11/2022] Open
Abstract
Metabolism, including catabolism and anabolism, is a basic cellular process necessary for cell survival. T lymphocytes have a distinct metabolism that can determine both fate and function. T-cell activation depends on glycolysis to obtain materials and energy for proliferation and effector function. Importantly, T cells utilize different metabolic processes under different conditions and diseases. Allogeneic hematopoietic cell transplantation (allo-HCT) is a classic immunotherapy for hematological malignancies; however, the development of graft-versus-host disease (GVHD) is a major factor limiting the success of allo-HCT. T cells in the donor graft drive GVHD by mounting a robust immunological attack against recipient normal tissues. Hence, understanding T-cell metabolism after allo-HCT would provide potential metabolic targets for the control of GVHD and primary tumor relapse. The purpose of the current review is to highlight the key metabolic pathways involved in alloantigen-activated T cells and to discuss how manipulating these pathways can serve as potential new therapeutic strategies to induce immune tolerance after allo-transplantation. We will also summarize the recent progress in regulating T-cell metabolism in bone marrow transplantation by targeting novel metabolic regulators or immune checkpoint molecules.
Collapse
Affiliation(s)
- Hung D Nguyen
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, United States
| | - Sandeepkumar Kuril
- Department of Pediatric Ematology-Oncology, Medical University of South Carolina, Charleston, SC, United States
| | - David Bastian
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, United States
| | - Xue-Zhong Yu
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, United States.,Department of Medicine, Medical University of South Carolina, Charleston, SC, United States
| |
Collapse
|
424
|
Kuo SM. Does Modification of the Large Intestinal Microbiome Contribute to the Anti-Inflammatory Activity of Fermentable Fiber? Curr Dev Nutr 2018; 2:nzx004. [PMID: 30377676 PMCID: PMC6201682 DOI: 10.3945/cdn.117.001180] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2017] [Accepted: 11/21/2017] [Indexed: 12/16/2022] Open
Abstract
Fiber is an inadequately understood and insufficiently consumed nutrient. This review examines the possible causal relation between fiber-induced microbiome changes and the anti-inflammatory activity of fiber. To demonstrate the dominant role of fermentable plant fiber in shaping the intestinal microbiome, animal and human fiber-feeding studies are reviewed. Using culture-, PCR-, and sequencing-based microbial analyses, a higher prevalence of Bifidobacterium and Lactobacillus genera was observed from the feeding of different types of fermentable fiber. This finding was reported in studies performed on several host species including human. Health conditions and medications that are linked to intestinal microbial alterations likely also change the nutrient environment of the large intestine. The unique gene clusters of Bifidobacterium and Lactobacillus that enable the catabolism of plant glycans and the ability of Bifidobacterium and Lactobacillus to reduce the colonization of proteobacteria probably contribute to their prevalence in a fiber-rich intestinal environment. The fiber-induced microbiome changes could contribute to the anti-inflammatory activity of fiber. Although most studies did not measure fecal microbial density or total daily fecal microbial output (colon microbial load), limited evidence suggests that the increase in intestinal commensal microbial load plays an important role in the anti-inflammatory activity of fiber. Various probiotic supplements, including Bifidobacterium and Lactobacillus, showed anti-inflammatory activity only in the presence of fiber, which promoted microbial growth as indicated by increasing plasma short-chain fatty acids. Probiotics alone or pure fiber administered under sterile conditions showed no anti-inflammatory activity. The potential mechanisms that could mediate the anti-inflammatory effect of common microbial metabolites are reviewed, but more in vivo trials are needed. Future studies including simultaneous microbial composition and load measurements are also important.
Collapse
Affiliation(s)
- Shiu-Ming Kuo
- Department of Exercise and Nutrition Sciences, University at Buffalo, SUNY, Buffalo, NY
| |
Collapse
|
425
|
Chaput N, Lepage P, Coutzac C, Soularue E, Le Roux K, Monot C, Boselli L, Routier E, Cassard L, Collins M, Vaysse T, Marthey L, Eggermont A, Asvatourian V, Lanoy E, Mateus C, Robert C, Carbonnel F. Baseline gut microbiota predicts clinical response and colitis in metastatic melanoma patients treated with ipilimumab. Ann Oncol 2018; 28:1368-1379. [PMID: 28368458 DOI: 10.1093/annonc/mdx108] [Citation(s) in RCA: 899] [Impact Index Per Article: 128.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Background Ipilimumab, an immune checkpoint inhibitor targeting CTLA-4, prolongs survival in a subset of patients with metastatic melanoma (MM) but can induce immune-related adverse events, including enterocolitis. We hypothesized that baseline gut microbiota could predict ipilimumab anti-tumor response and/or intestinal toxicity. Patients and methods Twenty-six patients with MM treated with ipilimumab were prospectively enrolled. Fecal microbiota composition was assessed using 16S rRNA gene sequencing at baseline and before each ipilimumab infusion. Patients were further clustered based on microbiota patterns. Peripheral blood lymphocytes immunophenotypes were studied in parallel. Results A distinct baseline gut microbiota composition was associated with both clinical response and colitis. Compared with patients whose baseline microbiota was driven by Bacteroides (cluster B, n = 10), patients whose baseline microbiota was enriched with Faecalibacterium genus and other Firmicutes (cluster A, n = 12) had longer progression-free survival (P = 0.0039) and overall survival (P = 0.051). Most of the baseline colitis-associated phylotypes were related to Firmicutes (e.g. relatives of Faecalibacterium prausnitzii and Gemmiger formicilis), whereas no colitis-related phylotypes were assigned to Bacteroidetes. A low proportion of peripheral blood regulatory T cells was associated with cluster A, long-term clinical benefit and colitis. Ipilimumab led to a higher inducible T-cell COStimulator induction on CD4+ T cells and to a higher increase in serum CD25 in patients who belonged to Faecalibacterium-driven cluster A. Conclusion Baseline gut microbiota enriched with Faecalibacterium and other Firmicutes is associated with beneficial clinical response to ipilimumab and more frequent occurrence of ipilimumab-induced colitis.
Collapse
Affiliation(s)
- N Chaput
- Laboratory of Immunomonitoring in Oncology, CNRS-UMS 3655 and INSERM-US23, Gustave Roussy Cancer Campus, Villejuif.,Faculty of Pharmacy, Chatenay-Malabry
| | - P Lepage
- Micalis Institute, INRA, AgroParisTech, Paris
| | - C Coutzac
- Laboratory of Immunomonitoring in Oncology, CNRS-UMS 3655 and INSERM-US23, Gustave Roussy Cancer Campus, Villejuif.,Faculty of Medicine, University Paris-Saclay, Le Kremlin Bicêtre
| | - E Soularue
- Laboratory of Immunomonitoring in Oncology, CNRS-UMS 3655 and INSERM-US23, Gustave Roussy Cancer Campus, Villejuif.,Faculty of Medicine, University Paris-Saclay, Le Kremlin Bicêtre.,Department of Gastroenterology, Kremlin Bicêtre Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP), Le Kremlin Bicêtre
| | - K Le Roux
- Micalis Institute, INRA, AgroParisTech, Paris
| | - C Monot
- Micalis Institute, INRA, AgroParisTech, Paris
| | - L Boselli
- Laboratory of Immunomonitoring in Oncology, CNRS-UMS 3655 and INSERM-US23, Gustave Roussy Cancer Campus, Villejuif
| | - E Routier
- Dermatology Unit, Department of Medicine, Gustave Roussy, Cancer Campus, Villejuif
| | - L Cassard
- Laboratory of Immunomonitoring in Oncology, CNRS-UMS 3655 and INSERM-US23, Gustave Roussy Cancer Campus, Villejuif
| | - M Collins
- Faculty of Medicine, University Paris-Saclay, Le Kremlin Bicêtre.,Department of Gastroenterology, Kremlin Bicêtre Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP), Le Kremlin Bicêtre
| | - T Vaysse
- Faculty of Medicine, University Paris-Saclay, Le Kremlin Bicêtre.,Department of Gastroenterology, Kremlin Bicêtre Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP), Le Kremlin Bicêtre
| | - L Marthey
- Faculty of Medicine, University Paris-Saclay, Le Kremlin Bicêtre.,Department of Gastroenterology, Kremlin Bicêtre Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP), Le Kremlin Bicêtre
| | - A Eggermont
- Dermatology Unit, Department of Medicine, Gustave Roussy, Cancer Campus, Villejuif.,INSERM U1015, Gustave Roussy, Cancer Campus, Villejuif
| | - V Asvatourian
- Biostatistics and Epidemiology Unit, Gustave Roussy Cancer Campus (GRCC), Villejuif.,University Paris-Saclay, Université Paris-Sud, UVSQ, CESP, INSERM, Villejuif, France
| | - E Lanoy
- Biostatistics and Epidemiology Unit, Gustave Roussy Cancer Campus (GRCC), Villejuif.,University Paris-Saclay, Université Paris-Sud, UVSQ, CESP, INSERM, Villejuif, France
| | - C Mateus
- Faculty of Medicine, University Paris-Saclay, Le Kremlin Bicêtre
| | - C Robert
- Faculty of Medicine, University Paris-Saclay, Le Kremlin Bicêtre.,Dermatology Unit, Department of Medicine, Gustave Roussy, Cancer Campus, Villejuif
| | - F Carbonnel
- Faculty of Medicine, University Paris-Saclay, Le Kremlin Bicêtre.,Department of Gastroenterology, Kremlin Bicêtre Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP), Le Kremlin Bicêtre
| |
Collapse
|
426
|
Chisolm DA, Weinmann AS. Connections Between Metabolism and Epigenetics in Programming Cellular Differentiation. Annu Rev Immunol 2018; 36:221-246. [PMID: 29328786 DOI: 10.1146/annurev-immunol-042617-053127] [Citation(s) in RCA: 95] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Researchers are intensifying efforts to understand the mechanisms by which changes in metabolic states influence differentiation programs. An emerging objective is to define how fluctuations in metabolites influence the epigenetic states that contribute to differentiation programs. This is because metabolites such as S-adenosylmethionine, acetyl-CoA, α-ketoglutarate, 2-hydroxyglutarate, and butyrate are donors, substrates, cofactors, and antagonists for the activities of epigenetic-modifying complexes and for epigenetic modifications. We discuss this topic from the perspective of specialized CD4+ T cells as well as effector and memory T cell differentiation programs. We also highlight findings from embryonic stem cells that give mechanistic insight into how nutrients processed through pathways such as glycolysis, glutaminolysis, and one-carbon metabolism regulate metabolite levels to influence epigenetic events and discuss similar mechanistic principles in T cells. Finally, we highlight how dysregulated environments, such as the tumor microenvironment, might alter programming events.
Collapse
Affiliation(s)
- Danielle A Chisolm
- Department of Microbiology, University of Alabama at Birmingham, Alabama 35294, USA; ,
| | - Amy S Weinmann
- Department of Microbiology, University of Alabama at Birmingham, Alabama 35294, USA; ,
| |
Collapse
|
427
|
Bonvalet M, Daillère R, Roberti MP, Rauber C, Zitvogel L. The Impact of the Intestinal Microbiota in Therapeutic Responses Against Cancer. Oncoimmunology 2018. [DOI: 10.1007/978-3-319-62431-0_27] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
|
428
|
Riwes M, Reddy P. Microbial metabolites and graft versus host disease. Am J Transplant 2018; 18:23-29. [PMID: 28742948 DOI: 10.1111/ajt.14443] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Revised: 07/04/2017] [Accepted: 07/08/2017] [Indexed: 01/25/2023]
Abstract
The health of mammals is a reflection of the diversity and composition of the intestinal microbiota. Alterations in the composition and functions of the intestinal microbiota have been implicated in multiple disease processes. The impact of the microbiota in health and disease is in part a function of the nutrient processing and release of metabolites. Recent studies have uncovered a major role for microbial metabolites in the function of the host immune system by which they influence disease processes such as acute graft versus host disease (GVHD), which is the main complication of allogeneic hematopoietic cell transplantation (allo-HCT). The mechanisms of acute GVHD regulation by the complex microbial community and the metabolites released by them are unclear. In this review we summarize major findings of how microbial metabolites interact with the immune system and discuss how these interactions could impact acute GVHD.
Collapse
Affiliation(s)
- M Riwes
- Blood and Marrow Transplant Program, Division of Hematology/Oncology, Department of Internal Medicine, University of Michigan Comprehensive Cancer Center, Ann Arbor, MI, USA
| | - P Reddy
- Blood and Marrow Transplant Program, Division of Hematology/Oncology, Department of Internal Medicine, University of Michigan Comprehensive Cancer Center, Ann Arbor, MI, USA
| |
Collapse
|
429
|
Shallis RM, Terry CM, Lim SH. Changes in intestinal microbiota and their effects on allogeneic stem cell transplantation. Am J Hematol 2018; 93:122-128. [PMID: 28842931 DOI: 10.1002/ajh.24896] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Revised: 07/31/2017] [Accepted: 08/22/2017] [Indexed: 12/30/2022]
Abstract
The human intestinal microbiota is essential for microbial homeostasis, regulation of metabolism, and intestinal immune tolerance. Rapidly evolving understanding of the importance of the microbiota implicates changes in the composition and function of intestinal microbial communities in an assortment of systemic conditions. Complications following allogeneic stem cell transplant now join the ever-expanding list of pathologic states regulated by intestinal microbiota. Dysbiosis, or disruption of the normal ecology of this microbiome, has been directly implicated in the pathogenesis of entities such as Clostridium difficile infections, graft-versus-host disease (GVHD), and most recently disease relapse, all of which are major causes of morbidity and mortality in patients undergoing allogeneic stem cell transplant. In this review, we elucidate the key origins of microbiotic alterations and discuss how dysbiosis influences complications following allogeneic stem cell transplant. Our emerging understanding of the importance of a balanced and diverse intestinal microbiota is prompting investigation into the appropriate treatment of dysbiosis, reliable and early detection of such, and ultimately its prevention in patients to improve the outcome following allogeneic hematopoietic stem cell transplant.
Collapse
Affiliation(s)
- Rory M. Shallis
- Division of Hematology and Oncology; Rhode Island Hospital/Brown University Warren Alpert School of Medicine; Providence Rhode Island
| | - Christopher M. Terry
- Division of Hematology and Oncology; Rhode Island Hospital/Brown University Warren Alpert School of Medicine; Providence Rhode Island
| | - Seah H. Lim
- Division of Hematology and Oncology; Rhode Island Hospital/Brown University Warren Alpert School of Medicine; Providence Rhode Island
| |
Collapse
|
430
|
Reddy P, Ferrara JL. Graft-Versus-Host Disease and Graft-Versus-Leukemia Responses. Hematology 2018. [DOI: 10.1016/b978-0-323-35762-3.00108-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
|
431
|
Naymagon S, Naymagon L, Wong SY, Ko HM, Renteria A, Levine J, Colombel JF, Ferrara J. Acute graft-versus-host disease of the gut: considerations for the gastroenterologist. Nat Rev Gastroenterol Hepatol 2017; 14:711-726. [PMID: 28951581 PMCID: PMC6240460 DOI: 10.1038/nrgastro.2017.126] [Citation(s) in RCA: 109] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Haematopoietic stem cell transplantation (HSCT) is central to the management of many haematological disorders. A frequent complication of HSCT is acute graft-versus-host disease (GVHD), a condition in which immune cells from the donor attack healthy recipient tissues. The gastrointestinal system is among the most common sites affected by acute GVHD, and severe manifestations of acute GVHD of the gut portends a poor prognosis in patients after HSCT. Acute GVHD of the gastrointestinal tract presents both diagnostic and therapeutic challenges. Although the clinical manifestations are nonspecific and overlap with those of infection and drug toxicity, diagnosis is ultimately based on clinical criteria. As reliable serum biomarkers have not yet been validated outside of clinical trials, endoscopic and histopathological evaluation continue to be utilized in diagnosis. Once a diagnosis of gastrointestinal acute GVHD is established, therapy with systemic corticosteroids is typically initiated, and non-responders can be treated with a wide range of second-line therapies. In addition to treating the underlying disease, the management of complications including profuse diarrhoea, severe malnutrition and gastrointestinal bleeding is paramount. In this Review, we discuss strategies for the diagnosis and management of acute GVHD of the gastrointestinal tract as they pertain to the practising gastroenterologist.
Collapse
Affiliation(s)
- Steven Naymagon
- Henry D. Janowitz Division of Gastroenterology, Icahn School of Medicine at Mount Sinai
| | - Leonard Naymagon
- Division of Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai
| | - Serre-Yu Wong
- Henry D. Janowitz Division of Gastroenterology, Icahn School of Medicine at Mount Sinai
| | - Huaibin Mabel Ko
- Henry D. Janowitz Division of Gastroenterology, Icahn School of Medicine at Mount Sinai.,Lillian and Henry M. Stratton-Hans Popper Department of Pathology and Laboratory Medicine, Icahn School of Medicine at Mount Sinai, One Gustave Levy Place, New York, New York 10029, USA
| | - Anne Renteria
- Division of Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai
| | - John Levine
- Division of Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai
| | | | - James Ferrara
- Division of Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai
| |
Collapse
|
432
|
Affiliation(s)
- Robert Zeiser
- From the Department of Hematology, Oncology, and Stem Cell Transplantation, Faculty of Medicine, Freiburg University Medical Center, Freiburg, Germany (R.Z.); and the Department of Pediatrics, Division of Blood and Marrow Transplantation, University of Minnesota, Minneapolis (B.R.B.)
| | - Bruce R Blazar
- From the Department of Hematology, Oncology, and Stem Cell Transplantation, Faculty of Medicine, Freiburg University Medical Center, Freiburg, Germany (R.Z.); and the Department of Pediatrics, Division of Blood and Marrow Transplantation, University of Minnesota, Minneapolis (B.R.B.)
| |
Collapse
|
433
|
Abstract
PURPOSE OF REVIEW The purpose of this review is to summarize recent advances in our understanding of the complex immunology of intestinal transplantation and allograft rejection. RECENT FINDINGS Recent findings highlight the importance of the intestinal microbiome for intestinal homeostasis and the role of newly discovered innate lymphoid cells in intestinal transplantation. In addition, the role of host antimicrobial Th17 responses in the pathogenesis of inflammatory bowel diseases and intestinal allograft rejection has been further elucidated. SUMMARY Research on the complex immune system of the intestine has continued to reveal more intricacies and connections with each study performed, making treatment of intestinal transplant patients more multifaceted. The interaction, communication, and relationships between areas such as the microbiome, innate lymphoid cells, and Th17 cells reveal possible targets for therapy and further areas requiring ongoing research.
Collapse
|
434
|
Farowski F, Bücker V, Vehreschild JJ, Biehl L, Cruz-Aguilar R, Scheid C, Holtick U, Jazmati N, Wisplinghoff H, Cornely OA, Vehreschild MJGT. Impact of choice, timing, sequence and combination of broad-spectrum antibiotics on the outcome of allogeneic haematopoietic stem cell transplantation. Bone Marrow Transplant 2017; 53:52-57. [PMID: 29131156 DOI: 10.1038/bmt.2017.203] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Revised: 08/02/2017] [Accepted: 08/02/2017] [Indexed: 02/02/2023]
Abstract
Recent data link the incidence of intestinal GvHD (iGvHD) after allogeneic haematopoietic stem cell transplantation (aSCT) to exposure with piperacillin-tazobactam or imipenem-cilastatin. To assess relevance of timing, duration, sequence and combination of antibiotic treatment in this setting, we applied a time-dependent model to our aSCT cohort. Patients from the prospective Cologne Cohort of Neutropenic Patients (CoCoNut) undergoing aSCT from January 2007 to April 2013 were included into a time-dependent multivariate Cox proportional hazards regression model with backward-stepwise selection. In 399 eligible patients, cumulative antibiotic exposure (hazard ratio (HR) 2.46; 95% confidence interval (95% CI) 1.59-3.81; P<0.001) and exposure to sequential treatment with penicillin derivatives and carbapenems (HR 6.22, 95% CI 1.27-30.31), but not to the individual classes, were associated with iGvHD at day 100. Glycopeptides were assessed as a risk factor (HR 3.73, 95% CI 1.51-9.19), but not considered independent, since their use was dependent on previous exposure to penicillin derivatives and carbapenems. Patients with iGvHD presented with increased non-relapse mortality at day 365 (HR 3.51; 95% CI 2.10-5.89; P<0.001). We identified sequential exposure to penicillin derivatives and carbapenems as well as overall exposure to antibiotics as independent risk factors for iGVHD. Confirmation of these findings in larger, prospective cohorts is necessary.
Collapse
Affiliation(s)
- F Farowski
- Department I of Internal Medicine, University Hospital of Cologne, Cologne, Germany.,German Centre for Infection Research (DZIF), Partner Site Bonn-Cologne, Cologne, Germany
| | - V Bücker
- Department I of Internal Medicine, University Hospital of Cologne, Cologne, Germany
| | - J J Vehreschild
- Department I of Internal Medicine, University Hospital of Cologne, Cologne, Germany.,German Centre for Infection Research (DZIF), Partner Site Bonn-Cologne, Cologne, Germany
| | - L Biehl
- Department I of Internal Medicine, University Hospital of Cologne, Cologne, Germany.,German Centre for Infection Research (DZIF), Partner Site Bonn-Cologne, Cologne, Germany
| | - R Cruz-Aguilar
- Department I of Internal Medicine, University Hospital of Cologne, Cologne, Germany.,German Centre for Infection Research (DZIF), Partner Site Bonn-Cologne, Cologne, Germany
| | - C Scheid
- Department I of Internal Medicine, University Hospital of Cologne, Cologne, Germany
| | - U Holtick
- Department I of Internal Medicine, University Hospital of Cologne, Cologne, Germany
| | - N Jazmati
- German Centre for Infection Research (DZIF), Partner Site Bonn-Cologne, Cologne, Germany.,Institute for Medical Microbiology, Immunology and Hygiene, University Hospital of Cologne, Cologne, Germany
| | - H Wisplinghoff
- Institute for Medical Microbiology, Immunology and Hygiene, University Hospital of Cologne, Cologne, Germany
| | - O A Cornely
- Department I of Internal Medicine, University Hospital of Cologne, Cologne, Germany.,German Centre for Infection Research (DZIF), Partner Site Bonn-Cologne, Cologne, Germany.,Clinical Trials Centre Cologne, ZKS Köln, Cologne, Germany.,Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - M J G T Vehreschild
- Department I of Internal Medicine, University Hospital of Cologne, Cologne, Germany.,German Centre for Infection Research (DZIF), Partner Site Bonn-Cologne, Cologne, Germany
| |
Collapse
|
435
|
Perkey E, Maillard I. New Insights into Graft-Versus-Host Disease and Graft Rejection. ANNUAL REVIEW OF PATHOLOGY-MECHANISMS OF DISEASE 2017; 13:219-245. [PMID: 29099650 DOI: 10.1146/annurev-pathol-020117-043720] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Allogeneic transplantation of foreign organs or tissues has lifesaving potential, but can lead to serious complications. After solid organ transplantation, immune-mediated rejection mandates the use of prolonged global immunosuppression and limits the life span of transplanted allografts. After bone marrow transplantation, donor-derived immune cells can trigger life-threatening graft-versus-host disease. T cells are central mediators of alloimmune complications and the target of most existing therapeutic interventions. We review recent progress in identifying multiple cell types in addition to T cells and new molecular pathways that regulate pathogenic alloreactivity. Key discoveries include the cellular subsets that function as potential sources of alloantigens, the cross talk of innate lymphoid cells with damaged epithelia and with the recipient microbiome, the impact of the alarmin interleukin-33 on alloreactivity, and the role of Notch ligands expressed by fibroblastic stromal cells in alloimmunity. While refining our understanding of transplantation immunobiology, these findings identify new therapeutic targets and new areas of investigation.
Collapse
Affiliation(s)
- Eric Perkey
- Life Sciences Institute, University of Michigan, Ann Arbor, Michigan 48109, USA;
| | - Ivan Maillard
- Life Sciences Institute, University of Michigan, Ann Arbor, Michigan 48109, USA; .,Department of Internal Medicine, Division of Hematology-Oncology, and Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, Michigan 48109, USA.,Department of Medicine, Division of Hematology-Oncology, and Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA;
| |
Collapse
|
436
|
Bull JMC. A review of immune therapy in cancer and a question: can thermal therapy increase tumor response? Int J Hyperthermia 2017; 34:840-852. [PMID: 28974121 DOI: 10.1080/02656736.2017.1387938] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Immune therapy is a successful cancer treatment coming into its own. This is because checkpoint molecules, adoptive specific lymphocyte transfer and chimeric antigen T-cell (CAR-T) therapy are able to induce more durable responses in an increasing number of malignancies compared to chemotherapy. In addition, immune therapies are able to treat bulky disease, whereas standard cytotoxic therapies cannot treat large tumour burdens. Checkpoint inhibitor monoclonal antibodies are becoming widely used in the clinic and although more complex, adoptive lymphocyte transfer and CAR-T therapies show promise. We are learning that there are nuances to predicting the successful use of the checkpoint inhibitors as well as to specific-antigen adoptive and CAR-T therapies. We are also newly aware of a here-to-fore unrealised natural force, the status of the microbiome. However, despite better understanding of mechanisms of action of the new immune therapies, the best responses to the new immune therapies remain 20-30%. Likely the best way to improve this somewhat low response rate for patients is to increase the patient's own immune response. Thermal therapy is a way to do this. All forms of thermal therapy, from fever-range systemic thermal therapy, to high-temperature HIFU and even cryotherapy improve the immune response pre-clinically. It is time to test the immune therapies with thermal therapy in vivo to test for optimal timing of the combinations that will best enhance tumour response and then to begin to test the immune therapies with thermal therapy in the clinic as soon as possible.
Collapse
Affiliation(s)
- Joan M C Bull
- a Division of Oncology, Department of Internal Medicine , The University of Texas Medical School at Houston , Houston , TX , USA
| |
Collapse
|
437
|
Hayase E, Hashimoto D, Nakamura K, Noizat C, Ogasawara R, Takahashi S, Ohigashi H, Yokoi Y, Sugimoto R, Matsuoka S, Ara T, Yokoyama E, Yamakawa T, Ebata K, Kondo T, Hiramine R, Aizawa T, Ogura Y, Hayashi T, Mori H, Kurokawa K, Tomizuka K, Ayabe T, Teshima T. R-Spondin1 expands Paneth cells and prevents dysbiosis induced by graft-versus-host disease. J Exp Med 2017; 214:3507-3518. [PMID: 29066578 PMCID: PMC5716036 DOI: 10.1084/jem.20170418] [Citation(s) in RCA: 86] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2017] [Revised: 07/09/2017] [Accepted: 09/07/2017] [Indexed: 12/18/2022] Open
Abstract
The intestinal microbial ecosystem is actively regulated by Paneth cell-derived antimicrobial peptides such as α-defensins. Various disorders, including graft-versus-host disease (GVHD), disrupt Paneth cell functions, resulting in unfavorably altered intestinal microbiota (dysbiosis), which further accelerates the underlying diseases. Current strategies to restore the gut ecosystem are bacteriotherapy such as fecal microbiota transplantation and probiotics, and no physiological approach has been developed so far. In this study, we demonstrate a novel approach to restore gut microbial ecology by Wnt agonist R-Spondin1 (R-Spo1) or recombinant α-defensin in mice. R-Spo1 stimulates intestinal stem cells to differentiate to Paneth cells and enhances luminal secretion of α-defensins. Administration of R-Spo1 or recombinant α-defensin prevents GVHD-mediated dysbiosis, thus representing a novel and physiological approach at modifying the gut ecosystem to restore intestinal homeostasis and host-microbiota cross talk toward therapeutic benefits.
Collapse
Affiliation(s)
- Eiko Hayase
- Department of Hematology, Faculty of Medicine, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Daigo Hashimoto
- Department of Hematology, Faculty of Medicine, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Kiminori Nakamura
- Department of Cell Biological Science, Graduate School of Life Science, Faculty of Advanced Life Science, Hokkaido University, Sapporo, Japan
| | - Clara Noizat
- Department of Hematology, Faculty of Medicine, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Reiki Ogasawara
- Department of Hematology, Faculty of Medicine, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Shuichiro Takahashi
- Department of Hematology, Faculty of Medicine, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Hiroyuki Ohigashi
- Department of Hematology, Faculty of Medicine, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Yuki Yokoi
- Innate Immunity Laboratory, Graduate School of Life Science, Hokkaido University, Sapporo, Japan
| | - Rina Sugimoto
- Innate Immunity Laboratory, Graduate School of Life Science, Hokkaido University, Sapporo, Japan
| | - Satomi Matsuoka
- Department of Hematology, Faculty of Medicine, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Takahide Ara
- Department of Hematology, Faculty of Medicine, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Emi Yokoyama
- Department of Hematology, Faculty of Medicine, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Tomohiro Yamakawa
- Department of Hematology, Faculty of Medicine, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Ko Ebata
- Department of Hematology, Faculty of Medicine, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Takeshi Kondo
- Department of Hematology, Faculty of Medicine, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Rina Hiramine
- Department of Protein Science Laboratory, Graduate School of Life Science, Hokkaido University, Sapporo, Japan.,Global Station for Soft Matter, Global Institution for Collaborative Research and Education, Hokkaido University, Sapporo, Japan
| | - Tomoyasu Aizawa
- Department of Protein Science Laboratory, Graduate School of Life Science, Hokkaido University, Sapporo, Japan.,Global Station for Soft Matter, Global Institution for Collaborative Research and Education, Hokkaido University, Sapporo, Japan
| | - Yoshitoshi Ogura
- Department of Bacteriology, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Tetsuya Hayashi
- Department of Bacteriology, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Hiroshi Mori
- Center for Information Biology, National Institute of Genetics, Mishima, Japan
| | - Ken Kurokawa
- Center for Information Biology, National Institute of Genetics, Mishima, Japan.,Department of Life Science and Technology, Tokyo Institute of Technology, Tokyo, Japan
| | - Kazuma Tomizuka
- Innovative Technology Labs, Research Functions Unit, Research & Development Division, Kyowa Hakko Kirin, Tokyo, Japan
| | - Tokiyoshi Ayabe
- Department of Cell Biological Science, Graduate School of Life Science, Faculty of Advanced Life Science, Hokkaido University, Sapporo, Japan
| | - Takanori Teshima
- Department of Hematology, Faculty of Medicine, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| |
Collapse
|
438
|
Bisaccia J, Soudja S. [Microbiome-derived butyrate alleviates intestinal graft versus host reaction]. Med Sci (Paris) 2017; 33:862-864. [PMID: 28994381 DOI: 10.1051/medsci/20173310016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Affiliation(s)
- Joseph Bisaccia
- Master de cancérologie, module d'immunologie - virologie, université de Lyon, université Claude Bernard Lyon 1, Lyon, France
| | - Saidi Soudja
- Centre de recherches en cancérologie, UMR Inserm 1052, CNRS 5286, Centre Léon Bérard, Lyon, France
| |
Collapse
|
439
|
Indoxyl 3-sulfate inhibits maturation and activation of human monocyte-derived dendritic cells. Immunobiology 2017; 223:239-245. [PMID: 29100619 DOI: 10.1016/j.imbio.2017.10.014] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Revised: 09/19/2017] [Accepted: 10/03/2017] [Indexed: 01/01/2023]
Abstract
Indole is produced from l-tryptophan by commensal bacteria and further metabolized to indoxyl 3-sulfate (I3S) in the liver. Physiologic concentrations of I3S are related to a lower risk to develop graft versus host disease in allogeneic stem cell transplanted patients pointing towards an immunoregulatory function of I3S. Here we investigated the impact of I3S on the maturation of human monocyte-derived dendritic cells (DCs). Even pathophysiologic concentrations of I3S did not affect viability of mature DCs, but I3S decreased the expression of co-stimulatory molecules such as CD80 and CD86 on mature DCs. Furthermore, I3S inhibited IL-12 and IL-6 secretion by mature DCs while IL-10 was significantly upregulated. Co-culture of I3S-treated mature DCs with allogeneic T cells revealed no alteration in T cell proliferation. However, interferon gamma and TNF production of T cells was suppressed. As I3S exerted no direct effect on T cells, the defect in T cell activation was mediated by I3S-treated mature DCs. Our study suggests an anti-inflammatory and tolerizing effect of I3S on human DCs.
Collapse
|
440
|
Mancini N, Greco R, Pasciuta R, Barbanti MC, Pini G, Morrow OB, Morelli M, Vago L, Clementi N, Giglio F, Lupo Stanghellini MT, Forcina A, Infurnari L, Marktel S, Assanelli A, Carrabba M, Bernardi M, Corti C, Burioni R, Peccatori J, Sormani MP, Banfi G, Ciceri F, Clementi M. Enteric Microbiome Markers as Early Predictors of Clinical Outcome in Allogeneic Hematopoietic Stem Cell Transplant: Results of a Prospective Study in Adult Patients. Open Forum Infect Dis 2017; 4:ofx215. [PMID: 29226172 PMCID: PMC5714175 DOI: 10.1093/ofid/ofx215] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2017] [Accepted: 09/28/2017] [Indexed: 12/18/2022] Open
Abstract
Background Infections and graft-vs-host disease (GvHD) still represent major, not easily predictable complications in allogeneic hematopoietic stem cell transplant (allo-HSCT). Both conditions have been correlated to altered enteric microbiome profiles during the peritransplant period. The main objective of this study was to identify possible early microbiome-based markers useful in pretransplant risk stratification. Methods Stool samples were collected from 96 consecutive patients at the beginning of the pretransplant conditioning regimen (T0) and at 10 (T1) and 30 (T2) days following transplant. When significant in univariate analysis, the identified microbiome markers were used in multivariate regression analyses, together with other significant clinical variables for allo-HSCT-related risk stratification. Four main outcomes were addressed: (1) septic complications, (2) GvHD, (3) relapse of the underlying disease, and (4) mortality. Results The presence of >5% proinflammatory Enterobacteriaceae at T0 was the only significant marker for the risk of microbiologically confirmed sepsis. Moreover, ≤10% Lachnospiraceae at T0 was the only significant factor for increased risk of overall mortality, including death from both infectious and noninfectious causes.Finally, a low bacterial alpha-diversity (Shannon index ≤ 1.3) at T1 was the only variable significantly correlating with an increased risk of GvHD within 30 days. Conclusions Microbiome markers can be useful in the very early identification of patients at risk for major transplant-related complications, offering new tools for individualized preemptive or therapeutic strategies to improve allo-HSCT outcomes.
Collapse
Affiliation(s)
- Nicasio Mancini
- Laboratory of Microbiology and Virology, San Raffaele Scientific Institute, Milan, Italy.,Department of Microbiology and Virology, University "Vita-Salute" San Raffaele, Milan, Italy
| | - Raffaella Greco
- Hematology and Bone Marrow Transplantation Unit, Division of Regenerative Medicine, Stem Cells and Gene Therapy, San Raffaele Scientific Institute, Milan, Italy
| | - Renée Pasciuta
- Laboratory of Microbiology and Virology, San Raffaele Scientific Institute, Milan, Italy
| | - Maria Chiara Barbanti
- Hematology and Bone Marrow Transplantation Unit, Division of Regenerative Medicine, Stem Cells and Gene Therapy, San Raffaele Scientific Institute, Milan, Italy
| | - Giacomo Pini
- Laboratory of Microbiology and Virology, San Raffaele Scientific Institute, Milan, Italy
| | - Olivia Beatrice Morrow
- Laboratory of Microbiology and Virology, San Raffaele Scientific Institute, Milan, Italy
| | - Mara Morelli
- Hematology and Bone Marrow Transplantation Unit, Division of Regenerative Medicine, Stem Cells and Gene Therapy, San Raffaele Scientific Institute, Milan, Italy
| | - Luca Vago
- Hematology and Bone Marrow Transplantation Unit, Division of Regenerative Medicine, Stem Cells and Gene Therapy, San Raffaele Scientific Institute, Milan, Italy
| | - Nicola Clementi
- Department of Microbiology and Virology, University "Vita-Salute" San Raffaele, Milan, Italy
| | - Fabio Giglio
- Hematology and Bone Marrow Transplantation Unit, Division of Regenerative Medicine, Stem Cells and Gene Therapy, San Raffaele Scientific Institute, Milan, Italy
| | - Maria Teresa Lupo Stanghellini
- Hematology and Bone Marrow Transplantation Unit, Division of Regenerative Medicine, Stem Cells and Gene Therapy, San Raffaele Scientific Institute, Milan, Italy
| | - Alessandra Forcina
- Hematology and Bone Marrow Transplantation Unit, Division of Regenerative Medicine, Stem Cells and Gene Therapy, San Raffaele Scientific Institute, Milan, Italy
| | - Laura Infurnari
- Laboratory of Microbiology and Virology, San Raffaele Scientific Institute, Milan, Italy
| | - Sarah Marktel
- Hematology and Bone Marrow Transplantation Unit, Division of Regenerative Medicine, Stem Cells and Gene Therapy, San Raffaele Scientific Institute, Milan, Italy
| | - Andrea Assanelli
- Hematology and Bone Marrow Transplantation Unit, Division of Regenerative Medicine, Stem Cells and Gene Therapy, San Raffaele Scientific Institute, Milan, Italy
| | - Matteo Carrabba
- Hematology and Bone Marrow Transplantation Unit, Division of Regenerative Medicine, Stem Cells and Gene Therapy, San Raffaele Scientific Institute, Milan, Italy
| | - Massimo Bernardi
- Hematology and Bone Marrow Transplantation Unit, Division of Regenerative Medicine, Stem Cells and Gene Therapy, San Raffaele Scientific Institute, Milan, Italy
| | - Consuelo Corti
- Hematology and Bone Marrow Transplantation Unit, Division of Regenerative Medicine, Stem Cells and Gene Therapy, San Raffaele Scientific Institute, Milan, Italy
| | - Roberto Burioni
- Laboratory of Microbiology and Virology, San Raffaele Scientific Institute, Milan, Italy.,Department of Microbiology and Virology, University "Vita-Salute" San Raffaele, Milan, Italy
| | - Jacopo Peccatori
- Hematology and Bone Marrow Transplantation Unit, Division of Regenerative Medicine, Stem Cells and Gene Therapy, San Raffaele Scientific Institute, Milan, Italy
| | | | - Giuseppe Banfi
- Galeazzi Scientific Institute and University "Vita-Salute" San Raffaele, Milan, Italy
| | - Fabio Ciceri
- Hematology and Bone Marrow Transplantation Unit, Division of Regenerative Medicine, Stem Cells and Gene Therapy, San Raffaele Scientific Institute, Milan, Italy.,Department of Hematology and Bone Marrow Transplantation, University "Vita-Salute" San Raffaele, Milan, Italy
| | - Massimo Clementi
- Laboratory of Microbiology and Virology, San Raffaele Scientific Institute, Milan, Italy.,Department of Microbiology and Virology, University "Vita-Salute" San Raffaele, Milan, Italy
| |
Collapse
|
441
|
Associations between acute gastrointestinal GvHD and the baseline gut microbiota of allogeneic hematopoietic stem cell transplant recipients and donors. Bone Marrow Transplant 2017; 52:1643-1650. [PMID: 28967895 DOI: 10.1038/bmt.2017.200] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2017] [Revised: 07/28/2017] [Accepted: 08/04/2017] [Indexed: 12/13/2022]
Abstract
Growing evidence suggests that host-microbiota interactions influence GvHD risk following allogeneic hematopoietic stem cell transplant. However, little is known about the influence of the transplant recipient's pre-conditioning microbiota nor the influence of the transplant donor's microbiota. Our study examines associations between acute gastrointestinal GvHD (agGvHD) and 16S rRNA fecal bacterial profiles in a prospective cohort of N=57 recipients before preparative conditioning, as well as N=22 of their paired HLA-matched sibling donors. On average, recipients had lower fecal bacterial diversity (P=0.0002) and different phylogenetic membership (UniFrac P=0.001) than the healthy transplant donors. Recipients with lower phylogenetic diversity had higher overall mortality rates (hazard ratio=0.37, P=0.008), but no statistically significant difference in agGvHD risk. In contrast, high bacterial donor diversity was associated with decreased agGvHD risk (odds ratio=0.12, P=0.038). Further investigation is warranted as to whether selection of hematopoietic stem cell transplant donors with high gut microbiota diversity and/or other specific compositional attributes may reduce agGvHD incidence, and by what mechanisms.
Collapse
|
442
|
High incidence of extensive chronic graft-versus-host disease in patients with the REG3A rs7588571 non-GG genotype. PLoS One 2017; 12:e0185213. [PMID: 28945764 PMCID: PMC5612655 DOI: 10.1371/journal.pone.0185213] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2017] [Accepted: 09/10/2017] [Indexed: 01/06/2023] Open
Abstract
Regenerating islet-derived protein 3 alpha (REG3A) is a biomarker of lower gastrointestinal graft-versus-host disease (GVHD); however, the biological role of REG3A in the pathophysiology of GVHD is not understood. Here, we examined the association between a single nucleotide polymorphism in the REG3A gene, rs7588571, which is located upstream and within 2 kb of the REG3A gene, and transplant outcomes including the incidence of GVHD. The study population consisted of 126 adult Japanese patients who had undergone bone marrow transplantation from a HLA-matched sibling. There was no association between rs7588571 polymorphism and the incidence of acute GVHD. However, a significantly higher incidence of extensive chronic GVHD was observed in patients with the rs7588571 non-GG genotype than in those with the GG genotype (Odds ratio 2.6; 95% confidence interval, 1.1–6.0; P = 0.029). Semi-quantitative reverse transcription PCR demonstrated that the rs7588571 non-GG genotype exhibited a significantly lower REG3A mRNA expression level than the GG genotype (P = 0.032), and Western blot analysis demonstrated that the rs7588571 non-GG genotype exhibited a trend toward lower REG3A protein expression level than the GG genotype (P = 0.053). Since REG proteins have several activities that function to control intestinal microbiota, and since intestinal dysbiosis is in part responsible for the development of GVHD, our findings lead to the novel concept that REG3A could have some protective effect in the pathogenesis of GVHD through the regulation of gut microbiota.
Collapse
|
443
|
Short chain fatty acids ameliorate immune-mediated uveitis partially by altering migration of lymphocytes from the intestine. Sci Rep 2017; 7:11745. [PMID: 28924192 PMCID: PMC5603543 DOI: 10.1038/s41598-017-12163-3] [Citation(s) in RCA: 86] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Accepted: 09/04/2017] [Indexed: 02/07/2023] Open
Abstract
Short chain fatty acids (SCFA) are metabolites of intestinal bacteria resulting from fermentation of dietary fiber. SCFA are protective in various animal models of inflammatory disease. We investigated the effects of exogenous administration of SFCAs, particularly propionate, on uveitis using an inducible model of experimental autoimmune uveitis (EAU). Oral SCFA administration attenuated uveitis severity in a mouse strain-dependent manner through regulatory T cell induction among lymphocytes in the intestinal lamina propria (LPL) and cervical lymph nodes (CLN). SCFA also suppressed effector T cell induction in the CLN and mesenteric lymph nodes (MLN). Alterations in intestinal morphology and gene expression demonstrated in the EAU model prior to the onset of uveitis were blunted by oral SCFA administration. Using a Kaede transgenic mouse, we demonstrated enhanced leukocyte trafficking between the intestine and the eye in EAU. Propionate suppressed T effector cell migration between the intestine and the spleen in EAU Kaede mice. In conclusion, our findings support exogenous administration of SCFAs as a potential treatment strategy for uveitis through the stabilization of subclinical intestinal alterations that occur in inflammatory diseases including uveitis, as well as prevention of trafficking of leukocytes between the gastrointestinal tract and extra-intestinal tissues.
Collapse
|
444
|
Shashikanth N, Yeruva S, Ong MLDM, Odenwald MA, Pavlyuk R, Turner JR. Epithelial Organization: The Gut and Beyond. Compr Physiol 2017; 7:1497-1518. [DOI: 10.1002/cphy.c170003] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
445
|
Bai L, Zhou P, Li D, Ju X. Changes in the gastrointestinal microbiota of children with acute lymphoblastic leukaemia and its association with antibiotics in the short term. J Med Microbiol 2017; 66:1297-1307. [PMID: 28855025 DOI: 10.1099/jmm.0.000568] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Purpose. To detect the alteration of gut bacteria in children with ALL and analyse the impact of short-term-use of antibiotics on the changes caused by ALL.Methodology. We collected faecal samples from both children with ALL and healthy children. According to their medication history with antibiotics, we classified the samples into ALL+ATBx, ALL, CON+ATBx and CON groups. Next-generation sequencing was performed to identify the gut bacteria according to the MiSeq platform. The Shannon index, Simpson index, Chao index and Ace index were used to represent the alpha diversity of gut bacteria. The beta diversity was estimated using the principles of co-ordinate analysis and non-metric multi-dimensional scaling. The taxon composition and presence of biomarkers were then determined through bioinformatics.Results. With regard to alpha diversity, the Shannon index and Simpson index differed significantly between the ALL and CON groups, as well as the CON+ATBx and CON groups, but not the ALL+ATBx and CON+ATBx groups. With regard to beta diversity, the ALL and CON separated clearly into clusters, as did ALL+ATBx and CON+ATBx. There were differences in composition among the four groups at different taxonomy hierarchies. More bacteria showed an obvious difference between the paired groups ALL and CON than did for the paired groups ALL+ATBx and CON+ATBx. The area under the receiver operating characteristic curves for Bacteroidales and Enterococcaceae used to predict ALL were 0.735 and 0.724, respectively.Conclusion. ALL induced structural changes of the gut microbiota, with the alpha diversity being significantly weakened by antibiotics, but not beta diversity. Bacteroidales and Enterococcaceae can be referred to as biomarkers for ALL.
Collapse
Affiliation(s)
- Lu Bai
- Department of Pediatrics, Qilu Hospital of Shandong University, Jinan 250012, PR China
| | - Panpan Zhou
- Department of Pediatrics, Qilu Hospital of Shandong University, Jinan 250012, PR China
| | - Dong Li
- Cryomedicine Lab, Qilu Hospital of Shandong University, Jinan 250012, PR China
| | - Xiuli Ju
- Department of Pediatrics, Qilu Hospital of Shandong University, Jinan 250012, PR China
| |
Collapse
|
446
|
Unraveling the gut microbiome of the long-lived naked mole-rat. Sci Rep 2017; 7:9590. [PMID: 28852094 PMCID: PMC5575099 DOI: 10.1038/s41598-017-10287-0] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Accepted: 08/07/2017] [Indexed: 01/16/2023] Open
Abstract
The naked mole-rat (Heterocephalus glaber) is a subterranean mouse-sized African mammal that shows astonishingly few age-related degenerative changes and seems to not be affected by cancer. These features make this wild rodent an excellent model to study the biology of healthy aging and longevity. Here we characterize for the first time the intestinal microbial ecosystem of the naked mole-rat in comparison to humans and other mammals, highlighting peculiarities related to the specific living environment, such as the enrichment in bacteria able to utilize soil sulfate as a terminal electron acceptor to sustain an anaerobic oxidative metabolism. Interestingly, some compositional gut microbiota peculiarities were also shared with human gut microbial ecosystems of centenarians and Hadza hunter-gatherers, considered as models of a healthy gut microbiome and of a homeostatic and highly adaptive gut microbiota-host relationship, respectively. In addition, we found an enrichment of short-chain fatty acids and carbohydrate degradation products in naked mole-rat compared to human samples. These data confirm the importance of the gut microbial ecosystem as an adaptive partner for the mammalian biology and health, independently of the host phylogeny.
Collapse
|
447
|
Daguindau E, Gautier T, Chagué C, Pais de Barros JP, Deckert V, Lagrost L, Saas P. Is It Time to Reconsider the Lipopolysaccharide Paradigm in Acute Graft-Versus-Host Disease? Front Immunol 2017; 8:952. [PMID: 28848554 PMCID: PMC5553011 DOI: 10.3389/fimmu.2017.00952] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Accepted: 07/25/2017] [Indexed: 12/16/2022] Open
Affiliation(s)
- Etienne Daguindau
- Université Bourgogne Franche-Comté, EFS Bourgogne Franche-Comté, INSERM, UMR1098, Besançon, France
- Department of Hematology, University Hospital of Besançon, Besançon, France
- LipSTIC LabEx, FHU INCREASE, Besançon, France
| | - Thomas Gautier
- INSERM, Université Bourgogne Franche-Comté, LNC UMR1231, Dijon, France
- LipSTIC LabEx, Dijon, France
| | - Cécile Chagué
- Université Bourgogne Franche-Comté, EFS Bourgogne Franche-Comté, INSERM, UMR1098, Besançon, France
- Department of Hematology, University Hospital of Besançon, Besançon, France
| | - Jean-Paul Pais de Barros
- INSERM, Université Bourgogne Franche-Comté, LNC UMR1231, Dijon, France
- LipSTIC LabEx, Dijon, France
| | - Valérie Deckert
- INSERM, Université Bourgogne Franche-Comté, LNC UMR1231, Dijon, France
- LipSTIC LabEx, Dijon, France
| | - Laurent Lagrost
- INSERM, Université Bourgogne Franche-Comté, LNC UMR1231, Dijon, France
- LipSTIC LabEx, Dijon, France
- University Hospital of Dijon, Dijon, France
| | - Philippe Saas
- Université Bourgogne Franche-Comté, EFS Bourgogne Franche-Comté, INSERM, UMR1098, Besançon, France
- Department of Hematology, University Hospital of Besançon, Besançon, France
- LipSTIC LabEx, FHU INCREASE, Besançon, France
| |
Collapse
|
448
|
Role of the intestinal mucosa in acute gastrointestinal GVHD. Blood 2017; 128:2395-2402. [PMID: 27856471 DOI: 10.1182/blood-2016-06-716738] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Accepted: 09/14/2016] [Indexed: 12/11/2022] Open
Abstract
Intestinal graft-versus-host disease (GVHD) remains a significant obstacle to the success of allogeneic hematopoietic cell transplantation. The intestinal mucosa comprises the inner lining of the intestinal tract and maintains close proximity with commensal microbes that reside within the intestinal lumen. Recent advances have significantly improved our understanding of the interactions between the intestinal mucosa and the enteric microbiota. Changes in host mucosal tissue and commensals posttransplant have been actively investigated, and provocative insights into mucosal immunity and the enteric microbiota are now being translated into clinical trials of novel approaches for preventing and treating acute GVHD. In this review, we summarize recent findings related to aspects of the intestinal mucosa during acute GVHD.
Collapse
|
449
|
Integrated meta-omic analyses of the gastrointestinal tract microbiome in patients undergoing allogeneic hematopoietic stem cell transplantation. Transl Res 2017; 186:79-94.e1. [PMID: 28686852 DOI: 10.1016/j.trsl.2017.06.008] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Revised: 05/17/2017] [Accepted: 06/12/2017] [Indexed: 02/06/2023]
Abstract
In patients undergoing allogeneic hematopoietic stem cell transplantation (allo-HSCT), treatment-induced changes to the gastrointestinal tract (GIT) microbiome have been linked to adverse outcomes, most notably graft-versus-host disease (GvHD). However, it is presently unknown whether this relationship is causal or consequential. Here, we performed an integrated meta-omic analysis to probe deeper into the GIT microbiome changes during allo-HSCT and its accompanying treatments. We used 16S and 18S rRNA gene amplicon sequencing to resolve archaea, bacteria, and eukaryotes within the GIT microbiomes of 16 patients undergoing allo-HSCT for the treatment of hematologic malignancies. These results revealed a major shift in the GIT microbiome after allo-HSCT including a marked reduction in bacterial diversity, accompanied by only limited changes in eukaryotes and archaea. An integrated analysis of metagenomic and metatranscriptomic data was performed on samples collected from a patient before and after allo-HSCT for acute myeloid leukemia. This patient developed severe GvHD, leading to death 9 months after allo-HSCT. In addition to drastically decreased bacterial diversity, the post-treatment microbiome showed a higher overall number and higher expression levels of antibiotic resistance genes (ARGs). One specific Escherichia coli strain causing a paravertebral abscess was linked to GIT dysbiosis, suggesting loss of intestinal barrier integrity. The apparent selection for bacteria expressing ARGs suggests that prophylactic antibiotic administration may adversely affect the overall treatment outcome. We therefore assert that such analyses including information about the selection of pathogenic bacteria expressing ARGs may assist clinicians in "personalizing" regimens for individual patients to improve overall outcomes.
Collapse
|
450
|
Rescigno M. The microbiota revolution: Excitement and caution. Eur J Immunol 2017; 47:1406-1413. [PMID: 28675439 DOI: 10.1002/eji.201646576] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Revised: 05/05/2017] [Accepted: 06/29/2017] [Indexed: 12/21/2022]
Abstract
Scientific progress is characterized by important technological advances. Next-generation DNA sequencing has, in the past few years, led to a major scientific revolution: the microbiome revolution. It has become possible to generate a fingerprint of the whole microbiota of any given environment. As it becomes clear that the microbiota affects several aspects of our lives, each new scientific finding should ideally be analyzed in light of these communities. For instance, animal experimentation should consider animal sources and husbandry; human experimentation should include analysis of microenvironmental cues that might affect the microbiota, including diet, antibiotic, and drug use, genetics. When analyzing the activity of a drug, we should remember that, according to the microbiota of the host, different drug activities might be observed, either due to modification or degradation by the microbiota, or because the microbiota changes the immune system of the host in a way that makes that drug more or less effective. This minireview will not be a comprehensive review on the interaction between the host and microbiota, but it will aim at creating awareness on why we should not forget the contribution of the microbiota in any single aspect of biology.
Collapse
Affiliation(s)
- Maria Rescigno
- Department of Experimental Oncology, European Institute of Oncology, Milan, Italy.,Dipartimento di Scienze della Salute, Universita' di Milano, Milan, Italy
| |
Collapse
|