1
|
Ehx G, Ritacco C, Baron F. Pathophysiology and preclinical relevance of experimental graft-versus-host disease in humanized mice. Biomark Res 2024; 12:139. [PMID: 39543777 PMCID: PMC11566168 DOI: 10.1186/s40364-024-00684-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Accepted: 11/06/2024] [Indexed: 11/17/2024] Open
Abstract
Graft-versus-host disease (GVHD) is a life-threatening complication of allogeneic hematopoietic cell transplantations (allo-HCT) used for the treatment of hematological malignancies and other blood-related disorders. Until recently, the discovery of actionable molecular targets to treat GVHD and their preclinical testing was almost exclusively based on modeling allo-HCT in mice by transplanting bone marrow and splenocytes from donor mice into MHC-mismatched recipient animals. However, due to fundamental differences between human and mouse immunology, the translation of these molecular targets into the clinic can be limited. Therefore, humanized mouse models of GVHD were developed to circumvent this limitation. In these models, following the transplantation of human peripheral blood mononuclear cells (PBMCs) into immunodeficient mice, T cells recognize and attack mouse organs, inducing GVHD. Thereby, humanized mice provide a platform for the evaluation of the effects of candidate therapies on GVHD mediated by human immune cells in vivo. Understanding the pathophysiology of this xenogeneic GVHD is therefore crucial for the design and interpretation of experiments performed with this model. In this article, we comprehensively review the cellular and molecular mechanisms governing GVHD in the most commonly used model of xenogeneic GVHD: PBMC-engrafted NOD/LtSz-PrkdcscidIL2rγtm1Wjl (NSG) mice. By re-analyzing public sequencing data, we also show that the clonal expansion and the transcriptional program of T cells in humanized mice closely reflect those in humans. Finally, we highlight the strengths and limitations of this model, as well as arguments in favor of its biological relevance for studying T-cell reactions against healthy tissues or cancer cells.
Collapse
Affiliation(s)
- Grégory Ehx
- Laboratory of Hematology, GIGA Institute, University of Liege, Liege, Belgium.
- Walloon Excellence in Life Sciences and Biotechnology (WELBIO) Department, WEL Research Institute, Wavre, Belgium.
| | - Caroline Ritacco
- Laboratory of Hematology, GIGA Institute, University of Liege, Liege, Belgium
| | - Frédéric Baron
- Laboratory of Hematology, GIGA Institute, University of Liege, Liege, Belgium
- Department of Medicine, Division of Hematology, CHU of Liege, University of Liege, Liege, Belgium
| |
Collapse
|
2
|
Farhan S, Mazur I, Hartzell S, Xie P, Neme K, German A, Mikulandric N, Patel K, Wu M, Kortam N, Yaseen A, Sweidan A, Latack K, Emole J, Peres E, Abidi MH, Ramesh M. Ciprofloxacin versus levofloxacin prophylaxis in hematopoietic stem cell transplantation: A randomized trial. Int J Infect Dis 2024; 147:107172. [PMID: 39019103 DOI: 10.1016/j.ijid.2024.107172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 07/09/2024] [Accepted: 07/10/2024] [Indexed: 07/19/2024] Open
Abstract
OBJECTIVES We aimed to assess whether there is a difference between ciprofloxacin and levofloxacin as prophylaxis in hematopoietic stem cell transplant (SCT) recipients. METHODS This is a prospective, randomized trial in patients receiving SCT at Henry Ford Health in the United States of America. We randomly assigned patients (1:1) to receive ciprofloxacin or levofloxacin. The primary outcome was incidence of bloodstream bacterial infections (BSI) up to day 60 after SCT. RESULTS Between June 4, 2018, and May 23, 2022, we randomly assigned 308 consecutive patients to receive ciprofloxacin (154 patients) or levofloxacin (154 patients). BSI was similar in both the ciprofloxacin and levofloxacin groups (18 [11.7%] vs 18 [11.7%]). Pneumonia was more frequent in the ciprofloxacin group compared to the levofloxacin group (18 [18%] vs 7 [23%]; relative risk 2.57, 95% CI 1.11-5.98; p = 0.028). There were no differences in neutrophil engraftment, fever, Clostridium difficile infection, relapse incidence, overall survival, nonrelapse mortality, length of stay post-SCT, or intensive care unit admission. CONCLUSION Although both prophylaxis regimens demonstrated the same efficacy in SCT recipients, levofloxacin prophylaxis led to less pneumonia in the first 60 days post-SCT. TRIAL REGISTRATION This study is registered on ClinicalTrials.gov, NCT03850379.
Collapse
Affiliation(s)
- Shatha Farhan
- Stem Cell Transplant and Cellular Therapy, Henry Ford Health, Detroit, MI, USA.
| | - Izabela Mazur
- Stem Cell Transplant and Cellular Therapy, Henry Ford Health, Detroit, MI, USA
| | - Susan Hartzell
- Wayne State University School of Medicine, Detroit, MI, USA
| | - Peter Xie
- MetroHealth System, Cleveland, OH, USA
| | - Klodiana Neme
- Stem Cell Transplant and Cellular Therapy, Henry Ford Health, Detroit, MI, USA
| | - Angela German
- Stem Cell Transplant and Cellular Therapy, Henry Ford Health, Detroit, MI, USA
| | - Nancy Mikulandric
- Stem Cell Transplant and Cellular Therapy, Henry Ford Health, Detroit, MI, USA
| | - Kunj Patel
- Department of Internal Medicine, Henry Ford Hospital, Detroit, MI, USA
| | - Min Wu
- Wayne State University School of Medicine, Detroit, MI, USA
| | - Neda Kortam
- Division of Rheumatology, University of Michigan, Ann Arbor, MI, USA
| | - Aseel Yaseen
- Division of Endocrinology, Diabetes, & Metabolism, Henry Ford Health, Detroit, MI, USA
| | - Aroob Sweidan
- Hematology Oncology, Henry Ford Health, Detroit, MI, USA
| | - Katie Latack
- Public Health Sciences, Henry Ford Health, Detroit, MI, USA
| | - Josephine Emole
- Stem Cell Transplant and Cellular Therapy, Henry Ford Health, Detroit, MI, USA
| | - Edward Peres
- Stem Cell Transplant and Cellular Therapy, Henry Ford Health, Detroit, MI, USA
| | - Muneer H Abidi
- Stem Cell Transplant and Cellular Therapy, Henry Ford Health, Detroit, MI, USA
| | - Mayur Ramesh
- Transplant Infectious Diseases and Immunotherapy, Henry Ford Hospital, Detroit, MI, USA
| |
Collapse
|
3
|
Wenger V, Zeiser R. Deciphering the role of the major histocompatibility complex, the intestinal microbiome and metabolites in the pathogenesis of acute graft-versus-host disease. Best Pract Res Clin Haematol 2024; 37:101567. [PMID: 39396261 DOI: 10.1016/j.beha.2024.101567] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 07/01/2024] [Accepted: 07/23/2024] [Indexed: 10/15/2024]
Abstract
Allogeneic hematologic stem cell transplantation is a cornerstone in modern hematological treatment, yet its efficacy is compromised by acute Graft-versus-Host Disease. In acute Graft-versus-Host Disease, conditioning regimen induced epithelial damage leads to release of damage and pathogen associated molecular patters which in turns triggers activation of alloreactive donor T cells, ultimately resulting in destruction of healthy tissue. Advances in major histocompatibility complex typing and preclinical studies using tissue specific major histocompatibility complex deletion have illuminated the contributions of both, hematopoietic and non-hematopoietic cells to acute Graft-versus-Host Disease pathophysiology. Concurrently, high-throughput sequencing techniques have enabled researchers to recognize the significant impact of the intestinal microbiome and newly discovered metabolites in the pathophysiology of acute Graft-versus-Host Disease. In this review, we discuss the implications of major histocompatibility complex expression on hematopoietic and non-hematopoietic cells, the effect on the intestinal microbiome and the metabolic alterations that contribute to acute Graft-versus-Host Disease. By combining these findings, we hope to untangle the complexity of acute Graft-versus-Host Disease, ultimately paving the way for the development of novel and more effective treatmen options in patients.
Collapse
Affiliation(s)
- Valentin Wenger
- Department of Medicine I, Medical Center - University of Freiburg, Faculty of Medicine, Albert Ludwigs University (ALU), Freiburg, Germany
| | - Robert Zeiser
- Department of Medicine I, Medical Center - University of Freiburg, Faculty of Medicine, Albert Ludwigs University (ALU), Freiburg, Germany; German Cancer Consortium (DKTK), Freiburg, and German Cancer Research Center (DKFZ), Heidelberg, Germany; Signalling Research Centres BIOSS and CIBSS - Centre for Integrative Biological Signalling Studies, University of Freiburg, Germany.
| |
Collapse
|
4
|
Maurer K, Antin JH. The graft versus leukemia effect: donor lymphocyte infusions and cellular therapy. Front Immunol 2024; 15:1328858. [PMID: 38558819 PMCID: PMC10978651 DOI: 10.3389/fimmu.2024.1328858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 02/28/2024] [Indexed: 04/04/2024] Open
Abstract
Allogeneic hematopoietic stem cell transplantation (HSCT) is a potentially curative therapy for many hematologic malignancies as well as non-malignant conditions. Part of the curative basis underlying HSCT for hematologic malignancies relies upon induction of the graft versus leukemia (GVL) effect in which donor immune cells recognize and eliminate residual malignant cells within the recipient, thereby maintaining remission. GVL is a clinically evident phenomenon; however, specific cell types responsible for inducing this effect and molecular mechanisms involved remain largely undefined. One of the best examples of GVL is observed after donor lymphocyte infusions (DLI), an established therapy for relapsed disease or incipient/anticipated relapse. DLI involves infusion of peripheral blood lymphocytes from the original HSCT donor into the recipient. Sustained remission can be observed in 20-80% of patients treated with DLI depending upon the underlying disease and the intrinsic burden of targeted cells. In this review, we will discuss current knowledge about mechanisms of GVL after DLI, experimental strategies for augmenting GVL by manipulation of DLI (e.g. neoantigen vaccination, specific cell type selection/depletion) and research outlook for improving DLI and cellular immunotherapies for hematologic malignancies through better molecular definition of the GVL effect.
Collapse
Affiliation(s)
| | - Joseph H. Antin
- Division of Hematologic Malignancies, Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
5
|
Enriquez J, McDaniel Mims B, Stroever S, dos Santos AP, Jones-Hall Y, Furr KL, Grisham MB. Influence of Housing Temperature and Genetic Diversity on Allogeneic T Cell-Induced Tissue Damage in Mice. PATHOPHYSIOLOGY 2023; 30:522-547. [PMID: 37987308 PMCID: PMC10661280 DOI: 10.3390/pathophysiology30040039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 11/12/2023] [Accepted: 11/18/2023] [Indexed: 11/22/2023] Open
Abstract
The objective of this study was to determine how housing temperature and genetic diversity affect the onset and severity of allogeneic T cell-induced tissue damage in mice subjected to reduced intensity conditioning (RIC). We found that adoptive transfer of allogeneic CD4+ T cells from inbred donors into sub-lethally irradiated inbred recipients (I→I) housed at standard housing temperatures (ST; 22-24 °C) induced extensive BM and spleen damage in the absence of injury to any other tissue. Although engraftment of T cells in RIC-treated mice housed at their thermo-neutral temperature (TNT; 30-32 °C) also developed similar BM and spleen damage, their survival was markedly and significantly increased when compared to their ST counterparts. In contrast, the adoptive transfer of allogeneic T cells into RIC-treated outbred CD1 recipients failed to induce disease in any tissue at ST or TNT. The lack of tissue damage was not due to defects in donor T cell trafficking to BM or spleen but was associated with the presence of large numbers of B cells and myeloid cells within these tissues that are known to contain immunosuppressive regulatory B cells and myeloid-derived suppressor cells. These data demonstrate, for the first time, that housing temperature affects the survival of RIC-treated I→I mice and that RIC-conditioned outbred mice are resistant to allogeneic T cell-induced BM and spleen damage.
Collapse
Affiliation(s)
- Josue Enriquez
- Department of Microbiology and Immunology, University of Gothenburg, 40530 Gothenburg, Sweden
| | - Brianyell McDaniel Mims
- Department of Oral Health Sciences, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Stephanie Stroever
- Department of Medical Education, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - Andrea Pires dos Santos
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, West Lafayette, IN 47907, USA
| | - Yava Jones-Hall
- Department of Veterinary Pathobiology, School of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843, USA
| | - Kathryn L. Furr
- Department of Immunology and Molecular Microbiology, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - Matthew B. Grisham
- Department of Immunology and Molecular Microbiology, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| |
Collapse
|
6
|
Scott AP, Henden A, Kennedy GA, Tey SK. PET assessment of acute gastrointestinal graft versus host disease. Bone Marrow Transplant 2023; 58:973-979. [PMID: 37537245 PMCID: PMC10471499 DOI: 10.1038/s41409-023-02038-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 06/29/2023] [Accepted: 07/07/2023] [Indexed: 08/05/2023]
Abstract
Acute gastrointestinal graft versus host disease (GI-GVHD) is a common complication following allogeneic haematopoietic cell transplantation (HCT), and is characterised by severe morbidity, frequent treatment-refractoriness, and high mortality. Early, accurate identification of GI-GVHD could allow for therapeutic interventions to ameliorate its severity, improve response rates and survival; however, standard endoscopic biopsy is inadequately informative in terms of diagnostic sensitivity or outcome prediction. In an era where rapid technological and laboratory advances have dramatically expanded our understanding of GI-GVHD biology and potential therapeutic targets, there is substantial scope for novel investigations that can precisely guide GI-GVHD management. In particular, the combination of tissue-based biomarker assessment (plasma cytokines, faecal microbiome) and molecular imaging by positron emission tomography (PET) offers the potential for non-invasive, real-time in vivo assessment of donor:recipient immune activity within the GI tract for GI-GVHD prediction or diagnosis. In this article, we review the evidence regarding GI-GVHD diagnosis, and examine the potential roles and translational opportunities posed by these novel diagnostic tools, with a focus on the evolving role of PET.
Collapse
Affiliation(s)
- Ashleigh P Scott
- Department of Haematology and Bone Marrow Transplant, Royal Brisbane and Women's Hospital, Brisbane, QLD, Australia.
- School of Medicine, The University of Queensland, Brisbane, QLD, Australia.
| | - Andrea Henden
- Department of Haematology and Bone Marrow Transplant, Royal Brisbane and Women's Hospital, Brisbane, QLD, Australia
- School of Medicine, The University of Queensland, Brisbane, QLD, Australia
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Glen A Kennedy
- Department of Haematology and Bone Marrow Transplant, Royal Brisbane and Women's Hospital, Brisbane, QLD, Australia
- School of Medicine, The University of Queensland, Brisbane, QLD, Australia
| | - Siok-Keen Tey
- Department of Haematology and Bone Marrow Transplant, Royal Brisbane and Women's Hospital, Brisbane, QLD, Australia
- School of Medicine, The University of Queensland, Brisbane, QLD, Australia
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| |
Collapse
|
7
|
Mohammadpour H, Tsuji T, MacDonald CR, Sarow JL, Rosenheck H, Daneshmandi S, Choi JE, Qiu J, Matsuzaki J, Witkiewicz AK, Attwood K, Blazar BR, Odunsi K, Repasky EA, McCarthy PL. Galectin-3 expression in donor T cells reduces GvHD severity and lethality after allogeneic hematopoietic cell transplantation. Cell Rep 2023; 42:112250. [PMID: 36924493 PMCID: PMC10116561 DOI: 10.1016/j.celrep.2023.112250] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 01/05/2023] [Accepted: 02/25/2023] [Indexed: 03/17/2023] Open
Abstract
Abundant donor cytotoxic T cells that attack normal host organs remain a major problem for patients receiving allogeneic hematopoietic cell transplantation (allo-HCT). Despite an increase in our knowledge of the pathobiology of acute graft versus host disease (aGvHD), the mechanisms regulating the proliferation and function of donor T cells remain unclear. Here, we show that activated donor T cells express galectin-3 (Gal-3) after allo-HCT. In both major and minor histocompatibility-mismatched models of murine aGvHD, expression of Gal-3 is associated with decreased T cell activation and suppression of the secretion of effector cytokines, including IFN-γ and GM-CSF. Mechanistically, Gal-3 results in activation of NFAT signaling, which can induce T cell exhaustion. Gal-3 overexpression in human T cells prevents severe disease by suppressing cytotoxic T cells in xenogeneic aGvHD models. Together, these data identify the Gal-3-dependent regulatory pathway in donor T cells as a critical component of inflammation in aGvHD.
Collapse
Affiliation(s)
- Hemn Mohammadpour
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA.
| | - Takemasa Tsuji
- Center for Immunotherapy, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Cameron R MacDonald
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Joseph L Sarow
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Hanna Rosenheck
- Department of Medicine, Transplant and Cellular Therapy Program, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Saeed Daneshmandi
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA; Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Jee Eun Choi
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Jingxin Qiu
- Department of Molecular and Cellular Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Junko Matsuzaki
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Agnieszka K Witkiewicz
- Department of Pathology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Kristopher Attwood
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Bruce R Blazar
- Department of Pediatrics, Division of Blood & Marrow Transplant & Cellular Therapy, University of Minnesota, Minneapolis, MN 55455, USA
| | - Kunle Odunsi
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Elizabeth A Repasky
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Philip L McCarthy
- Center for Immunotherapy, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA.
| |
Collapse
|
8
|
Kreft A, Schulze L, Ries I, Schindeldecker M, Neumann H. Histological diagnosis of acute graft-versus-host disease in different sites of the upper gastrointestinal tract with correlation to endoscopic findings. Leuk Lymphoma 2023; 64:433-439. [PMID: 36335433 DOI: 10.1080/10428194.2022.2142056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Graft-versus-host disease (GvHD) involving the intestine is a threat to patients after allogeneic hematopoietic stem cell transplantation (alloHSCT). We evaluated biopsies from different sites of the upper gastrointestinal tract (GIT) of 97 patients after alloHSCT. Forty-six patients with clinical symptoms consistent with upper GI GvHD revealed histological features of GvHD in the esophagus, stomach, and/or duodenum. Biopsies of the duodenum and esophagus were significantly more sensitive for signs of GvHD than those of the gastric antrum or corpus. The histological features of GvHD were significantly correlated with the endoscopic findings of ulcers, erosion, atrophy, and white plaques; however, the sensitivity and specificity of the latter were low. In univariate analysis, overall mortality was significantly associated with histological GvHD signs in all four sites. Nonrelapse mortality was associated with histologic GvHD features in the antrum only. Regarding GvHD diagnosis, biopsies of the upper gastrointestinal tract should include the duodenum and/or esophagus.
Collapse
Affiliation(s)
- Andreas Kreft
- Institute of Pathology, University Medical Center Mainz, Mainz, Germany
| | - Lisa Schulze
- Institute of Pathology, University Medical Center Mainz, Mainz, Germany
| | - Isabelle Ries
- 3rd Medical Department, Hematology, Oncology and Pneumology, University Medical Center Mainz, Germany
| | | | - Helmut Neumann
- GastroZentrum Lippe, Bad Salzuflen, Germany.,Department of Medicine 1, University Medical Center Mainz, Mainz, Germany
| |
Collapse
|
9
|
Yamamoto A, Kambara Y, Fujiwara H. Impact of oral microbiota on pathophysiology of GVHD. Front Immunol 2023; 14:1132983. [PMID: 36969182 PMCID: PMC10033631 DOI: 10.3389/fimmu.2023.1132983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Accepted: 02/23/2023] [Indexed: 03/29/2023] Open
Abstract
Allogeneic transplantation of hematopoietic cells is the only curative therapy for several hematopoietic disease in which patients receive cytotoxic conditioning regimens followed by infusion of hematopoietic stem cells. Although the outcomes have improved over the past decades, graft-versus-host-disease (GVHD), the most common life-threatening complication, remains a major cause of non-relapse morbidity and mortality. Pathophysiology of acute GVHD characterized by host antigen-presenting cells after tissue damage and donor T-cells is well studied, and additionally the importance of recipient microbiota in the intestine is elucidated in the GVHD setting. Oral microbiota is the second most abundant bacterial flora in the body after the intestinal tract, and it is related to chronic inflammation and carcinogenesis. Recently, composition of the oral microbiome in GVHD related to transplantation has been characterized and several common patterns, dysbiosis and enrichment of the specific bacterial groups, have been reported. This review focuses on the role of the oral microbiota in the context of GVHD.
Collapse
Affiliation(s)
- Akira Yamamoto
- Department of Hematology and Oncology, Okayama University Hospital, Okayama, Japan
| | - Yui Kambara
- Department of Hematology and Oncology and Respiratory Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Hideaki Fujiwara
- Department of Hematology and Oncology, Okayama University Hospital, Okayama, Japan
- *Correspondence: Hideaki Fujiwara,
| |
Collapse
|
10
|
Møller DL, Kielsen K, Nielsen CH, Sengeløv H, Pedersen AE, Ryder LP, Müller K. Thymic stromal lymphopoietin levels after allogeneic hematopoietic stem cell transplantation. Immunopharmacol Immunotoxicol 2022; 44:1004-1012. [PMID: 35899395 DOI: 10.1080/08923973.2022.2102989] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND Thymic stromal lymphopoietin (TSLP) is an immunoregulatory, Th2-polarizing cytokine produced by epithelial cells. We hypothesized that TSLP affects immune reconstitution after hematopoietic stem cell transplantation (HSCT) leading to increased alloreactivity. METHODS We measured plasma TSLP by ELISA in 38 patients and assessed the immune reconstitution by flow cytometry. RESULTS TSLP levels rose after initiation of the conditioning to peak at day +21 after HSCT (p = .03), where TSLP levels correlated with counts of neutrophils (rho = 0.36, p = .04), monocytes (rho = 0.58, p = .006), and lymphocytes (rho = 0.59, p = .02). Overall absolute TSLP levels were not associated with acute or chronic graft-vs-host disease (a/cGvHD). However, patients mounting a sustained increase in TSLP levels at day +90 had a higher risk of cGvHD compared to patients who had returned to pre-conditioning levels at that stage (cumulative incidence: 77% vs. 38%, p = .01). CONCLUSION In conclusion, this study suggests a role of TSLP in immune reconstitution and alloreactivity post-HSCT. lymphopoietin (TSLP) is an immunoregulatory, Th2-polarizing cytokine produced by epithelial cells. We hypothesized that TSLP affects immune reconstitution after hematopoietic stem cell transplantation (HSCT) leading to increased alloreactivity. We measured plasma TSLP by ELISA in 38 patients and assessed the immune reconstitution by flow cytometry.
Collapse
Affiliation(s)
- Dina Leth Møller
- Department of Pediatrics and Adolescent Medicine, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark.,Institute for Inflammation Research, Department of Rheumatology and Spine Diseases, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Katrine Kielsen
- Department of Pediatrics and Adolescent Medicine, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark.,Institute for Inflammation Research, Department of Rheumatology and Spine Diseases, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Claus Henrik Nielsen
- Institute for Inflammation Research, Department of Rheumatology and Spine Diseases, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Henrik Sengeløv
- Department of Hematology, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | | | - Lars Peter Ryder
- The Tissue Typing Laboratory, Department of Clinical Immunology, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Klaus Müller
- Department of Pediatrics and Adolescent Medicine, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark.,Institute for Inflammation Research, Department of Rheumatology and Spine Diseases, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark.,Institute of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
11
|
Chhabra S, Szabo A, Clurman A, McShane K, Waters N, Eastwood D, Samanas L, Fei T, Armijo G, Abedin S, Longo W, Hari P, Hamadani M, Shah NN, Runaas L, Jerkins JH, van den Brink M, Peled JU, Drobyski WR. Mitigation of gastrointestinal graft- versus-host disease with tocilizumab prophylaxis is accompanied by preservation of microbial diversity and attenuation of enterococcal domination. Haematologica 2022; 108:250-256. [PMID: 36106394 PMCID: PMC9827178 DOI: 10.3324/haematol.2022.281309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Indexed: 02/05/2023] Open
Affiliation(s)
- Saurabh Chhabra
- Bone Marrow Transplant and Cellular Therapy Program, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI,S.CHHABRA -
| | - Aniko Szabo
- Division of Biostatistics, Institute for Health and Equity, Medical College of Wisconsin, Milwaukee, WI,*AS and AC contributed equally as co-second authors
| | - Annelie Clurman
- Department of Immunology, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY,*AS and AC contributed equally as co-second authors
| | - Katelynn McShane
- Cancer Center Clinical Trials Office, Medical College of Wisconsin, Milwaukee, WI
| | - Nicholas Waters
- Department of Immunology, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Daniel Eastwood
- Division of Biostatistics, Institute for Health and Equity, Medical College of Wisconsin, Milwaukee, WI
| | - Lisa Samanas
- Department of Pharmacy, Froedtert Health, Milwaukee, WI
| | - Teng Fei
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Gabriel Armijo
- Department of Immunology, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Sameen Abedin
- Bone Marrow Transplant and Cellular Therapy Program, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI
| | - Walter Longo
- Bone Marrow Transplant and Cellular Therapy Program, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI
| | - Parameswaran Hari
- Bone Marrow Transplant and Cellular Therapy Program, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI
| | - Mehdi Hamadani
- Bone Marrow Transplant and Cellular Therapy Program, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI
| | - Nirav N. Shah
- Bone Marrow Transplant and Cellular Therapy Program, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI
| | - Lyndsey Runaas
- Bone Marrow Transplant and Cellular Therapy Program, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI
| | - James H. Jerkins
- Bone Marrow Transplant and Cellular Therapy Program, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI
| | - Marcel van den Brink
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, Weill Cornell Medical College, New York, NY, USA
| | - Jonathan U. Peled
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, Weill Cornell Medical College, New York, NY, USA,JUP and WRD contributed equally as co-senior authors
| | - William R. Drobyski
- Bone Marrow Transplant and Cellular Therapy Program, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI,JUP and WRD contributed equally as co-senior authors
| |
Collapse
|
12
|
Zeng C, Cheng TT, Ma X, Liu Y, Hua J, Chen X, Wang SY, Xu YJ. The absence of AhR in CD4 + T cells in patients with acute graft-versus-host disease may be related to insufficient CTCF expression. Clin Epigenetics 2022; 14:109. [PMID: 36056390 PMCID: PMC9440523 DOI: 10.1186/s13148-022-01330-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Accepted: 08/26/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Acute graft-versus-host disease (aGVHD) is a life-threatening complication of allogeneic hematopoietic stem cell transplantation (allo-HSCT). Accumulating evidence suggests that imbalanced Treg/Th17 ratio accelerates the progression of aGVHD. The aryl hydrocarbon receptor (AhR) is a basic helix-loop-helix transcription factor activated through cognate ligand binding. Current evidence supports that AhR plays a critical regulatory role in the differentiation of Treg and Th17 cells. However, the relationship between AhR and aGVHD remains unclear. RESULTS Our results showed that AhR expression was downregulated significantly in CD4+ T cells from patients with aGVHD compared with the non-aGVHD group. We also discovered that after activating AhR deficient CD4+ T cells, the expression levels of the activation markers-CD40L, CD134 and CD137 and cell proliferation activity were significantly higher than those of AhR-expressing CD4+ T cells. Restoring the expression of AhR in aGVHD CD4+ T cells resulted in significantly increased percentage of Tregs and associated gene transcripts, including Foxp3, IL-10 and CD39. In contrast, Th17 cell amounts and the transcription of related genes, including RORγt, IL-17A and IL-17F, were significantly reduced. We confirmed that CTCF recruited EP300 and TET2 to bind to the AhR promoter region and promoted AhR expression by mediating histone H3K9/K14 hyperacetylation and DNA demethylation in this region. The low expression of CTCF caused histone hypoacetylation and DNA hypermethylation of the AhR promoter, resulting in insufficient expression in aGVHD CD4+ T cells. CONCLUSIONS CTCF is an important inducer of AhR transcription. Insufficient expression of CTCF leads to excessive AhR downregulation, resulting in substantial CD4+ T cell activation and Th17/Treg ratio increase, thereby mediating the occurrence of aGVHD.
Collapse
Affiliation(s)
- Cong Zeng
- Department of Hematology, Xiangya Hospital, Central South University, Changsha, Hunan, China.,National Clinical Research Center for Geriatric Diseases, Xiangya Hospital, Changsha, China.,Hunan Hematology Oncology Clinical Medical Research Center, Changsha, China
| | - Ting-Ting Cheng
- Department of Hematology, Xiangya Hospital, Central South University, Changsha, Hunan, China.,National Clinical Research Center for Geriatric Diseases, Xiangya Hospital, Changsha, China.,Hunan Hematology Oncology Clinical Medical Research Center, Changsha, China
| | - Xia Ma
- Department of Hematology, Xiangya Hospital, Central South University, Changsha, Hunan, China.,National Clinical Research Center for Geriatric Diseases, Xiangya Hospital, Changsha, China.,Hunan Hematology Oncology Clinical Medical Research Center, Changsha, China
| | - Yi Liu
- Department of Hematology, Xiangya Hospital, Central South University, Changsha, Hunan, China.,National Clinical Research Center for Geriatric Diseases, Xiangya Hospital, Changsha, China.,Hunan Hematology Oncology Clinical Medical Research Center, Changsha, China
| | - Juan Hua
- Department of Hematology, Xiangya Hospital, Central South University, Changsha, Hunan, China.,National Clinical Research Center for Geriatric Diseases, Xiangya Hospital, Changsha, China.,Hunan Hematology Oncology Clinical Medical Research Center, Changsha, China
| | - Xu Chen
- Department of Hematology, Xiangya Hospital, Central South University, Changsha, Hunan, China.,National Clinical Research Center for Geriatric Diseases, Xiangya Hospital, Changsha, China.,Hunan Hematology Oncology Clinical Medical Research Center, Changsha, China
| | - Shi-Yu Wang
- Department of Hematology, Xiangya Hospital, Central South University, Changsha, Hunan, China.,National Clinical Research Center for Geriatric Diseases, Xiangya Hospital, Changsha, China.,Hunan Hematology Oncology Clinical Medical Research Center, Changsha, China
| | - Ya-Jing Xu
- Department of Hematology, Xiangya Hospital, Central South University, Changsha, Hunan, China. .,National Clinical Research Center for Geriatric Diseases, Xiangya Hospital, Changsha, China. .,Hunan Hematology Oncology Clinical Medical Research Center, Changsha, China. .,National Clinical Research Center for Hematologic Diseases, The First Affiliated Hospital of Soochow University, Suzhou, China.
| |
Collapse
|
13
|
Guo H, Li R, Wang M, Hou Y, Liu S, Peng T, Zhao X, Lu L, Han Y, Shao Y, Chang Y, Li C, Huang X. Multiomics Analysis Identifies SOCS1 as Restraining T Cell Activation and Preventing Graft-Versus-Host Disease. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2200978. [PMID: 35585676 PMCID: PMC9313503 DOI: 10.1002/advs.202200978] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 04/14/2022] [Indexed: 05/03/2023]
Abstract
Graft-versus-host disease (GVHD) is a major life-threatening complication of allogeneic hematopoietic stem cell transplantation (allo-HSCT). Inflammatory signaling pathways promote T-cell activation and are involved in the pathogenesis of GVHD. Suppressor of cytokine signaling 1 (SOCS1) is a critical negative regulator for several inflammatory cytokines. However, its regulatory role in T-cell activation and GVHD has not been elucidated. Multiomics analysis of the transcriptome and chromatin structure of granulocyte-colony-stimulating-factor (G-CSF)-administered hyporesponsive T cells from healthy donors reveal that G-CSF upregulates SOCS1 by reorganizing the chromatin structure around the SOCS1 locus. Parallel in vitro and in vivo analyses demonstrate that SOCS1 is critical for restraining T cell activation. Loss of Socs1 in T cells exacerbates GVHD pathogenesis and diminishes the protective role of G-CSF in GVHD mouse models. Further analysis shows that SOCS1 inhibits T cell activation not only by inhibiting the colony-stimulating-factor 3 receptor (CSF3R)/Janus kinase 2 (JAK2)/signal transducer and activator of transcription 3 (STAT3) pathway, but also by restraining activation of the inflammasome signaling pathway. Moreover, high expression of SOCS1 in T cells from patients correlates with low acute GVHD occurrence after HSCT. Overall, these findings identify that SOCS1 is critical for inhibiting T cell activation and represents a potential target for the attenuation of GVHD.
Collapse
Affiliation(s)
- Huidong Guo
- Peking University Institute of HematologyNational Clinical Research Center for Hematologic DiseaseBeijing Key Laboratory of Hematopoietic Stem Cell TransplantationSchool of Life SciencesPeking University People's HospitalPeking UniversityBeijing100044China
| | - Ruifeng Li
- Peking University Institute of HematologyNational Clinical Research Center for Hematologic DiseaseBeijing Key Laboratory of Hematopoietic Stem Cell TransplantationSchool of Life SciencesPeking University People's HospitalPeking UniversityBeijing100044China
- Peking‐Tsinghua Center for Life SciencesPeking UniversityBeijing100080China
- Institute for Immunology and School of MedicineTsinghua UniversityBeijing100084China
| | - Ming Wang
- Peking University Institute of HematologyNational Clinical Research Center for Hematologic DiseaseBeijing Key Laboratory of Hematopoietic Stem Cell TransplantationSchool of Life SciencesPeking University People's HospitalPeking UniversityBeijing100044China
| | - Yingping Hou
- Peking University Institute of HematologyNational Clinical Research Center for Hematologic DiseaseBeijing Key Laboratory of Hematopoietic Stem Cell TransplantationSchool of Life SciencesPeking University People's HospitalPeking UniversityBeijing100044China
- Peking‐Tsinghua Center for Life SciencesPeking UniversityBeijing100080China
| | - Shuoshuo Liu
- Institute for Immunology and School of MedicineTsinghua UniversityBeijing100084China
- Beijing Tsinghua Changgeng HospitalBeijing102218China
| | - Ting Peng
- Peking University Institute of HematologyNational Clinical Research Center for Hematologic DiseaseBeijing Key Laboratory of Hematopoietic Stem Cell TransplantationSchool of Life SciencesPeking University People's HospitalPeking UniversityBeijing100044China
| | - Xiang‐Yu Zhao
- Peking University Institute of HematologyNational Clinical Research Center for Hematologic DiseaseBeijing Key Laboratory of Hematopoietic Stem Cell TransplantationSchool of Life SciencesPeking University People's HospitalPeking UniversityBeijing100044China
| | - Liming Lu
- Shanghai Institute of ImmunologyShanghai Jiaotong University School of Medicine280 South Chongqing RoadShanghai200025China
| | - Yali Han
- Shanghai Jiayin Biotechnology, Ltd.Shanghai200092China
| | - Yiming Shao
- Shanghai Jiayin Biotechnology, Ltd.Shanghai200092China
| | - Ying‐Jun Chang
- Peking University Institute of HematologyNational Clinical Research Center for Hematologic DiseaseBeijing Key Laboratory of Hematopoietic Stem Cell TransplantationSchool of Life SciencesPeking University People's HospitalPeking UniversityBeijing100044China
| | - Cheng Li
- Peking University Institute of HematologyNational Clinical Research Center for Hematologic DiseaseBeijing Key Laboratory of Hematopoietic Stem Cell TransplantationSchool of Life SciencesPeking University People's HospitalPeking UniversityBeijing100044China
- Center for Statistical ScienceCenter for BioinformaticsPeking UniversityBeijingChina
| | - Xiao‐Jun Huang
- Peking University Institute of HematologyNational Clinical Research Center for Hematologic DiseaseBeijing Key Laboratory of Hematopoietic Stem Cell TransplantationSchool of Life SciencesPeking University People's HospitalPeking UniversityBeijing100044China
- Peking‐Tsinghua Center for Life SciencesPeking UniversityBeijing100080China
- Research Unit of Key Technique for Diagnosis and Treatments of Hematologic Malignancies (2019RU029)Chinese Academy of Medical SciencesBeijing100730China
| |
Collapse
|
14
|
Ceramide synthase 6 impacts T-cell allogeneic response and graft-versus-host disease through regulating N-RAS/ERK pathway. Leukemia 2022; 36:1907-1915. [PMID: 35513703 PMCID: PMC9256768 DOI: 10.1038/s41375-022-01581-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 04/09/2022] [Accepted: 04/20/2022] [Indexed: 02/02/2023]
Abstract
Allogeneic hematopoietic cell transplantation (allo-HCT) is an effective immunotherapy for various hematologic malignances, predominantly through potent graft-versus-leukemia (GVL) effect. However, the mortality after allo-HCT is because of relapse of primary malignancy and followed by graft-vs-host-disease (GVHD) as a major cause of transplant-related mortality. Hence, strategies to limit GVHD while preserving the GVL effect are highly desirable. Ceramide, which serves a central role in sphingolipid metabolism, is generated by ceramide synthases (CerS1–6). In this study, we found that genetic or pharmacologic targeting of CerS6 prevented and reversed chronic GVHD (cGVHD). Furthermore, specific inhibition of CerS6 with ST1072 significantly ameliorated acute GVHD (aGVHD) while preserving the GVL effect, which differed from FTY720 that attenuated aGVHD but impaired GVL activity. At the cellular level, blockade of CerS6 restrained donor T cells from migrating into GVHD target organs and preferentially reduced activation of donor CD4 T cells. At the molecular level, CerS6 was required for optimal TCR signaling, CD3/PKCθ co-localization, and subsequent N-RAS activation and ERK signaling, especially on CD4+ T cells. The current study provides rationale and means for targeting CerS6 to control GVHD and leukemia relapse, which would enhance the efficacy of allo-HCT as an immunotherapy for hematologic malignancies in the clinic.
Collapse
|
15
|
Tarantino G, Saraceni F, Mancini G, Poiani M, Maroni L, Goteri G, Scortechini I, Fiorentini A, Dubbini MV, Marini F, Daretti L, Marzioni M, Bendia E, Benedetti A, Olivieri A. Gastrointestinal Complications after Allogeneic Hematopoietic Stem Cell Transplant: A Multidisciplinary Approach with Early Endoscopic Evaluation. Clin Hematol Int 2021; 3:161-168. [PMID: 34938988 PMCID: PMC8690703 DOI: 10.2991/chi.k.210826.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 08/26/2021] [Indexed: 11/25/2022] Open
Abstract
Gastrointestinal complications (GICs) represent the major cause of morbidity and mortality after allogeneic hematopoietic stem cell transplantation (allo-HSCT). Differential diagnosis of GICs is of paramount importance since early and reliable identification of graft-versus-host disease (GVHD) is essential for a correct management of the patients. The aim of the present retrospective study was to evaluate the occurrence of GICs after allo-HSCT and to assess the diagnostic performance of a quick endoscopic and histological assessment in the differential diagnosis between GVHD and other GI conditions. Between January 2015 and August 2019, 122 consecutive patients receiving an allo-HSCT were managed by an interdisciplinary team, supported by a dedicated endoscopic service. Clinical, therapeutic, endoscopic and histological data were analyzed for each patient. Collectively, 94 of the patients developed GICs (77%). A moderate–severe mucositis was the most frequent complication, occurring in 79 patients (84%). Acute GI-GVHD was diagnosed in 35 patients (37% of whom with GICs) and 19 of them with a moderate–severe grade. Infective acute colitis developed in eight patients, mainly due to Clostridium difficile (CD) and Cytomegalovirus infections (8.5%). Rectal biopsy showed the highest sensitivity and specificity (80% and 100%, respectively). However, when biopsy procedures were guided by symptoms and performed on apparently intact mucosa, upper histology also provided a high negative predictive value (80%). Our multidisciplinary approach with a quick endoscopic/histologic investigation in the patients receiving an allo-HSCT and who suffered GICs could improve diagnostic and therapeutic management in this challenging setting.
Collapse
Affiliation(s)
- Giuseppe Tarantino
- Clinica di Gastroenterologia, Epatologia ed Endoscopia Digestiva d'Urgenza, Via Conca 71, Ospedali Riuniti Ancona, Italy
| | - Francesco Saraceni
- Clinica Ematologica, Dipartimento di Medicina Interna, Ospedali Riuniti Ancona, Italy
| | - Giorgia Mancini
- Clinica Ematologica, Dipartimento di Medicina Interna, Ospedali Riuniti Ancona, Italy
| | - Monica Poiani
- Clinica Ematologica, Dipartimento di Medicina Interna, Ospedali Riuniti Ancona, Italy
| | - Luca Maroni
- Clinica di Gastroenterologia, Epatologia ed Endoscopia Digestiva d'Urgenza, Via Conca 71, Ospedali Riuniti Ancona, Italy
| | - Gaia Goteri
- Anatomia Patologica, Dipartimento di Medicina Interna, Ospedali Riuniti Ancona, Italy
| | - Ilaria Scortechini
- Clinica Ematologica, Dipartimento di Medicina Interna, Ospedali Riuniti Ancona, Italy
| | - Alessandro Fiorentini
- Clinica Ematologica, Dipartimento di Medicina Interna, Ospedali Riuniti Ancona, Italy
| | | | - Francesco Marini
- Malattie dell'Apparato Digerente, Endoscopia Digestiva e Malattie Infiammatorie Croniche Intestinali, Ospedali Riuniti Ancona, Italy
| | - Luigi Daretti
- Malattie dell'Apparato Digerente, Endoscopia Digestiva e Malattie Infiammatorie Croniche Intestinali, Ospedali Riuniti Ancona, Italy
| | - Marco Marzioni
- Clinica di Gastroenterologia, Epatologia ed Endoscopia Digestiva d'Urgenza, Via Conca 71, Ospedali Riuniti Ancona, Italy
| | - Emanuele Bendia
- Malattie dell'Apparato Digerente, Endoscopia Digestiva e Malattie Infiammatorie Croniche Intestinali, Ospedali Riuniti Ancona, Italy
| | - Antonio Benedetti
- Clinica di Gastroenterologia, Epatologia ed Endoscopia Digestiva d'Urgenza, Via Conca 71, Ospedali Riuniti Ancona, Italy
| | - Attilio Olivieri
- Clinica Ematologica, Dipartimento di Medicina Interna, Ospedali Riuniti Ancona, Italy
| |
Collapse
|
16
|
Qi L, Huang X, He C, Ji D, Li F. Steroid-resistant intestinal aGVHD and refractory CMV and EBV infections complicated by haplo-HSCT were successfully rescued by FMT and CTL infusion. J Int Med Res 2021; 49:3000605211063292. [PMID: 34918995 PMCID: PMC8728789 DOI: 10.1177/03000605211063292] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Haploidentical hematopoietic stem cell transplantation (haplo-HSCT) produces similar survival outcomes as HLA-matched sibling donor allogeneic HCST in younger patients with acquired severe aplastic anemia (SAA). This study reported a 29-years-old man with SAA and intracranial hemorrhage who underwent haplo-HSCT with a modified BU/CY + ATG conditioning regimen. Neutrophil and platelet engraftment were both achieved on day 14 after HSCT. The patient developed grade IV acute graft-versus-host disease (aGVHD) on day 20 and acquired cytomegalovirus (CMV) and Epstein-Barr virus (EBV) infections on day 47. After the failure of methylprednisolone, basiliximab, ruxolitinib, and antiviral treatment, the patient was diagnosed with steroid-resistant grade IV aGVHD and refractory CMV and EBV infections. We performed fecal microbiota transplantation and infused CMV- and EBV-specific cytotoxic T lymphocytes. After that the stool volume and frequency gradually decreased, and viral DNA was undetectable on day 80. This report provides helpful clinical experience for treating steroid-resistant aGVHD and refractory viral infections.
Collapse
Affiliation(s)
- Ling Qi
- Department of Hematology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Xianbao Huang
- Department of Hematology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Cong He
- Institute of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Dexiang Ji
- Department of Hematology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Fei Li
- Department of Hematology, The First Affiliated Hospital of Nanchang University, Nanchang, China.,Institute of Hematology, Jiangxi Academy of Clinical Medical Sciences, Nanchang, China
| |
Collapse
|
17
|
Rayasam A, Drobyski WR. Translational Clinical Strategies for the Prevention of Gastrointestinal Tract Graft Versus Host Disease. Front Immunol 2021; 12:779076. [PMID: 34899738 PMCID: PMC8662938 DOI: 10.3389/fimmu.2021.779076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Accepted: 11/08/2021] [Indexed: 11/15/2022] Open
Abstract
Graft versus host disease (GVHD) is the major non-relapse complication associated with allogeneic hematopoietic stem cell transplantation (HSCT). Unfortunately, GVHD occurs in roughly half of patients following this therapy and can induce severe life-threatening side effects and premature mortality. The pathophysiology of GVHD is driven by alloreactive donor T cells that induce a proinflammatory environment to cause pathological damage in the skin, gastrointestinal (GI) tract, lung, and liver during the acute phase of this disease. Recent work has demonstrated that the GI tract is a pivotal target organ and a primary driver of morbidity and mortality in patients. Prevention of this complication has therefore emerged as an important goal of prophylaxis strategies given the primacy of this tissue site in GVHD pathophysiology. In this review, we summarize foundational pre-clinical studies that have been conducted in animal models to prevent GI tract GVHD and examine the efficacy of these approaches upon subsequent translation into the clinic. Specifically, we focus on therapies designed to block inflammatory cytokine pathways, inhibit cellular trafficking of alloreactive donor T cells to the GI tract, and reconstitute impaired regulatory networks for the prevention of GVHD in the GI tract.
Collapse
Affiliation(s)
- Aditya Rayasam
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, United States
| | - William R Drobyski
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, United States.,Bone Marrow Transplant Program, Medical College of Wisconsin, Milwaukee, WI, United States
| |
Collapse
|
18
|
Mohamed FA, Thangavelu G, Rhee SY, Sage PT, O’Connor RS, Rathmell JC, Blazar BR. Recent Metabolic Advances for Preventing and Treating Acute and Chronic Graft Versus Host Disease. Front Immunol 2021; 12:757836. [PMID: 34712243 PMCID: PMC8546182 DOI: 10.3389/fimmu.2021.757836] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Accepted: 09/13/2021] [Indexed: 01/14/2023] Open
Abstract
The therapeutic efficacy of allogeneic hematopoietic stem cell transplantation (allo-HSCT) is limited by the development of graft-versus-host disease (GVHD). In GVHD, rigorous pre-conditioning regimen resets the immune landscape and inflammatory milieu causing immune dysregulation, characterized by an expansion of alloreactive cells and a reduction in immune regulatory cells. In acute GVHD (aGVHD), the release of damage- and pathogen- associated molecular patterns from damaged tissue caused by the conditioning regimen sets the stage for T cell priming, activation and expansion further exacerbating tissue injury and organ damage, particularly in the gastrointestinal tract. Studies have shown that donor T cells utilize multiple energetic and biosynthetic pathways to mediate GVHD that can be distinct from the pathways used by regulatory T cells for their suppressive function. In chronic GVHD (cGVHD), donor T cells may differentiate into IL-21 producing T follicular helper cells or tissue resident T helper cells that cooperate with germinal center B cells or memory B cells, respectively, to produce allo- and auto-reactive antibodies with subsequent tissue fibrosis. Alternatively, donor T cells can become IFN- γ/IL-17 cytokine expressing T cells that mediate sclerodermatous skin injury. Patients refractory to the first line standard regimens for GVHD treatment have a poor prognosis indicating an urgent need for new therapies to restore the balance between effector and regulatory immune cells while preserving the beneficial graft-versus-tumor effect. Emerging data points toward a role for metabolism in regulating these allo- and auto-immune responses. Here, we will discuss the preclinical and clinical data available on the distinct metabolic demands of acute and chronic GVHD and recent efforts in identifying therapeutic targets using metabolomics. Another dimension of this review will examine the changing microbiome after allo-HSCT and the role of microbial metabolites such as short chain fatty acids and long chain fatty acids on regulating immune responses. Lastly, we will examine the metabolic implications of coinhibitory pathway blockade and cellular therapies in allo-HSCT. In conclusion, greater understanding of metabolic pathways involved in immune cell dysregulation during allo-HSCT may pave the way to provide novel therapies to prevent and treat GVHD.
Collapse
Affiliation(s)
- Fathima A. Mohamed
- Department of Pediatrics, Division of Blood & Marrow Transplant & Cellular Therapy, University of Minnesota Cancer Center, Minneapolis, MN, United States
| | - Govindarajan Thangavelu
- Department of Pediatrics, Division of Blood & Marrow Transplant & Cellular Therapy, University of Minnesota Cancer Center, Minneapolis, MN, United States
| | - Stephanie Y. Rhee
- Department of Pediatrics, Division of Blood & Marrow Transplant & Cellular Therapy, University of Minnesota Cancer Center, Minneapolis, MN, United States
| | - Peter T. Sage
- Renal Division, Transplantation Research Center, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Roddy S. O’Connor
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Center for Cellular Immunotherapies, Perelman School of Medicine, Philadelphia, PA, United States
| | - Jeffrey C. Rathmell
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Bruce R. Blazar
- Department of Pediatrics, Division of Blood & Marrow Transplant & Cellular Therapy, University of Minnesota Cancer Center, Minneapolis, MN, United States
| |
Collapse
|
19
|
Tourret M, Talvard-Balland N, Lambert M, Ben Youssef G, Chevalier MF, Bohineust A, Yvorra T, Morin F, Azarnoush S, Lantz O, Dalle JH, Caillat-Zucman S. Human MAIT cells are devoid of alloreactive potential: prompting their use as universal cells for adoptive immune therapy. J Immunother Cancer 2021; 9:jitc-2021-003123. [PMID: 34615705 PMCID: PMC8496386 DOI: 10.1136/jitc-2021-003123] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/13/2021] [Indexed: 12/20/2022] Open
Abstract
Background Mucosal-associated invariant T (MAIT) cells are semi-invariant T cells that recognize microbial antigens presented by the highly conserved MR1 molecule. MAIT cells are predominantly localized in the liver and barrier tissues and are potent effectors of antimicrobial defense. MAIT cells are very few at birth and accumulate gradually over a period of about 6 years during the infancy. The cytotoxic potential of MAIT cells, as well as their newly described regulatory and tissue repair functions, open the possibility of exploiting their properties in adoptive therapy. A prerequisite for their use as ‘universal’ cells would be a lack of alloreactive potential, which remains to be demonstrated. Methods We used ex vivo, in vitro and in vivo models to determine if human MAIT cells contribute to allogeneic responses. Results We show that recovery of MAIT cells after allogeneic hematopoietic stem cell transplantation recapitulates their slow physiological expansion in early childhood, independent of recovery of non-MAIT T cells. In vitro, signals provided by allogeneic cells and cytokines do not induce sustained MAIT cell proliferation. In vivo, human MAIT cells do not expand nor accumulate in tissues in a model of T-cell-mediated xenogeneic graft-versus-host disease in immunodeficient mice. Conclusions Altogether, these results provide evidence that MAIT cells are devoid of alloreactive potential and pave the way for harnessing their translational potential in universal adoptive therapy overcoming barriers of HLA disparity. Trial registration number ClinicalTrials.gov number NCT02403089.
Collapse
Affiliation(s)
- Marie Tourret
- INSERM UMR976, Human Immunology, Pathophysiology and Immunotherapy, Université de Paris, Paris, France
| | - Nana Talvard-Balland
- INSERM UMR976, Human Immunology, Pathophysiology and Immunotherapy, Université de Paris, Paris, France
| | - Marion Lambert
- INSERM UMR976, Human Immunology, Pathophysiology and Immunotherapy, Université de Paris, Paris, France
| | - Ghada Ben Youssef
- INSERM UMR976, Human Immunology, Pathophysiology and Immunotherapy, Université de Paris, Paris, France
| | - Mathieu F Chevalier
- INSERM UMR976, Human Immunology, Pathophysiology and Immunotherapy, Université de Paris, Paris, France
| | - Armelle Bohineust
- INSERM UMR976, Human Immunology, Pathophysiology and Immunotherapy, Université de Paris, Paris, France
| | - Thomas Yvorra
- INSERM UMR3666/U1143, Université PSL, Institut Curie, Paris, France
| | - Florence Morin
- Laboratoire d'Immunologie, Hôpital Saint-Louis, Assistance Publique-Hôpitaux de Paris (AP-HP), Université de Paris, Paris, France
| | - Saba Azarnoush
- Département d'Immuno-Hématologie, Hôpital Robert Debré, AP-HP, Université de Paris, Paris, France
| | - Olivier Lantz
- INSERM U932, Université PSL, Institut Curie, Paris, France.,Laboratoire d'immunologie clinique & Centre d'investigation Clinique en Biothérapie (CIC-BT1428), Institut Curie, Paris, France
| | - Jean-Hugues Dalle
- INSERM UMR976, Human Immunology, Pathophysiology and Immunotherapy, Université de Paris, Paris, France.,Département d'Immuno-Hématologie, Hôpital Robert Debré, AP-HP, Université de Paris, Paris, France
| | - Sophie Caillat-Zucman
- INSERM UMR976, Human Immunology, Pathophysiology and Immunotherapy, Université de Paris, Paris, France .,Laboratoire d'Immunologie, Hôpital Saint-Louis, Assistance Publique-Hôpitaux de Paris (AP-HP), Université de Paris, Paris, France
| |
Collapse
|
20
|
Jiang H, Fu D, Bidgoli A, Paczesny S. T Cell Subsets in Graft Versus Host Disease and Graft Versus Tumor. Front Immunol 2021; 12:761448. [PMID: 34675938 PMCID: PMC8525316 DOI: 10.3389/fimmu.2021.761448] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Accepted: 09/13/2021] [Indexed: 01/04/2023] Open
Abstract
Allogeneic hematopoietic cell transplantation (allo-HCT) is an essential therapeutic modality for patients with hematological malignancies and other blood disorders. Unfortunately, acute graft-versus-host disease (aGVHD) remains a major source of morbidity and mortality following allo-HCT, which limits its use in a broader spectrum of patients. Chronic graft-versus-host disease (cGVHD) also remains the most common long-term complication of allo-HCT, occurring in reportedly 30-70% of patients surviving more than 100 days. Chronic GVHD is also the leading cause of non-relapse mortality (NRM) occurring more than 2 years after HCT for malignant disease. Graft versus tumor (GVT) is a major component of the overall beneficial effects of allogeneic HCT in the treatment of hematological malignancies. Better understanding of GVHD pathogenesis is important to identify new therapeutic targets for GVHD prevention and therapy. Emerging data suggest opposing roles for different T cell subsets, e.g., IFN-γ producing CD4+ and CD8+ T cells (Th1 and Tc1), IL-4 producing T cells (Th2 and Tc2), IL-17 producing T cells (Th17 and Tc17), IL-9 producing T cells (Th9 and Tc9), IL-22 producing T cells (Th22), T follicular helper cells (Tfh), regulatory T-cells (Treg) and tissue resident memory T cells (Trm) in GVHD and GVT etiology. In this review, we first summarize the general description of the cytokine signals that promote the differentiation of T cell subsets and the roles of these T cell subsets in the pathogenesis of GVHD. Next, we extensively explore preclinical findings of T cell subsets in both GVHD/GVT animal models and humans. Finally, we address recent findings about the roles of T-cell subsets in clinical GVHD and current strategies to modulate T-cell differentiation for treating and preventing GVHD in patients. Further exploring and outlining the immune biology of T-cell differentiation in GVHD that will provide more therapeutic options for maintaining success of allo-HCT.
Collapse
Affiliation(s)
| | | | | | - Sophie Paczesny
- Department of Microbiology and Immunology and Pediatrics, Medical University of South Carolina, Charleston, SC, United States
| |
Collapse
|
21
|
Fujiwara H. Crosstalk Between Intestinal Microbiota Derived Metabolites and Tissues in Allogeneic Hematopoietic Cell Transplantation. Front Immunol 2021; 12:703298. [PMID: 34512627 PMCID: PMC8429959 DOI: 10.3389/fimmu.2021.703298] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 08/09/2021] [Indexed: 12/12/2022] Open
Abstract
Allogeneic hematopoietic stem cell transplantation (allo-HSCT) is an evidence based- cellular immunotherapy for hematological malignancies. Immune reactions not only promote graft-versus-tumor effects that kill hematological malignant cells but also graft-versus-host disease (GVHD) that is the primary complication characterized by systemic organ damages consisting of T-cells and antigen presenting cells (APCs) activation. GVHD has long been recognized as an immunological reaction that requires an immunosuppressive treatment targeting immune cells. However immune suppression cannot always prevent GVHD or effectively treat it once it has developed. Recent studies using high-throughput sequencing technology investigated the impact of microbial flora on GVHD and provided profound insights of the mechanism of GVHD other than immune cells. Allo-HSCT affects the intestinal microbiota and microbiome-metabolome axis that can alter intestinal homeostasis and the severity of experimental GVHD. This axis can potentially be manipulated via dietary intervention or metabolites produced by intestinal bacteria affected post-allo-HSCT. In this review, we discuss the mechanism of experimental GVHD regulation by the complex microbial community-metabolites-host tissue axis. Furthermore, we summarize the major findings of microbiome-based immunotherapeutic approaches that protect tissues from experimental GVHD. Understanding the complex relationships between gut microbiota-metabolites-host tissues axis provides crucial insight into the pathogenesis of GVHD and advances the development of new therapeutic approaches.
Collapse
Affiliation(s)
- Hideaki Fujiwara
- Department of Hematology and Oncology, Okayama University Hospital, Okayama, Japan
| |
Collapse
|
22
|
Henden AS, Koyama M, Robb RJ, Forero A, Kuns RD, Chang K, Ensbey KS, Varelias A, Kazakoff SH, Waddell N, Clouston AD, Giri R, Begun J, Blazar BR, Degli-Esposti MA, Kotenko SV, Lane SW, Bowerman KL, Savan R, Hugenholtz P, Gartlan KH, Hill GR. IFN-λ therapy prevents severe gastrointestinal graft-versus-host disease. Blood 2021; 138:722-737. [PMID: 34436524 PMCID: PMC8667051 DOI: 10.1182/blood.2020006375] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Immunopathology and intestinal stem cell (ISC) loss in the gastrointestinal (GI) tract is the prima facie manifestation of graft-versus-host disease (GVHD) and is responsible for significant mortality after allogeneic bone marrow transplantation (BMT). Approaches to prevent GVHD to date focus on immune suppression. Here, we identify interferon-λ (IFN-λ; interleukin-28 [IL-28]/IL-29) as a key protector of GI GVHD immunopathology, notably within the ISC compartment. Ifnlr1-/- mice displayed exaggerated GI GVHD and mortality independent of Paneth cells and alterations to the microbiome. Ifnlr1-/- intestinal organoid growth was significantly impaired, and targeted Ifnlr1 deficiency exhibited effects intrinsic to recipient Lgr5+ ISCs and natural killer cells. PEGylated recombinant IL-29 (PEG-rIL-29) treatment of naive mice enhanced Lgr5+ ISC numbers and organoid growth independent of both IL-22 and type I IFN and modulated proliferative and apoptosis gene sets in Lgr5+ ISCs. PEG-rIL-29 treatment improved survival, reduced GVHD severity, and enhanced epithelial proliferation and ISC-derived organoid growth after BMT. The preservation of ISC numbers in response to PEG-rIL-29 after BMT occurred both in the presence and absence of IFN-λ-signaling in recipient natural killer cells. IFN-λ is therefore an attractive and rapidly testable approach to prevent ISC loss and immunopathology during GVHD.
Collapse
Affiliation(s)
- Andrea S Henden
- Bone Marrow Transplantation Laboratory, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
- Department of Haematology and Bone Marrow Transplantation, Royal Brisbane and Women's Hospital, Brisbane, QLD, Australia
- Faculty of Medicine, The University of Queensland, Herston, QLD, Australia
| | - Motoko Koyama
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Renee J Robb
- Bone Marrow Transplantation Laboratory, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Adriana Forero
- Center for Innate Immunity and Immune Disease, University of Washington, Seattle, WA
| | - Rachel D Kuns
- Bone Marrow Transplantation Laboratory, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Karshing Chang
- Bone Marrow Transplantation Laboratory, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Kathleen S Ensbey
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Antiopi Varelias
- Bone Marrow Transplantation Laboratory, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Stephen H Kazakoff
- Genetics and Computational Biology Department, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Nicole Waddell
- Genetics and Computational Biology Department, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | | | - Rabina Giri
- Mater Research Institute, The University of Queensland-Translational Research Institute, Brisbane, QLD, Australia
| | - Jakob Begun
- Mater Research Institute, The University of Queensland-Translational Research Institute, Brisbane, QLD, Australia
| | - Bruce R Blazar
- Division of Blood and Marrow Transplantation, Department of Pediatrics, University of Minnesota, Minneapolis, MN
| | - Mariapia A Degli-Esposti
- Centre for Experimental Immunology, Lions Eye Institute, Perth, WA, Australia
- Infection and Immunity Program, Department of Microbiology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Sergei V Kotenko
- Center for Immunity and Inflammation, New Jersey Medical School, and
- Department of Microbiology, Biochemistry and Molecular Genetics, Rutgers Biomedical and Health Sciences (RBHS), Newark, NJ
| | - Steven W Lane
- Cancer Program, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Kate L Bowerman
- Australian Centre for Ecogenomics, School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD, Australia; and
| | - Ram Savan
- Center for Innate Immunity and Immune Disease, University of Washington, Seattle, WA
| | - Philip Hugenholtz
- Australian Centre for Ecogenomics, School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD, Australia; and
| | - Kate H Gartlan
- Bone Marrow Transplantation Laboratory, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
- Faculty of Medicine, The University of Queensland, Herston, QLD, Australia
| | - Geoffrey R Hill
- Bone Marrow Transplantation Laboratory, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA
- Division of Medical Oncology, The University of Washington, Seattle, WA
| |
Collapse
|
23
|
Tang SQ, Yao WL, Wang YZ, Zhang YY, Zhao HY, Wen Q, Wang Y, Xu LP, Zhang XH, Huang XJ, Kong Y. Improved function and balance in T cell modulation by endothelial cells in young people. Clin Exp Immunol 2021; 206:196-207. [PMID: 34382213 DOI: 10.1111/cei.13654] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 07/25/2021] [Accepted: 08/06/2021] [Indexed: 11/26/2022] Open
Abstract
Elderly individuals exhibit unbalanced bone marrow (BM) effector T cell subset differentiation, such as increased T helper type 1 (Th1) and T cytotoxic type 1 (Tc1) cell frequencies, but the underlying mechanism is still unclear. Endothelial cells (ECs), which are instructive components of the BM microenvironment, exhibit the phenotype of semi-professional antigen-presenting cells and regulate T cell recruitment and activation. Thus, we compared the frequency and function of BM ECs, especially their capacity to regulate effector T cell subsets, between young and elderly healthy individuals, and explored the underlying mechanism of this immunomodulatory discrepancy. Although the young and elderly EC percentages were comparable, young ECs showed fewer reactive oxygen species and better migratory and tube-forming abilities than elderly ECs. Notably, increased T cell activation molecules and inflammatory cytokines were found in elderly ECs which regulated T cells to differentiate into more proinflammatory T cells, including Th1 and Tc1 cells, than young ECs.
Collapse
Affiliation(s)
- Shu-Qian Tang
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Collaborative Innovation Center of Hematology, Peking University, Beijing, China
| | - Wei-Li Yao
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Collaborative Innovation Center of Hematology, Peking University, Beijing, China
| | - Ya-Zhe Wang
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Collaborative Innovation Center of Hematology, Peking University, Beijing, China
| | - Yuan-Yuan Zhang
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Collaborative Innovation Center of Hematology, Peking University, Beijing, China
| | - Hong-Yan Zhao
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Collaborative Innovation Center of Hematology, Peking University, Beijing, China
| | - Qi Wen
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Collaborative Innovation Center of Hematology, Peking University, Beijing, China
| | - Yu Wang
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Collaborative Innovation Center of Hematology, Peking University, Beijing, China
| | - Lan-Ping Xu
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Collaborative Innovation Center of Hematology, Peking University, Beijing, China
| | - Xiao-Hui Zhang
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Collaborative Innovation Center of Hematology, Peking University, Beijing, China
| | - Xiao-Jun Huang
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Collaborative Innovation Center of Hematology, Peking University, Beijing, China
| | - Yuan Kong
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Collaborative Innovation Center of Hematology, Peking University, Beijing, China
| |
Collapse
|
24
|
McDaniel Mims B, Enriquez J, Pires dos Santos A, Jones-Hall Y, Dowd S, Furr KL, Grisham MB. Antibiotic administration exacerbates acute graft vs. host disease-induced bone marrow and spleen damage in lymphopenic mice. PLoS One 2021; 16:e0254845. [PMID: 34358240 PMCID: PMC8346256 DOI: 10.1371/journal.pone.0254845] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Accepted: 07/04/2021] [Indexed: 12/29/2022] Open
Abstract
Background Hematopoietic stem cell transplantation is a potential cure for certain life-threatening malignant and nonmalignant diseases. However, experimental and clinical studies have demonstrated that pre-transplant myeloablative conditioning damages the gut leading to translocation of intestinal bacteria and the development of acute graft vs. host disease (aGVHD). The overall objective of this study was to determine whether administration of broad spectrum antibiotics (Abx) affects the onset and/or severity of aGVHD in lymphopenic mice that were not subjected to toxic, pre-transplant conditioning. Results We found that treatment of NK cell-depleted recombination activating gene-1-deficient (-NK/RAG) recipients with an Abx cocktail containing vancomycin and neomycin for 7 days prior to and 4 weeks following adoptive transfer of allogeneic CD4+ T cells, exacerbated the development of aGVHD-induced BM failure and spleen damage when compared to untreated–NK/RAG recipients engrafted with syngeneic or allogeneic T cells. Abx-treated mice exhibited severe anemia and monocytopenia as well as marked reductions in BM- and spleen-residing immune cells. Blinded histopathological analysis confirmed that Abx-treated mice engrafted with allogeneic T cells suffered significantly more damage to the BM and spleen than did untreated mice engrafted with allogeneic T cells. Abx-induced exacerbation of BM and spleen damage correlated with a dramatic reduction in fecal bacterial diversity, marked loss of anaerobic bacteria and remarkable expansion of potentially pathogenic bacteria. Conclusions We conclude that continuous Abx treatment may aggravate aGVHD-induced tissue damage by reducing short chain fatty acid-producing anaerobes (e.g. Clostridium, Blautia) and/or by promoting the expansion of pathobionts (e.g. Akkermansia) and opportunistic pathogens (Cronobacter).
Collapse
Affiliation(s)
- Brianyell McDaniel Mims
- Department of Immunology and Molecular Microbiology, Texas Tech University Health Sciences Center, Lubbock, TX, United States of America
| | - Josue Enriquez
- Department of Immunology and Molecular Microbiology, Texas Tech University Health Sciences Center, Lubbock, TX, United States of America
| | - Andrea Pires dos Santos
- College of Veterinary Medicine, Department of Comparative Pathobiology, Purdue University, West Lafayette, IN, United States of America
| | - Yava Jones-Hall
- College of Veterinary Medicine and Biomedical Sciences, Department of Veterinary Pathobiology, Texas A&M University, College Station, TX, United States of America
| | - Scot Dowd
- MR DNA (Molecular Research), Shallowater, TX, United States of America
| | - Kathryn L. Furr
- Department of Immunology and Molecular Microbiology, Texas Tech University Health Sciences Center, Lubbock, TX, United States of America
| | - Matthew B. Grisham
- Department of Immunology and Molecular Microbiology, Texas Tech University Health Sciences Center, Lubbock, TX, United States of America
- * E-mail:
| |
Collapse
|
25
|
Ehx G, Ritacco C, Hannon M, Dubois S, Delens L, Willems E, Servais S, Drion P, Beguin Y, Baron F. Comprehensive analysis of the immunomodulatory effects of rapamycin on human T cells in graft-versus-host disease prophylaxis. Am J Transplant 2021; 21:2662-2674. [PMID: 33512760 DOI: 10.1111/ajt.16505] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 12/24/2020] [Accepted: 01/18/2021] [Indexed: 01/25/2023]
Abstract
Graft-versus-host disease (GVHD) is a major cause of toxicity after allogeneic hematopoietic cell transplantation (allo-HCT). While rapamycin (RAPA) is commonly used in GVHD prophylaxis in combination with a calcineurin inhibitor (CNI), the understanding of its mechanism of action on human T cells is still incomplete. Here, we performed an extensive analysis of RAPA effects on human T cells in a humanized mouse model of GVHD, in ex-vivo T cell cultures and in patients given RAPA plus tacrolimus as GVHD prophylaxis after nonmyeloablative allo-HCT. We demonstrate that RAPA mitigates GVHD by decreasing T cell engraftment and differentiation, inhibiting CD8+ T cell activation and increasing the long-term IL-2 secretion, thereby supporting regulatory T cell (Treg) proliferation. In contrast, graft-versus-leukemia effects were not abrogated, as RAPA-treated T cells had increased resistance to apoptosis and retained their effector function and proliferative capacity upon re-stimulation. Importantly, we found that RAPA impact on Treg and CD8+ T cells was closely dependent upon IL-2 signaling and that therapeutic options interfering with IL-2, such as calcineurin inhibitors, antagonize the IL-2-dependent promotion of Treg mediated by RAPA. Our results suggest that RAPA immunological efficacy could be improved in combination with drugs having possible synergistic effects such as the hypomethylating agent 5-azacytidine.
Collapse
Affiliation(s)
- Grégory Ehx
- Groupe Interdisciplinaire de Génoprotéomique Appliquée (GIGA)-I3, University of Liège, Liège, Belgium
| | - Caroline Ritacco
- Groupe Interdisciplinaire de Génoprotéomique Appliquée (GIGA)-I3, University of Liège, Liège, Belgium
| | - Muriel Hannon
- Groupe Interdisciplinaire de Génoprotéomique Appliquée (GIGA)-I3, University of Liège, Liège, Belgium
| | - Sophie Dubois
- Groupe Interdisciplinaire de Génoprotéomique Appliquée (GIGA)-I3, University of Liège, Liège, Belgium
| | - Loic Delens
- Groupe Interdisciplinaire de Génoprotéomique Appliquée (GIGA)-I3, University of Liège, Liège, Belgium
| | - Evelyne Willems
- Department of Medicine, Division of Hematology, CHU and University of Liège, Liège, Belgium
| | - Sophie Servais
- Groupe Interdisciplinaire de Génoprotéomique Appliquée (GIGA)-I3, University of Liège, Liège, Belgium.,Department of Medicine, Division of Hematology, CHU and University of Liège, Liège, Belgium
| | - Pierre Drion
- Experimental Surgery, GIGA-R & Credec, University of Liège, Liège, Belgium
| | - Yves Beguin
- Groupe Interdisciplinaire de Génoprotéomique Appliquée (GIGA)-I3, University of Liège, Liège, Belgium.,Department of Medicine, Division of Hematology, CHU and University of Liège, Liège, Belgium
| | - Frédéric Baron
- Groupe Interdisciplinaire de Génoprotéomique Appliquée (GIGA)-I3, University of Liège, Liège, Belgium.,Department of Medicine, Division of Hematology, CHU and University of Liège, Liège, Belgium
| |
Collapse
|
26
|
Tkachev V, Kaminski J, Potter EL, Furlan SN, Yu A, Hunt DJ, McGuckin C, Zheng H, Colonna L, Gerdemann U, Carlson J, Hoffman M, Olvera J, English C, Baldessari A, Panoskaltsis-Mortari A, Watkins B, Qayed M, Suessmuth Y, Betz K, Bratrude B, Langston A, Horan JT, Ordovas-Montanes J, Shalek AK, Blazar BR, Roederer M, Kean LS. Spatiotemporal single-cell profiling reveals that invasive and tissue-resident memory donor CD8 + T cells drive gastrointestinal acute graft-versus-host disease. Sci Transl Med 2021; 13:13/576/eabc0227. [PMID: 33441422 DOI: 10.1126/scitranslmed.abc0227] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Accepted: 12/11/2020] [Indexed: 12/11/2022]
Abstract
Organ infiltration by donor T cells is critical to the development of acute graft-versus-host disease (aGVHD) in recipients after allogeneic hematopoietic stem cell transplant (allo-HCT). However, deconvoluting the transcriptional programs of newly recruited donor T cells from those of tissue-resident T cells in aGVHD target organs remains a challenge. Here, we combined the serial intravascular staining technique with single-cell RNA sequencing to dissect the tightly connected processes by which donor T cells initially infiltrate tissues and then establish a pathogenic tissue residency program in a rhesus macaque allo-HCT model that develops aGVHD. Our results enabled creation of a spatiotemporal map of the transcriptional programs controlling donor CD8+ T cell infiltration into the primary aGVHD target organ, the gastrointestinal (GI) tract. We identified the large and small intestines as the only two sites demonstrating allo-specific, rather than lymphodepletion-driven, T cell infiltration. GI-infiltrating donor CD8+ T cells demonstrated a highly activated, cytotoxic phenotype while simultaneously developing a canonical tissue-resident memory T cell (TRM) transcriptional signature driven by interleukin-15 (IL-15)/IL-21 signaling. We found expression of a cluster of genes directly associated with tissue invasiveness, including those encoding adhesion molecules (ITGB2), specific chemokines (CCL3 and CCL4L1) and chemokine receptors (CD74), as well as multiple cytoskeletal proteins. This tissue invasion transcriptional signature was validated by its ability to discriminate the CD8+ T cell transcriptome of patients with GI aGVHD from those of GVHD-free patients. These results provide insights into the mechanisms controlling tissue occupancy of target organs by pathogenic donor CD8+ TRM cells during aGVHD in primate transplant recipients.
Collapse
Affiliation(s)
- Victor Tkachev
- Division of Pediatric Hematology/Oncology, Boston Children's Hospital, Department of Pediatric Oncology, Dana-Farber Cancer Institute, Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA.
| | - James Kaminski
- Division of Pediatric Hematology/Oncology, Boston Children's Hospital, Department of Pediatric Oncology, Dana-Farber Cancer Institute, Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA.,Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - E Lake Potter
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20858, USA
| | - Scott N Furlan
- Fred Hutchinson Cancer Research Center, Department of Pediatrics, University of Washington, Seattle, WA 98109, USA
| | - Alison Yu
- Division of Pediatric Hematology/Oncology, Boston Children's Hospital, Department of Pediatric Oncology, Dana-Farber Cancer Institute, Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Daniel J Hunt
- Division of Pediatric Hematology/Oncology, Boston Children's Hospital, Department of Pediatric Oncology, Dana-Farber Cancer Institute, Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Connor McGuckin
- Division of Pediatric Hematology/Oncology, Boston Children's Hospital, Department of Pediatric Oncology, Dana-Farber Cancer Institute, Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Hengqi Zheng
- University of Washington, Seattle, WA 98195, USA
| | - Lucrezia Colonna
- Fred Hutchinson Cancer Research Center, Department of Pediatrics, University of Washington, Seattle, WA 98109, USA
| | - Ulrike Gerdemann
- Division of Pediatric Hematology/Oncology, Boston Children's Hospital, Department of Pediatric Oncology, Dana-Farber Cancer Institute, Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | | | - Michelle Hoffman
- Fred Hutchinson Cancer Research Center, Department of Pediatrics, University of Washington, Seattle, WA 98109, USA
| | - Joe Olvera
- Division of Pediatric Hematology/Oncology, Boston Children's Hospital, Department of Pediatric Oncology, Dana-Farber Cancer Institute, Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Chris English
- Washington National Primate Research Center, Seattle, WA 98195, USA
| | | | - Angela Panoskaltsis-Mortari
- Division of Blood and Marrow Transplantation, Department of Pediatrics, Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55454, USA
| | | | - Muna Qayed
- Emory University School of Medicine, Atlanta, GA 30322, USA
| | | | - Kayla Betz
- Division of Pediatric Hematology/Oncology, Boston Children's Hospital, Department of Pediatric Oncology, Dana-Farber Cancer Institute, Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Brandi Bratrude
- Division of Pediatric Hematology/Oncology, Boston Children's Hospital, Department of Pediatric Oncology, Dana-Farber Cancer Institute, Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | | | - John T Horan
- Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Jose Ordovas-Montanes
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA.,Division of Gastroenterology, Boston Children's Hospital and Program in Immunology, Harvard Medical School, Boston, MA 02115, USA.,Harvard Stem Cell Institute, Cambridge, MA 02138, USA
| | - Alex K Shalek
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA.,Institute for Medical Engineering and Science (IMES), Department of Chemistry, and Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA 02142, USA.,Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA
| | - Bruce R Blazar
- Division of Blood and Marrow Transplantation, Department of Pediatrics, Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55454, USA
| | - Mario Roederer
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20858, USA
| | - Leslie S Kean
- Division of Pediatric Hematology/Oncology, Boston Children's Hospital, Department of Pediatric Oncology, Dana-Farber Cancer Institute, Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
27
|
Haring E, Zeiser R, Apostolova P. Interfering With Inflammation: Heterogeneous Effects of Interferons in Graft- Versus-Host Disease of the Gastrointestinal Tract and Inflammatory Bowel Disease. Front Immunol 2021; 12:705342. [PMID: 34249014 PMCID: PMC8264264 DOI: 10.3389/fimmu.2021.705342] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 06/10/2021] [Indexed: 12/14/2022] Open
Abstract
The intestine can be the target of several immunologically mediated diseases, including graft-versus-host disease (GVHD) and inflammatory bowel disease (IBD). GVHD is a life-threatening complication that occurs after allogeneic hematopoietic stem cell transplantation. Involvement of the gastrointestinal tract is associated with a particularly high mortality. GVHD development starts with the recognition of allo-antigens in the recipient by the donor immune system, which elicits immune-mediated damage of otherwise healthy tissues. IBD describes a group of immunologically mediated chronic inflammatory diseases of the intestine. Several aspects, including genetic predisposition and immune dysregulation, are responsible for the development of IBD, with Crohn’s disease and ulcerative colitis being the two most common variants. GVHD and IBD share multiple key features of their onset and development, including intestinal tissue damage and loss of intestinal barrier function. A further common feature in the pathophysiology of both diseases is the involvement of cytokines such as type I and II interferons (IFNs), amongst others. IFNs are a family of protein mediators produced as a part of the inflammatory response, typically to pathogens or malignant cells. Diverse, and partially paradoxical, effects have been described for IFNs in GVHD and IBD. This review summarizes current knowledge on the role of type I, II and III IFNs, including basic concepts and controversies about their functions in the context of GVHD and IBD. In addition, therapeutic options, research developments and remaining open questions are addressed.
Collapse
Affiliation(s)
- Eileen Haring
- Department of Medicine I, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,German Cancer Consortium (DKTK), Partner Site Freiburg and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Robert Zeiser
- Department of Medicine I, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,German Cancer Consortium (DKTK), Partner Site Freiburg and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Petya Apostolova
- Department of Medicine I, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,German Cancer Consortium (DKTK), Partner Site Freiburg and German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
28
|
Update in clinical and mouse microbiota research in allogeneic haematopoietic cell transplantation. Curr Opin Hematol 2021; 27:360-367. [PMID: 33003084 DOI: 10.1097/moh.0000000000000616] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
PURPOSE OF REVIEW The intestinal microbiota plays a critical role in intestinal homeostasis and immune regulation and has been recognized as a predictor of clinical outcome in patients undergoing allogeneic haematopoietic cell transplantation (allo-HCT) and specifically a determinant of the severity of graft-versus-host disease (GVHD) in mouse models. As GVHD is the most important cause of nonrelapse mortality (NRM) after allo-HCT, understanding the mechanisms by which modifying the microbiota may prevent or decrease the severity of GVHD would represent an important advance. RECENT FINDINGS Microbiota injury was observed globally and higher diversity at peri-engraftment was associated with lower mortality. Lactose is a dietary factor that promotes post-allo-HCT Enterococcus expansion, which is itself associated with mortality from GVHD in patients and exacerbates GVHD in mice. Bacterial and fungal bloodstream infections are preceded by intestinal colonization with a corresponding organism, supporting the gut as a source for many bloodstream infections. Metabolomic profiling studies showed that GVHD is associated with changes in faecal and plasma microbiota-derived molecules. SUMMARY In this review, we highlight some of the most recent and important findings in clinical and mouse microbiota research, as it relates to allo-HCT. Many of these are already being translated into clinical trials that have the potential to change future practice in the care of patients.
Collapse
|
29
|
Takahashi H, Okayama N, Yamaguchi N, Nomura M, Miyahara Y, Mahbub MH, Hase R, Morishima Y, Suehiro Y, Yamasaki T, Tamada K, Takahashi S, Tojo A, Tanabe T. Analysis of Relationships between Immune Checkpoint and Methylase Gene Polymorphisms and Outcomes after Unrelated Bone Marrow Transplantation. Cancers (Basel) 2021; 13:cancers13112752. [PMID: 34206082 PMCID: PMC8199545 DOI: 10.3390/cancers13112752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 05/25/2021] [Accepted: 05/26/2021] [Indexed: 11/23/2022] Open
Abstract
Simple Summary Hematopoietic stem-cell transplantation (HSCT) is a curative therapy for blood disorders. Unrelated bone marrow transplantation (uBMT) is a type of allogeneic HSCT that uses the bone marrow of an unrelated donor. While HLA mismatch is a risk factor for poor outcomes in HSCT, such as graft-versus-host disease (GVHD), the importance of non-HLA single-nucleotide polymorphisms (SNPs) remains unclear. The clinical application of immune checkpoint and chromatin methylation inhibitors to cancer has been attracting attention. In the present study, we retrospectively genotyped five SNPs in four immune checkpoint genes, BTLA, PD-1, LAG3, and CTLA4, and two SNPs in methylase genes, DNMT1 and EZH2, in 999 uBMT pairs. Although no correlations were observed between these SNPs and post-uBMT outcomes, recipient EZH2 SNP exhibited a low p-value in the analysis of grade 2–4 acute GVHD (p = 0.010). This SNP may be useful for outcome predictions and needs to be confirmed in a larger-scale study. Abstract Unrelated bone marrow transplantation (uBMT) is performed to treat blood disorders, and it uses bone marrow from an unrelated donor as the transplant source. Although the importance of HLA matching in uBMT has been established, that of other genetic factors, such as single-nucleotide polymorphisms (SNPs), remains unclear. The application of immunoinhibitory receptors as anticancer drugs has recently been attracting attention. This prompted us to examine the importance of immunoinhibitory receptor SNPs in uBMT. We retrospectively genotyped five single-nucleotide polymorphisms (SNPs) in the immune checkpoint genes, BTLA, PD-1, LAG3, and CTLA4, and two SNPs in the methylase genes, DNMT1 and EZH2, in 999 uBMT donor–recipient pairs coordinated through the Japan Marrow Donor Program matched at least at HLA-A, -B, and -DRB1. No correlations were observed between these SNPs and post-uBMT outcomes (p > 0.005). This result questions the usefulness of these immune checkpoint gene polymorphisms for predicting post-BMT outcomes. However, the recipient EZH2 histone methyltransferase gene SNP, which encodes the D185H substitution, exhibited a low p-value in regression analysis of grade 2–4 acute graft-versus-host disease (p = 0.010). Due to a low minor allele frequency, this SNP warrants further investigation in a larger-scale study.
Collapse
Affiliation(s)
- Hidekazu Takahashi
- Department of Public Health and Preventive Medicine, Yamaguchi University Graduate School of Medicine, Ube 755-8505, Japan; (H.T.); (N.Y.); (M.N.); (M.M.); (R.H.)
| | - Naoko Okayama
- Division of Laboratory, Yamaguchi University Hospital, Ube 755-8505, Japan; (N.O.); (Y.M.); (T.Y.)
| | - Natsu Yamaguchi
- Department of Public Health and Preventive Medicine, Yamaguchi University Graduate School of Medicine, Ube 755-8505, Japan; (H.T.); (N.Y.); (M.N.); (M.M.); (R.H.)
| | - Moe Nomura
- Department of Public Health and Preventive Medicine, Yamaguchi University Graduate School of Medicine, Ube 755-8505, Japan; (H.T.); (N.Y.); (M.N.); (M.M.); (R.H.)
| | - Yuta Miyahara
- Division of Laboratory, Yamaguchi University Hospital, Ube 755-8505, Japan; (N.O.); (Y.M.); (T.Y.)
| | - MH Mahbub
- Department of Public Health and Preventive Medicine, Yamaguchi University Graduate School of Medicine, Ube 755-8505, Japan; (H.T.); (N.Y.); (M.N.); (M.M.); (R.H.)
| | - Ryosuke Hase
- Department of Public Health and Preventive Medicine, Yamaguchi University Graduate School of Medicine, Ube 755-8505, Japan; (H.T.); (N.Y.); (M.N.); (M.M.); (R.H.)
| | - Yasuo Morishima
- Division of Epidemiology and Prevention, Aichi Cancer Center Research Institute, Nagoya 464-8681, Japan;
| | - Yutaka Suehiro
- Department of Oncology and Laboratory Medicine, Yamaguchi University Graduate School of Medicine, Ube 755-8505, Japan;
| | - Takahiro Yamasaki
- Division of Laboratory, Yamaguchi University Hospital, Ube 755-8505, Japan; (N.O.); (Y.M.); (T.Y.)
- Department of Oncology and Laboratory Medicine, Yamaguchi University Graduate School of Medicine, Ube 755-8505, Japan;
| | - Koji Tamada
- Department of Immunology, Yamaguchi University Graduate School of Medicine, Ube 755-8505, Japan;
| | - Satoshi Takahashi
- Department of Hematology and Oncology, Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan;
| | - Arinobu Tojo
- Tokyo Medical and Dental University, Tokyo 113-8510, Japan;
| | - Tsuyoshi Tanabe
- Department of Public Health and Preventive Medicine, Yamaguchi University Graduate School of Medicine, Ube 755-8505, Japan; (H.T.); (N.Y.); (M.N.); (M.M.); (R.H.)
- Correspondence:
| |
Collapse
|
30
|
Imran MM, Ahmad U, Usman U, Ali M, Shaukat A, Gul N. Neutrophil/lymphocyte ratio-A marker of COVID-19 pneumonia severity. Int J Clin Pract 2021; 75:e13698. [PMID: 32892477 DOI: 10.1111/ijcp.13698] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Accepted: 12/26/2020] [Indexed: 01/07/2023] Open
Abstract
AIM To determine the efficacy of neutrophil/lymphocyte ratio (NLR) as a marker of the severity of COVID-19 pneumonia in the South-Asian population. METHODS This was a prospective, cross-sectional, analytic study conducted at HDU/ICU of District Headquarter Hospital, Faisalabad, Pakistan, from May through July 2020. Sixty-three eligible patients, admitted to the HDU/ICU, were prospectively enrolled in the study. Their NLR, C-reactive protein, serum albumin and serum fibrinogen were measured. Patients' demographic characteristics, comorbidities, clinical manifestations of COVID-19 infection, medication use and history of lung malignancy were retrieved from their medical history. Patients were categorised into either a general group (with mild COVID-19) or a heavy group (with moderate to severe COVID-19). RESULTS There were significant differences between the two groups in diabetes prevalence, NLR, C-reactive protein and serum albumin. NLR and C-reactive protein were positively correlated (P < .001, P = .04, respectively) whereas serum albumin was negatively correlated (P = .009) with severe COVID-19. NLR was found to be an independent risk factor for severe COVID-19 pneumonia in the heavy group (OR = 1.264, 95% CI: 1.046~1.526, P = .015). The calculated AUC using ROC for NLR was 0.831, with an optimal limit of 4.795, sensitivity of 0.83 and specificity of 0.75, which is highly suggestive of NLR being a marker for the early detection of deteriorating severe COVID-19 infection. CONCLUSION NLR can be used as an early warning signal for deteriorating severe COVID-19 infection and can provide an objective basis for early identification and management of severe COVID-19 pneumonia.
Collapse
Affiliation(s)
- Mehr Muhammad Imran
- Department of Pulmonology, District Headquarter Hospital, Faisalabad, Pakistan
| | - Umair Ahmad
- Department of Medicine, Allied Hospital, Faisalabad, Pakistan
- Medical Unit II, Faisalabad Medical University, Faisalabad, Pakistan
| | - Umer Usman
- Department of Pulmonology, District Headquarter Hospital, Faisalabad, Pakistan
- Medical Unit II, Faisalabad Medical University, Faisalabad, Pakistan
| | - Majid Ali
- College of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia
- Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA, Australia
| | - Aamir Shaukat
- Department of Pulmonology, District Headquarter Hospital, Faisalabad, Pakistan
| | - Noor Gul
- Department of Pulmonology, District Headquarter Hospital, Faisalabad, Pakistan
| |
Collapse
|
31
|
Liu W, Zhou N, Liu Y, Zhang W, Li X, Wang Y, Zheng R, Zhang Y. Mesenchymal stem cell exosome-derived miR-223 alleviates acute graft-versus-host disease via reducing the migration of donor T cells. Stem Cell Res Ther 2021; 12:153. [PMID: 33637123 PMCID: PMC7913292 DOI: 10.1186/s13287-021-02159-2] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 01/12/2021] [Indexed: 12/11/2022] Open
Abstract
Background Mesenchymal stem cells (MSCs) have been utilized in treating acute graft-versus-host disease (aGvHD) as they show strong immunosuppressive capacity through the release of various mediators, including immunosuppressive molecules, growth factors, chemokines, and exosomes. MicroRNAs (miRNAs) derived from MSC exosomes (MSCs-Exo) play a critical role in the regulation of immune responses. However, the function of miRNAs in treating aGvHD remains unknown. Here, we performed expression profiling of exosome-miRNAs from human umbilical cord MSCs (huc-MSCs) and murine compact bone MSCs (mb-MSCs) to investigate their immunoregulation effects in aGvHD. Methods Huc-MSCs-Exo and mb-MSCs-Exo were isolated and constructed MSCs-Exo-derived miRNA expression profiling using high-throughput sequencing. High expression of miR-223 was identified in both kinds of MSCs-Exo by bioinformatics analysis and quantitative real-time PCR (qPCR). In vitro cell crawling assay, transmigration assay and adhesion assay were subsequently applied to investigate the regulation of miR-223 on T cells. MiR-223 target gene was analyzed by western blot, luciferase analysis, and qPCR. Moreover, murine aGvHD model was established by infusing splenocytes and bone marrow nuclear cells from C57BL/6j mice (H-2Kb) into BALB/c recipient mice (H-2Kd). For therapeutic effect, MSCs or miR-223 Agomir were injected via tail vein. The general conditions of the mice in each group were monitored. Hematoxylin-eosin (H&E) staining was used to detect pathological changes of mice spleen, liver, and intestine. Mechanistically, immunofluorescence and flow cytometry were used to evaluate donor T cell migration, and enzyme-linked immunosorbent assay (ELISA) was used to detect the expression of serum inflammatory cytokines IFN-γ, TNF-α, and IL-17. Results High-throughput sequencing revealed high expression of miR-223 in huc-MSCs-Exo and mb-MSCs-Exo. MiR-223 could restrain adhesion and migration of T cells by inhibiting ICAM-1 expression in mouse lymphatic endothelial cells. MiR-223Agomir infusion attenuated aGvHD clinical symptoms, reduced the donor T cell infiltration into the spleen, liver, and intestine, and decreased inflammatory cytokines IFN-γ, TNF-α, and IL-17. Conclusion MSCs-Exo-derived miR-223 could attenuate aGvHD in mice through decreasing donor T cell migration. Our results unveil a new role of MSCs-Exo containing miR-223 in the treatment of aGvHD. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-021-02159-2.
Collapse
Affiliation(s)
- Weijiang Liu
- Department of Experimental Hematology and Biochemistry, Beijing Institute of Radiation Medicine, 27 Taiping Road, Beijing, 100085, People's Republic of China
| | - Na Zhou
- Department of Experimental Hematology and Biochemistry, Beijing Institute of Radiation Medicine, 27 Taiping Road, Beijing, 100085, People's Republic of China.,Department of Pediatrics, Tianjin Medical University General Hospital, 154 Anshan Road, Tianjin, 300052, People's Republic of China
| | - Yuanlin Liu
- Department of Experimental Hematology and Biochemistry, Beijing Institute of Radiation Medicine, 27 Taiping Road, Beijing, 100085, People's Republic of China
| | - Wei Zhang
- Department of Experimental Hematology and Biochemistry, Beijing Institute of Radiation Medicine, 27 Taiping Road, Beijing, 100085, People's Republic of China.,Department of Medical Administration, The Sixth Medical Center of PLA General Hospital, Beijing, 100048, China
| | - Xue Li
- Department of Experimental Hematology and Biochemistry, Beijing Institute of Radiation Medicine, 27 Taiping Road, Beijing, 100085, People's Republic of China
| | - Yang Wang
- Department of Experimental Hematology and Biochemistry, Beijing Institute of Radiation Medicine, 27 Taiping Road, Beijing, 100085, People's Republic of China
| | - Rongxiu Zheng
- Department of Pediatrics, Tianjin Medical University General Hospital, 154 Anshan Road, Tianjin, 300052, People's Republic of China.
| | - Yi Zhang
- Department of Experimental Hematology and Biochemistry, Beijing Institute of Radiation Medicine, 27 Taiping Road, Beijing, 100085, People's Republic of China.
| |
Collapse
|
32
|
Sofi MH, Wu Y, Ticer T, Schutt S, Bastian D, Choi HJ, Tian L, Mealer C, Liu C, Westwater C, Armeson KE, Alekseyenko AV, Yu XZ. A single strain of Bacteroides fragilis protects gut integrity and reduces GVHD. JCI Insight 2021; 6:136841. [PMID: 33554953 PMCID: PMC7934839 DOI: 10.1172/jci.insight.136841] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Accepted: 12/16/2020] [Indexed: 12/20/2022] Open
Abstract
Graft-versus-host disease (GVHD) is a pathological process caused by an exaggerated donor lymphocyte response to host antigens after allogeneic hematopoietic cell transplantation (allo-HCT). Donor T cells undergo extensive clonal expansion and differentiation, which culminate in damage to recipient target organs. Damage to the gastrointestinal tract is a main contributor to morbidity and mortality. The loss of diversity among intestinal bacteria caused by pretransplant conditioning regimens leads to an outgrowth of opportunistic pathogens and exacerbated GVHD after allo-HCT. Using murine models of allo-HCT, we found that an increase of Bacteroides in the intestinal microbiota of the recipients was associated with reduced GVHD in mice given fecal microbial transplantation. Administration of Bacteroides fragilis through oral gavage increased gut microbiota diversity and beneficial commensal bacteria and significantly ameliorated acute and chronic GVHD development. Preservation of gut integrity following B. fragilis exposure was likely attributed to increased short chain fatty acids, IL-22, and regulatory T cells, which in turn improved gut tight junction integrity and reduced inflammatory cytokine production of pathogenic T cells. The current study provides a proof of concept that a single strain of commensal bacteria can be a safe and effective means to protect gut integrity and ameliorate GVHD after allo-HCT.
Collapse
Affiliation(s)
- M Hanief Sofi
- Department of Microbiology and Immunology, Hollings Cancer Center, College of Medicine, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Yongxia Wu
- Department of Microbiology and Immunology, Hollings Cancer Center, College of Medicine, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Taylor Ticer
- Department of Microbiology and Immunology, Hollings Cancer Center, College of Medicine, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Steven Schutt
- Department of Microbiology and Immunology, Hollings Cancer Center, College of Medicine, Medical University of South Carolina, Charleston, South Carolina, USA
| | - David Bastian
- Department of Microbiology and Immunology, Hollings Cancer Center, College of Medicine, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Hee-Jin Choi
- Department of Microbiology and Immunology, Hollings Cancer Center, College of Medicine, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Linlu Tian
- Department of Microbiology and Immunology, Hollings Cancer Center, College of Medicine, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Corey Mealer
- Department of Microbiology and Immunology, Hollings Cancer Center, College of Medicine, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Chen Liu
- Department of Pathology, Yale School of Medicine, New Haven, Connecticut, USA
| | - Caroline Westwater
- Department of Microbiology and Immunology, Hollings Cancer Center, College of Medicine, Medical University of South Carolina, Charleston, South Carolina, USA.,Department of Oral Health Sciences, College of Dental Medicine, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Kent E Armeson
- Biomedical Informatics Center and Department of Public Health Sciences, College of Medicine, and Department of Healthcare Leadership & Management, College of Public Health Sciences, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Alexander V Alekseyenko
- Department of Oral Health Sciences, College of Dental Medicine, Medical University of South Carolina, Charleston, South Carolina, USA.,Biomedical Informatics Center and Department of Public Health Sciences, College of Medicine, and Department of Healthcare Leadership & Management, College of Public Health Sciences, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Xue-Zhong Yu
- Department of Microbiology and Immunology, Hollings Cancer Center, College of Medicine, Medical University of South Carolina, Charleston, South Carolina, USA
| |
Collapse
|
33
|
Abstract
OBJECTIVES Villous atrophy (VA) is not pathognomonic of celiac disease (CD). We aimed at reporting distribution, clinical, and immunohistochemical features of seronegative VA (SNVA) in a pediatric population. METHODS We retrospectively collected data from patients who underwent intestinal biopsies between 2010 and 2017 and showed VA without serum CD-associated autoantibodies. Marsh-Oberhuber grading was used. Density of intraepithelial lymphocytes (IELs) expressing CD3 or TCRγδ+ receptor and of lamina propria CD25+ cells was assessed by immunohistochemistry. Intestinal deposits of anti-tissue tranglutaminase2 (anti-TG2) were also investigated by double immunofluorescence. RESULTS Over a 7-year period, 64 out of 1282 patients with VA had negative serum CD serology. Diagnoses were: inflammatory bowel diseases (IBD) (21/64), Gastro-Esophageal Reflux Disease (GERD) (12/64), food allergy (8/64), infections (7/64, of which 3 HIV infections), immune deficiency (3/64), short bowel syndrome (3/64), congenital diarrhea (2/64), other/inconclusive diagnosis (8/64). Forty-four, 15, and 5 showed Marsh 3a, 3b, and 3c lesion, respectively. The latter category included 2 patients with Crohn disease, 2 with immunodeficiencies, 1 with lymphohistiocytosis. In 41/46 (89%) patients, mononuclear CD25+ cells were above the cut-off, indicating mucosal inflammation but only 18/46 (39%) had IELs and TCRγδ + IELs above limits of normality. In 10 of 46 (22%) patients, a positive immunofluorescence indicated the presence of anti-TG2 mucosal antibodies. CONCLUSIONS SNVA is not rare representing up to 5% of the cases of VA. Most patients have a Marsh 3a lesion. Immunohistochemical analysis may be helpful in excluding CD, whereas the finding of mucosal anti-TG2, particularly with a weak staining, shows no absolute specificity for CD.
Collapse
|
34
|
Khuat LT, Dave M, Murphy WJ. The emerging roles of the gut microbiome in allogeneic hematopoietic stem cell transplantation. Gut Microbes 2021; 13:1966262. [PMID: 34455917 PMCID: PMC8436969 DOI: 10.1080/19490976.2021.1966262] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Accepted: 07/12/2021] [Indexed: 02/04/2023] Open
Abstract
Allogeneic hematopoietic stem cell transplantation (allo-HSCT) is used for the treatment of hematologic cancers and disorders. However, graft-versus-host disease (GVHD) in which the donor immune cells attack the genetically-disparate recipient is a significant cause of morbidity. Acute GVHD is an inflammatory condition and the gastrointestinal system is a major organ affected but is also tied to beneficial graft-versus-tumor (GVT) effects. There is increasing interest on the role of the microbiome on immune function as well as on cancer progression and immunotherapy outcomes. However, there are still significant unanswered questions on the role the microbiome plays in GVHD progression or how to exploit the microbiome in GVHD prevention or treatment. In this review, concepts of HSCT with the focus on GVHD pathogenesis as well as issues in preclinical models used to study GVHD will be discussed with an emphasis on the impact of the microbiome. Factors affecting the microbiome and GVHD outcome such as obesity are also examined. The bridging of preclinical models and clinical outcomes in relation to the role of the microbiome will also be discussed along with possibilities for therapeutic exploitation.
Collapse
Affiliation(s)
- Lam T. Khuat
- Department of Dermatology, School of Medicine, University of California, Davis, CA, USA
| | - Maneesh Dave
- Division of Gastroenterology, Department of Internal Medicine, School of Medicine, University of California, Davis, CA, USA
| | - William J. Murphy
- Department of Dermatology, School of Medicine, University of California, Davis, CA, USA
- Department of Internal Medicine, School of Medicine, University of California, Davis, CA, USAs
| |
Collapse
|
35
|
Hua J, Chen Y, Fu B, Chen X, Xu XJ, Yang SH, Chen C, Xu YJ. Downregulation of p53 by Insufficient CTCF in CD4 + T Cells Is an Important Factor Inducing Acute Graft-Versus-Host Disease. Front Immunol 2020; 11:568637. [PMID: 33133081 PMCID: PMC7550539 DOI: 10.3389/fimmu.2020.568637] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Accepted: 08/26/2020] [Indexed: 01/10/2023] Open
Abstract
Recent evidence indicates that p53 plays a protective role against various systemic autoimmune diseases by suppressing pro-inflammatory cytokine production and reducing the number of pathogenic T cells. However, whether abnormal p53 expression participates in the development of acute graft-versus-host disease (aGVHD) remains unclear. In this study, we demonstrated that p53 was downregulated in CD4+ T cells from patients with aGVHD compared with the non-aGVHD group. Furthermore, we confirmed that low expression of CCCTC-binding factor (CTCF) in CD4+ T cells from aGVHD cases is an important factor affecting histone H3K9/K14 hypoacetylation in the p53 promoter and p53 downregulation. Restoring CTCF expression in CD4+ T cells from aGVHD patients increased p53 amounts and corrected the imbalance of Th17 cells/Tregs. Taken together, these results provide novel insights into p53 downregulation in CD4+ T cells from aGVHD patients.
Collapse
Affiliation(s)
- Juan Hua
- Department of Hematology, Xiangya Hospital, Central South University, Changsha, China
| | - Yan Chen
- Department of Hematology, Xiangya Hospital, Central South University, Changsha, China
| | - Bin Fu
- Department of Hematology, Xiangya Hospital, Central South University, Changsha, China
| | - Xu Chen
- Department of Hematology, Xiangya Hospital, Central South University, Changsha, China
| | - Xue-Jun Xu
- Department of Hematology, Xiangya Hospital, Central South University, Changsha, China
| | - Shuang-Hui Yang
- Department of Hematology, Xiangya Hospital, Central South University, Changsha, China
| | - Cong Chen
- Department of Hematology, Xiangya Hospital, Central South University, Changsha, China
| | - Ya-Jing Xu
- Department of Hematology, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
36
|
Vandenhove B, Canti L, Schoemans H, Beguin Y, Baron F, Graux C, Kerre T, Servais S. How to Make an Immune System and a Foreign Host Quickly Cohabit in Peace? The Challenge of Acute Graft- Versus-Host Disease Prevention After Allogeneic Hematopoietic Cell Transplantation. Front Immunol 2020; 11:583564. [PMID: 33193397 PMCID: PMC7609863 DOI: 10.3389/fimmu.2020.583564] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 09/21/2020] [Indexed: 01/16/2023] Open
Abstract
Allogeneic hematopoietic cell transplantation (alloHCT) has been used as cellular immunotherapy against hematological cancers for more than six decades. Its therapeutic efficacy relies on the cytoreductive effects of the conditioning regimen but also on potent graft-versus-tumor (GVT) reactions mediated by donor-derived immune cells. However, beneficial GVT effects may be counterbalanced by acute GVHD (aGVHD), a systemic syndrome in which donor immune cells attack healthy tissues of the recipient, resulting in severe inflammatory lesions mainly of the skin, gut, and liver. Despite standard prophylaxis regimens, aGVHD still occurs in approximately 20–50% of alloHCT recipients and remains a leading cause of transplant-related mortality. Over the past two decades, advances in the understanding its pathophysiology have helped to redefine aGVHD reactions and clinical presentations as well as developing novel strategies to optimize its prevention. In this review, we provide a brief overview of current knowledge on aGVHD immunopathology and discuss current approaches and novel strategies being developed and evaluated in clinical trials for aGVHD prevention. Optimal prophylaxis of aGVHD would prevent the development of clinically significant aGVHD, while preserving sufficient immune responsiveness to maintain beneficial GVT effects and immune defenses against pathogens.
Collapse
Affiliation(s)
- Benoît Vandenhove
- Laboratory of Hematology, GIGA-I3, GIGA Institute, University of Liège, Liège, Belgium
| | - Lorenzo Canti
- Laboratory of Hematology, GIGA-I3, GIGA Institute, University of Liège, Liège, Belgium
| | - Hélène Schoemans
- Department of Clinical Hematology, University Hospitals Leuven, KU Leuven, Leuven, Belgium
| | - Yves Beguin
- Laboratory of Hematology, GIGA-I3, GIGA Institute, University of Liège, Liège, Belgium.,Department of Clinical Hematology, CHU of Liège, University of Liège, Liège, Belgium
| | - Frédéric Baron
- Laboratory of Hematology, GIGA-I3, GIGA Institute, University of Liège, Liège, Belgium.,Department of Clinical Hematology, CHU of Liège, University of Liège, Liège, Belgium
| | - Carlos Graux
- Department of Clinical Hematology, CHU UCL Namur (Godinne), Université Catholique de Louvain, Yvoir, Belgium
| | - Tessa Kerre
- Hematology Department, Ghent University Hospital, Ghent University, Ghent, Belgium
| | - Sophie Servais
- Laboratory of Hematology, GIGA-I3, GIGA Institute, University of Liège, Liège, Belgium.,Department of Clinical Hematology, CHU of Liège, University of Liège, Liège, Belgium
| |
Collapse
|
37
|
Yao WL, Wen Q, Zhao HY, Tang SQ, Zhang YY, Wang Y, Xu LP, Zhang XH, Huang XJ, Kong Y. Different subsets of haematopoietic cells and immune cells in bone marrow between young and older donors. Clin Exp Immunol 2020; 203:137-149. [PMID: 33020903 DOI: 10.1111/cei.13531] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2020] [Revised: 09/19/2020] [Accepted: 09/21/2020] [Indexed: 12/27/2022] Open
Abstract
Young donors are reported to be associated with better transplant outcomes than older donors in allogeneic hematopoietic stem cell transplantation (allo-HSCT), but the mechanism is still unclear. The current study compared the different subsets of haematopoietic stem cells (HSCs) and their progenitors as well as immune cells in bone marrow (BM) between young and older donors. The frequencies of HSCs, multipotent progenitors (MPPs) and myeloid progenitors, including common myeloid progenitors (CMPs) and megakaryocyte-erythroid progenitors (MEPs), were decreased, whereas those of lymphoid progenitors, including multi-potent lymphoid progenitors (MLPs) and common lymphoid progenitors (CLPs), were increased in the BM of young donors compared with in that of older donors. Lower reactive oxygen species (ROS) levels were observed in BM HSCs and six progenitor lines in young donors. Furthermore, young donors demonstrated higher frequencies of naive T cells and immune suppressor cells, such as alternative macrophages (M2) and lower frequencies of memory T cells and immune effectors, including T helper-1 and T cytotoxic-1 cells, in BM than older donors. Multivariate analysis demonstrated that donor age was independently correlated with BM HSC frequency. Although further validation is required, our results suggest that the differences in the frequency and immune differentiation potential of HSCs in BM between young donors and older donors may partly explain the different outcomes of allo-HSCT.
Collapse
Affiliation(s)
- W-L Yao
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Collaborative Innovation Center of Hematology, Peking University, Beijing, China
| | - Q Wen
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Collaborative Innovation Center of Hematology, Peking University, Beijing, China
| | - H-Y Zhao
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Collaborative Innovation Center of Hematology, Peking University, Beijing, China
| | - S-Q Tang
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Collaborative Innovation Center of Hematology, Peking University, Beijing, China
| | - Y-Y Zhang
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Collaborative Innovation Center of Hematology, Peking University, Beijing, China
| | - Y Wang
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Collaborative Innovation Center of Hematology, Peking University, Beijing, China
| | - L-P Xu
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Collaborative Innovation Center of Hematology, Peking University, Beijing, China
| | - X-H Zhang
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Collaborative Innovation Center of Hematology, Peking University, Beijing, China
| | - X-J Huang
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Collaborative Innovation Center of Hematology, Peking University, Beijing, China
| | - Y Kong
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Collaborative Innovation Center of Hematology, Peking University, Beijing, China
| |
Collapse
|
38
|
Pathogenic Bhlhe40+ GM-CSF+ CD4+ T cells promote indirect alloantigen presentation in the GI tract during GVHD. Blood 2020; 135:568-581. [PMID: 31880771 DOI: 10.1182/blood.2019001696] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Accepted: 12/10/2019] [Indexed: 12/23/2022] Open
Abstract
Gastrointestinal (GI) tract involvement is the major cause of morbidity and mortality in acute graft-versus-host disease (GVHD), and pathological damage is largely attributable to inflammatory cytokine production. Recently, granulocyte-macrophage colony stimulating factor (GM-CSF) has been identified as a cytokine that mediates inflammation in the GI tract, but the transcriptional program that governs GM-CSF production and the mechanism by which GM-CSF links adaptive to innate immunity within this tissue site have not been defined. In the current study, we identified Bhlhe40 as a key transcriptional regulator that governs GM-CSF production by CD4+ T cells and mediates pathological damage in the GI tract during GVHD. In addition, we observed that GM-CSF was not regulated by either interleukin 6 (IL-6) or IL-23, which are both potent inducers of GVHD-induced colonic pathology, indicating that GM-CSF constitutes a nonredundant inflammatory pathway in the GI tract. Mechanistically, GM-CSF had no adverse effect on regulatory T-cell reconstitution, but linked adaptive to innate immunity by enhancing the activation of donor-derived dendritic cells in the colon and subsequent accumulation of these cells in the mLNs. In addition, GM-CSF promoted indirect alloantigen presentation, resulting in the accumulation of donor-derived T cells with a proinflammatory cytokine phenotype in the colon. Thus, Bhlhe40+ GM-CSF+ CD4+ T cells constitute a colitogenic T-cell population that promotes indirect alloantigen presentation and pathological damage within the GI tract, positioning GM-CSF as a key regulator of GVHD in the colon and a potential therapeutic target for amelioration of this disease.
Collapse
|
39
|
Soto JR, Anthias C, Madrigal A, Snowden JA. Insights Into the Role of Vitamin D as a Biomarker in Stem Cell Transplantation. Front Immunol 2020; 11:966. [PMID: 32582151 PMCID: PMC7295104 DOI: 10.3389/fimmu.2020.00966] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Accepted: 04/24/2020] [Indexed: 12/19/2022] Open
Abstract
Vitamin D was discovered 100 years ago and since then multiple studies have consistently proved its effect on bone health and mineral metabolism. Further research has also explored its so-called "non-classical" biological effects, encompassing immune regulation and control of cell proliferation and differentiation. Vitamin D downregulates pro-inflammatory immune cells and subsequently their cytokine production, while enhancing the anti-inflammatory subsets, thus mediating inflammation and fostering a more tolerogenic environment. Its biological action is exerted through the vitamin D receptor, a nuclear receptor that mediates gene transcription and is expressed in most cells from the innate and adaptive immunity. Owing to its immune-modulatory properties, its role in cancer pathophysiology, hematology disorders and stem cell transplantation has also been investigated. Vitamin D deficiency causes immune imbalance and cytokine dysregulation, contributing to some autoimmune diseases. In the hematopoietic stem cell transplant setting this could lead to complications such as acute and chronic graft-versus-host disease, ultimately impacting transplant outcomes. Other factors have also been linked to this, including specific polymorphisms of the vitamin D receptor in both stem cell donors and recipients. Nevertheless, studies thus far have shown conflicting results and the use of vitamin D or its receptor as biomarkers has not been validated yet, therefore there are no evidence-based consensus guidelines to guide clinicians in their day-to-day practice. To gain more insight in this topic, we have reviewed the existent literature and gathered the current evidence. This is an overview of the role of serum vitamin D and its receptor as biomarkers for clinical outcomes in patients undergoing hematopoietic stem cell transplantation. Further prospective studies with larger cohorts are warranted to validate the viability of using serum vitamin D, and its receptor, as biomarkers in potential stem cell donors and patients, to identify those at risk of post-transplant complications and enable early therapeutic interventions.
Collapse
Affiliation(s)
- Jose Ros Soto
- Anthony Nolan Research Institute, Royal Free Hospital and University College London, London, United Kingdom
| | - Chloe Anthias
- Anthony Nolan Research Institute, Royal Free Hospital and University College London, London, United Kingdom
- Department of Haemato-Oncology, The Royal Marsden Hospital, Sutton, United Kingdom
| | - Alejandro Madrigal
- Anthony Nolan Research Institute, Royal Free Hospital and University College London, London, United Kingdom
| | - John A. Snowden
- Department of Haematology, Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, United Kingdom
| |
Collapse
|
40
|
Koldej RM, Perera T, Collins J, Ritchie DS. Association between P2X7 Polymorphisms and Post-Transplant Outcomes in Allogeneic Haematopoietic Stem Cell Transplantation. Int J Mol Sci 2020; 21:ijms21113772. [PMID: 32471261 PMCID: PMC7312926 DOI: 10.3390/ijms21113772] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 05/21/2020] [Accepted: 05/24/2020] [Indexed: 12/18/2022] Open
Abstract
Allogeneic stem cell transplantation (alloSCT) is a highly effective treatment method for haematologic malignancies. However, infection of acute organ dysfunction and graft versus host disease (GVHD) impact negatively on patient outcomes. Pre-transplant conditioning regimes are associated with high levels of immunogenic cell death and the release of extracellular ATP, which binds to the P2X7 receptor. It has been proposed that signaling through the P2X7 receptor may lead to activation of downstream effectors that influence alloSCT outcome. In this study, we examined the effect of gain-of-function (GOF) or loss-of-function (LOF) P2X7 Single Nucleotide Polymorphisms (SNP) in 453 paired alloSCT donors and recipients and correlated their presence or absence to the major post-transplant outcomes of acute GVHD, relapse free survival and overall survival. The allelic frequency of P2X7 SNP in recipients and donors was not different from those SNP for which there is published population data. The LOF SNP Glu496Ala was overrepresented in recipients who did not develop severe acute GVHD and was associated with improved overall survival in rare homozygous recipients, whereas the LOF SNP Ile568Asn was more common in patients with grade 1–4 GVHD but lost statistical association in patients with grade 2–4 aGVHD, and was associated with reduced overall survival in heterozygotes due to an excess of infection-related deaths. The GOF variant haplotype (homozygous Gln460Arg-Ala348Thr) had no impact on post-alloSCT outcomes. Overall, our data indicate that allelic variations in recipients or donors occurs at the same frequency as the general population and may have a minor, but clinically nominal, impact on post-alloSCT outcomes.
Collapse
Affiliation(s)
- Rachel M Koldej
- ACRF Translational Research Laboratory, Royal Melbourne Hospital, Melbourne, VIC 3050, Australia; (T.P.); (J.C.); (D.S.R.)
- Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Melbourne, VIC 3010, Australia
- Correspondence: ; Tel.: +61-3-8559-7264
| | - Travis Perera
- ACRF Translational Research Laboratory, Royal Melbourne Hospital, Melbourne, VIC 3050, Australia; (T.P.); (J.C.); (D.S.R.)
- Department of Clinical Haematology, Peter MacCallum Cancer Centre and Royal Melbourne Hospital, Melbourne, VIC 3000, Australia
| | - Jenny Collins
- ACRF Translational Research Laboratory, Royal Melbourne Hospital, Melbourne, VIC 3050, Australia; (T.P.); (J.C.); (D.S.R.)
- Department of Clinical Haematology, Peter MacCallum Cancer Centre and Royal Melbourne Hospital, Melbourne, VIC 3000, Australia
| | - David S Ritchie
- ACRF Translational Research Laboratory, Royal Melbourne Hospital, Melbourne, VIC 3050, Australia; (T.P.); (J.C.); (D.S.R.)
- Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Melbourne, VIC 3010, Australia
- Department of Clinical Haematology, Peter MacCallum Cancer Centre and Royal Melbourne Hospital, Melbourne, VIC 3000, Australia
| |
Collapse
|
41
|
Gavriilaki M, Sakellari I, Anagnostopoulos A, Gavriilaki E. The Impact of Antibiotic-Mediated Modification of the Intestinal Microbiome on Outcomes of Allogeneic Hematopoietic Cell Transplantation: Systematic Review and Meta-Analysis. Biol Blood Marrow Transplant 2020; 26:1738-1746. [PMID: 32447043 DOI: 10.1016/j.bbmt.2020.05.011] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 05/13/2020] [Indexed: 01/02/2023]
Abstract
Accumulating evidence points toward a protective role of intestinal microbiota diversity in allogeneic hematopoietic cell transplantation (allo-HCT). The purpose of this systematic review and meta-analysis is to determine the effect of antibiotic-mediated disruption of microbiota on main allo-HCT outcomes (graft-versus-host disease [GVHD], treatment-related mortality [TRM], overall survival [OS]). Following literature search, 2 reviewers screened eligible studies and assessed risk of bias (RoB). Meta-analysis was performed using Review Manager Software. Among 443 screened references, 18 were eligible for meta-analysis. In studies with genomic markers, grade II to IV acute GVHD was significantly reduced in patients not receiving gut decontamination (GD) (odds ratio [OR], 1.56; 95% confidence interval [CI], 1.20 to 2.04). In subgroup analysis, prophylaxis with systemic antibiotics conferred an increased risk of acute GVHD (OR, 1.65; 95% CI, 1.08 to 2.53). When we incorporated RoB, we found a positive correlation of intestinal GVHD with GD (OR, 1.77; 95% CI, 1.29 to 2.44). Patients with higher microbiota diversity presented increased OS (risk ratio [RR], 1.58; 95% CI, 1.19 to 2.09) and lower TRM (RR, 0.45; 95% CI, 0.26 to 0.76). Our findings confirm that GD and prophylaxis with systemic antibiotics increase acute and intestinal GVHD. Importantly, our meta-analysis was the first to show a significant effect of microbiota diversity on TRM and OS.
Collapse
Affiliation(s)
- Maria Gavriilaki
- Laboratory of Clinical Neurophysiology, AHEPA Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece.
| | - Ioanna Sakellari
- Hematology Department-BMT Unit, G. Papanicolaou Hospital, Thessaloniki, Greece
| | | | - Eleni Gavriilaki
- Hematology Department-BMT Unit, G. Papanicolaou Hospital, Thessaloniki, Greece
| |
Collapse
|
42
|
Woelfinger P, Epp K, Schaefer L, Kriege D, Theobald M, Bopp T, Wagner-Drouet EM. CD52-negative T cells predict acute graft-versus-host disease after an alemtuzumab-based conditioning regimen. Br J Haematol 2020; 191:253-262. [PMID: 32410220 DOI: 10.1111/bjh.16706] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Accepted: 04/09/2020] [Indexed: 11/28/2022]
Abstract
Allogeneic haematopoietic stem cell transplantation (HSCT) after a reduced-intensity conditioning (RIC) regimen with fludarabine, melphalan and alemtuzmab is an effective therapy for haematological malignancies. Alemtuzumab, a monoclonal antibody against CD52, a glycosylphosphatidylinositol-anchor-bound surface protein on lymphocytes, depletes T cells to prevent graft-versus-host disease (GVHD). Despite this, acute and chronic GVHD (a/cGVHD) remain life-threatening complications after HSCT. The aim of the present study was to identify parameters to predict GVHD. In 69 patients after HSCT, T-cell subsets were functionally analysed. Reconstitution of CD52neg T cells and CD52neg regulatory T cells (Tregs) correlated with onset, severity and clinical course of aGVHD. Patients with aGVHD showed significantly lower levels of CD52pos T cells compared to patients with cGVHD or without GVHD (P < 0·001). Analysis of T-cell reconstitution revealed a percentage of <40% of CD52pos CD4pos T cells or CD52pos Tregs at day +50 as a risk factor for the development of aGVHD. In contrast, CD52neg Tregs showed significant decreased levels of glycoprotein A repetitions predominant (GARP; P < 0·001), glucocorticoid-induced TNFR-related protein (GITR; P < 0·001), chemokine receptor (CXCR3; P = 0·023), C-C chemokine receptor type 5 (CCR5; P = 0·004), but increased levels of immunoglobulin-like transcript 3 (ILT3; P = 0·001), as well as a reduced suppressive capacity. We conclude that reconstitution of CD52neg T cells and CD52neg Tregs is a risk factor for development of aGVHD.
Collapse
Affiliation(s)
- Pascal Woelfinger
- Department of Hematology, Oncology and Pneumology, University Cancer Center Mainz (UCT), University Medical Center Mainz, Mainz, Germany
| | - Katharina Epp
- Department of Hematology, Oncology and Pneumology, University Cancer Center Mainz (UCT), University Medical Center Mainz, Mainz, Germany
| | - Lukas Schaefer
- Department of Hematology, Oncology and Pneumology, University Cancer Center Mainz (UCT), University Medical Center Mainz, Mainz, Germany
| | - Diana Kriege
- Department of Hematology, Oncology and Pneumology, University Cancer Center Mainz (UCT), University Medical Center Mainz, Mainz, Germany
| | - Matthias Theobald
- Department of Hematology, Oncology and Pneumology, University Cancer Center Mainz (UCT), University Medical Center Mainz, Mainz, Germany
| | - Tobias Bopp
- Institute for Immunology, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Eva-Maria Wagner-Drouet
- Department of Hematology, Oncology and Pneumology, University Cancer Center Mainz (UCT), University Medical Center Mainz, Mainz, Germany
| |
Collapse
|
43
|
Arsenic trioxide alleviates acute graft-versus-host disease by modulating macrophage polarization. SCIENCE CHINA-LIFE SCIENCES 2020; 63:1744-1754. [DOI: 10.1007/s11427-019-1691-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/26/2019] [Accepted: 03/27/2020] [Indexed: 12/24/2022]
|
44
|
Adom D, Rowan C, Adeniyan T, Yang J, Paczesny S. Biomarkers for Allogeneic HCT Outcomes. Front Immunol 2020; 11:673. [PMID: 32373125 PMCID: PMC7186420 DOI: 10.3389/fimmu.2020.00673] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2020] [Accepted: 03/25/2020] [Indexed: 12/23/2022] Open
Abstract
Allogeneic hematopoietic cell transplantation (HCT) remains the only curative therapy for many hematological malignant and non-malignant disorders. However, key obstacles to the success of HCT include graft-versus-host disease (GVHD) and disease relapse due to absence of graft-versus-tumor (GVT) effect. Over the last decade, advances in "omics" technologies and systems biology analysis, have allowed for the discovery and validation of blood biomarkers that can be used as diagnostic test and prognostic test (that risk-stratify patients before disease occurrence) for acute and chronic GVHD and recently GVT. There are also predictive biomarkers that categorize patients based on their likely to respond to therapy. Newer mathematical analysis such as machine learning is able to identify different predictors of GVHD using clinical characteristics pre-transplant and possibly in the future combined with other biomarkers. Biomarkers are not only useful to identify patients with higher risk of disease progression, but also help guide treatment decisions and/or provide a basis for specific therapeutic interventions. This review summarizes biomarkers definition, omics technologies, acute, chronic GVHD and GVT biomarkers currently used in clinic or with potential as targets for existing or new drugs focusing on novel published work.
Collapse
Affiliation(s)
- Djamilatou Adom
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, United States.,Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, United States.,Melvin and Bren Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Courtney Rowan
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, United States.,Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, United States.,Melvin and Bren Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Titilayo Adeniyan
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, United States.,Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, United States.,Melvin and Bren Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Jinfeng Yang
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, United States.,Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, United States.,Melvin and Bren Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Sophie Paczesny
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, United States.,Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, United States.,Melvin and Bren Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IN, United States
| |
Collapse
|
45
|
Emerging Role of Myeloid-derived Suppressor Cells in the Biology of Transplantation Tolerance. Transplantation 2020; 104:467-475. [DOI: 10.1097/tp.0000000000002996] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
46
|
Type I interferon signaling before hematopoietic stem cell transplantation lowers donor T cell activation via reduced allogenicity of recipient cells. Sci Rep 2019; 9:14955. [PMID: 31628411 PMCID: PMC6800427 DOI: 10.1038/s41598-019-51431-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Accepted: 09/30/2019] [Indexed: 12/21/2022] Open
Abstract
Recent studies highlight immunoregulatory functions of type I interferons (IFN-I) during the pathogenesis of graft-versus-host disease (GVHD) after allogeneic hematopoietic stem cell transplantation (allo-HSCT). We demonstrated that selective activation of IFN-I pathways including RIG-I/MAVS and cGAS/STING prior to allo-HSCT conditioning therapy can ameliorate the course of GVHD. However, direct effects of IFN-Is on immune cells remain ill characterized. We applied RIG-I agonists (3pRNA) to stimulate IFN-I production in murine models of conditioning therapy with total body irradiation (TBI) and GVHD. Using IFN-I receptor-deficient donor T cells and hematopoietic cells, we found that endogenous and RIG-I-induced IFN-Is do not reduce GVHD by acting on these cell types. However, 3pRNA applied before conditioning therapy reduced the ability of CD11c+ recipient cells to stimulate proliferation and interferon gamma expression of allogeneic T cells. Consistently, RIG-I activation before TBI reduced the proliferation of transplanted allogeneic T-cells. The reduced allogenicity of CD11c+ recipient cells was dependent on IFN-I signaling. Notably, this immunosuppressive function of DCs was restricted to a scenario where tissue damage occurs. Our findings uncover a context (damage by TBI) and IFN-I dependent modulation of T cells by DCs and extend the understanding about the cellular targets of IFN-I during allo-HSCT and GVHD.
Collapse
|
47
|
Gargiullo L, Del Chierico F, D’Argenio P, Putignani L. Gut Microbiota Modulation for Multidrug-Resistant Organism Decolonization: Present and Future Perspectives. Front Microbiol 2019; 10:1704. [PMID: 31402904 PMCID: PMC6671974 DOI: 10.3389/fmicb.2019.01704] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Accepted: 07/10/2019] [Indexed: 01/10/2023] Open
Abstract
The emergence of antimicrobial resistance (AMR) is of great concern to global public health. Treatment of multi-drug resistant (MDR) infections is a major clinical challenge: the increase in antibiotic resistance leads to a greater risk of therapeutic failure, relapses, longer hospitalizations, and worse clinical outcomes. Currently, there are no validated treatments for many MDR or pandrug-resistant (PDR) infections, and preventing the spread of these pathogens through hospital infection control procedures and antimicrobial stewardship programs is often the only tool available to healthcare providers. Therefore, new solutions to control the colonization of MDR pathogens are urgently needed. In this narrative review, we discuss current knowledge of microbiota-mediated mechanisms of AMR and strategies for MDR colonization control. We focus particularly on fecal microbiota transplantation for MDR intestinal decolonization and report updated literature on its current clinical use.
Collapse
Affiliation(s)
- Livia Gargiullo
- Division of Immunology and Infectious Diseases, University-Hospital Pediatric Department, Bambino Gesù Children’s Hospital, IRCSS, Rome, Italy
| | | | - Patrizia D’Argenio
- Division of Immunology and Infectious Diseases, University-Hospital Pediatric Department, Bambino Gesù Children’s Hospital, IRCSS, Rome, Italy
| | - Lorenza Putignani
- Human Microbiome Unit and Parasitology Unit, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| |
Collapse
|
48
|
Shao L, Pan S, Zhang QP, Jamal M, Chen LH, Yin Q, Wu YJ, Xiong J, Xiao RJ, Kwong YL, Zhou FL, Lie AKW. An Essential Role of Innate Lymphoid Cells in the Pathophysiology of Graft-vs.-Host Disease. Front Immunol 2019; 10:1233. [PMID: 31244831 PMCID: PMC6563595 DOI: 10.3389/fimmu.2019.01233] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2018] [Accepted: 05/15/2019] [Indexed: 12/14/2022] Open
Abstract
Allogeneic hematopoietic stem cell transplantation (Allo-HSCT) is the only curative treatment for multiple hematologic malignancies and non-malignant hematological diseases. However, graft-vs.-host disease (GVHD), one of the main complications after allo-HSCT, remains the major reason for morbidity and non-relapse mortality. Emerging evidence has demonstrated that innate lymphoid cells (ILCs) play a non-redundant role in the pathophysiology of GVHD. In this review, we will summarize previously published data regarding the role of ILCs in the pathogenesis of GVHD.
Collapse
Affiliation(s)
- Liang Shao
- Department of Hematology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Shan Pan
- Department of Immunology, School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Qiu-Ping Zhang
- Department of Immunology, School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Muhammad Jamal
- Department of Immunology, School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Lu-Hua Chen
- Department of Medicine, Li Ka Shing Faculty of Medicine, Faculty of Social Sciences, The University of Hong Kong, Hong Kong, China
| | - Qian Yin
- Department of Immunology, School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Ying-Jie Wu
- Department of Immunology, School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Jie Xiong
- Department of Immunology, School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Rui-Jing Xiao
- Department of Immunology, School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Yok-Lam Kwong
- Division of Hematology & BMT Center, Queen Mary Hospital, Hong Kong, China
| | - Fu-Ling Zhou
- Department of Hematology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Albert K W Lie
- Division of Hematology & BMT Center, Queen Mary Hospital, Hong Kong, China
| |
Collapse
|
49
|
Griesenauer B, Jiang H, Yang J, Zhang J, Ramadan AM, Egbosiuba J, Campa K, Paczesny S. ST2/MyD88 Deficiency Protects Mice against Acute Graft-versus-Host Disease and Spares Regulatory T Cells. THE JOURNAL OF IMMUNOLOGY 2019; 202:3053-3064. [PMID: 30979817 DOI: 10.4049/jimmunol.1800447] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Accepted: 03/18/2019] [Indexed: 12/11/2022]
Abstract
Acute graft-versus-host disease (aGVHD) hinders the efficacy of allogeneic hematopoietic cell transplantation (HCT). Plasma levels of soluble membrane-bound ST2 (ST2) are elevated in human and murine aGVHD and correlated to type 1 T cells response. ST2 signals through the adapter protein MyD88. The role of MyD88 in T cells during aGVHD has yet to be elucidated. We found that knocking out MyD88 in the donor T cells protected against aGVHD independent of IL-1R and TLR4 signaling in two murine HCT models. This protection was entirely driven by MyD88-/- CD4 T cells. Transplanting donor MyD88-/- conventional T cells (Tcons) with wild-type (WT) or MyD88-/- regulatory T cells (Tregs) lowered aGVHD severity and mortality. Transcriptome analysis of sorted MyD88-/- CD4 T cells from the intestine 10 d post-HCT showed lower levels of Il1rl1 (gene of ST2), Ifng, Csf2, Stat5, Batf, and Jak2 Transplanting donor ST2-/- Tcons with WT or ST2-/- Tregs showed a similar phenotype with what we observed when using donor MyD88-/- Tcons. Decreased ST2 was confirmed at the protein level with less secretion of soluble ST2 and more expression of ST2 compared with WT T cells. Our data suggest that Treg suppression from lack of MyD88 signaling in donor Tcons during alloreactivity uses the ST2 but not the IL-1R or TLR4 pathways, and ST2 represents a potential aGVHD therapeutic target sparing Tregs.
Collapse
Affiliation(s)
| | - Hua Jiang
- Indiana University School of Medicine, Indianapolis, IN 46202
| | - Jinfeng Yang
- Indiana University School of Medicine, Indianapolis, IN 46202
| | - Jilu Zhang
- Indiana University School of Medicine, Indianapolis, IN 46202
| | | | - Jane Egbosiuba
- Indiana University School of Medicine, Indianapolis, IN 46202
| | - Khaled Campa
- Indiana University School of Medicine, Indianapolis, IN 46202
| | - Sophie Paczesny
- Indiana University School of Medicine, Indianapolis, IN 46202
| |
Collapse
|
50
|
Grégoire C, Ritacco C, Hannon M, Seidel L, Delens L, Belle L, Dubois S, Vériter S, Lechanteur C, Briquet A, Servais S, Ehx G, Beguin Y, Baron F. Comparison of Mesenchymal Stromal Cells From Different Origins for the Treatment of Graft-vs.-Host-Disease in a Humanized Mouse Model. Front Immunol 2019; 10:619. [PMID: 31001253 PMCID: PMC6454068 DOI: 10.3389/fimmu.2019.00619] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Accepted: 03/08/2019] [Indexed: 12/23/2022] Open
Abstract
Mesenchymal stromal cells (MSCs) have potent immunomodulatory properties that make them an attractive tool against graft- vs.-host disease (GVHD). However, despite promising results in phase I/II studies, bone marrow (BM-) derived MSCs failed to demonstrate their superiority over placebo in the sole phase III trial reported thus far. MSCs from different tissue origins display different characteristics, but their therapeutic benefits have never been directly compared in GVHD. Here, we compared the impact of BM-, umbilical cord (UC-), and adipose-tissue (AT-) derived MSCs on T-cell function in vitro and assessed their efficacy for the treatment of GVHD induced by injection of human peripheral blood mononuclear cells in NOD-scid IL-2Rγnull HLA-A2/HHD mice. In vitro, resting BM- and AT-MSCs were more potent than UC-MSCs to inhibit lymphocyte proliferation, whereas UC- and AT-MSCs induced a higher regulatory T-cell (CD4+CD25+FoxP3+)/T helper 17 ratio. Interestingly, AT-MSCs and UC-MSCs activated the coagulation pathway at a higher level than BM-MSCs. In vivo, AT-MSC infusions were complicated by sudden death in 4 of 16 animals, precluding an analysis of their efficacy. Intravenous MSC infusions (UC- or BM- combined) failed to significantly increase overall survival (OS) in an analysis combining data from 80 mice (hazard ratio [HR] = 0.59, 95% confidence interval [CI] 0.32–1.08, P = 0.087). In a sensitivity analysis we also compared OS in control vs. each MSC group separately. The results for the BM-MSC vs. control comparison was HR = 0.63 (95% CI 0.30–1.34, P = 0.24) while the figures for the UC-MSC vs. control comparison was HR = 0.56 (95% CI 0.28–1.10, P = 0.09). Altogether, these results suggest that MSCs from various origins have different effects on immune cells in vitro and in vivo. However, none significantly prevented death from GVHD. Finally, our data suggest that the safety profile of AT-MSC and UC-MSC need to be closely monitored given their pro-coagulant activities in vitro.
Collapse
Affiliation(s)
- Céline Grégoire
- Hematology Research Unit, GIGA-I3, GIGA Institute, University of Liège, Liège, Belgium.,Department of Clinical Hematology, University Hospital Center of Liège, Liège, Belgium
| | - Caroline Ritacco
- Hematology Research Unit, GIGA-I3, GIGA Institute, University of Liège, Liège, Belgium
| | - Muriel Hannon
- Hematology Research Unit, GIGA-I3, GIGA Institute, University of Liège, Liège, Belgium
| | - Laurence Seidel
- Department of Biostatistics, SIMÉ, University Hospital Center of Liège, Liège, Belgium
| | - Loïc Delens
- Hematology Research Unit, GIGA-I3, GIGA Institute, University of Liège, Liège, Belgium
| | - Ludovic Belle
- Hematology Research Unit, GIGA-I3, GIGA Institute, University of Liège, Liège, Belgium
| | - Sophie Dubois
- Hematology Research Unit, GIGA-I3, GIGA Institute, University of Liège, Liège, Belgium
| | - Sophie Vériter
- Endocrine Cell Therapy, Centre of Tissue and Cellular Therapy, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Chantal Lechanteur
- Laboratory of Cell and Gene Therapy, University Hospital Center and University of Liège, Liège, Belgium
| | - Alexandra Briquet
- Laboratory of Cell and Gene Therapy, University Hospital Center and University of Liège, Liège, Belgium
| | - Sophie Servais
- Hematology Research Unit, GIGA-I3, GIGA Institute, University of Liège, Liège, Belgium.,Department of Clinical Hematology, University Hospital Center of Liège, Liège, Belgium
| | - Gregory Ehx
- Hematology Research Unit, GIGA-I3, GIGA Institute, University of Liège, Liège, Belgium
| | - Yves Beguin
- Hematology Research Unit, GIGA-I3, GIGA Institute, University of Liège, Liège, Belgium.,Department of Clinical Hematology, University Hospital Center of Liège, Liège, Belgium.,Laboratory of Cell and Gene Therapy, University Hospital Center and University of Liège, Liège, Belgium
| | - Frédéric Baron
- Hematology Research Unit, GIGA-I3, GIGA Institute, University of Liège, Liège, Belgium.,Department of Clinical Hematology, University Hospital Center of Liège, Liège, Belgium
| |
Collapse
|