201
|
Long J, Chang L, Shen Y, Gao WH, Wu YN, Dou HB, Huang MM, Wang Y, Fang WY, Shan JH, Wang YY, Zhu J, Chen Z, Hu J. Valproic Acid Ameliorates Graft-versus-Host Disease by Downregulating Th1 and Th17 Cells. THE JOURNAL OF IMMUNOLOGY 2015; 195:1849-57. [PMID: 26179902 DOI: 10.4049/jimmunol.1500578] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/10/2015] [Accepted: 06/02/2015] [Indexed: 01/27/2023]
Abstract
Graft-versus-host disease (GVHD) is the major complication after allogeneic bone marrow transplantation. Valproic acid (VPA) was described as a histone deacetylase inhibitor that had anti-inflammatory effects and reduced the production of proinflammatory cytokines in experimental autoimmune disease models. Using well-characterized mouse models of MHC-mismatched transplantation, we studied the effects of VPA on GVHD severity and graft-versus-leukemia (GVL) activity. Administration of VPA significantly attenuated the clinical severity of GVHD, the histopathology of GVHD-involved organs, and the overall mortality from GVHD. VPA downregulated Th1 and Th17 cell responses and cytokine production in vitro and in vivo, whereas its effect on GVHD was regulatory T cell independent. The effect of VPA was related to its ability to directly reduce the activity of Akt, an important regulator of T cell immune responses. Importantly, when mice received lethal doses of host-type acute leukemia cells, administration of VPA did not impair GVL activity and resulted in significantly improved leukemia-free survival. These findings reveal a unique role for VPA as a histone deacetylase inhibitor in reducing the donor CD4(+) T cells that contribute to GVHD, which may provide a strategy to reduce GVHD while preserving the GVL effect.
Collapse
Affiliation(s)
- Jun Long
- State Key Laboratory for Medical Genomics, Rui-Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Department of Hematology, Rui-Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Shanghai Institute of Hematology, Rui-Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Collaborative Innovation Center of Hematology, Rui-Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; and
| | - Li Chang
- State Key Laboratory for Medical Genomics, Rui-Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Department of Hematology, Rui-Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Shanghai Institute of Hematology, Rui-Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Collaborative Innovation Center of Hematology, Rui-Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; and
| | - Yan Shen
- Research Center for Experimental Medicine, Rui-Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Wen-Hui Gao
- State Key Laboratory for Medical Genomics, Rui-Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Department of Hematology, Rui-Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Shanghai Institute of Hematology, Rui-Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Collaborative Innovation Center of Hematology, Rui-Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; and
| | - Yue-Nv Wu
- State Key Laboratory for Medical Genomics, Rui-Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Department of Hematology, Rui-Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Shanghai Institute of Hematology, Rui-Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Collaborative Innovation Center of Hematology, Rui-Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; and
| | - Han-Bo Dou
- State Key Laboratory for Medical Genomics, Rui-Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Department of Hematology, Rui-Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Shanghai Institute of Hematology, Rui-Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Collaborative Innovation Center of Hematology, Rui-Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; and
| | - Meng-Meng Huang
- State Key Laboratory for Medical Genomics, Rui-Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Department of Hematology, Rui-Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Shanghai Institute of Hematology, Rui-Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Collaborative Innovation Center of Hematology, Rui-Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; and
| | - Ying Wang
- State Key Laboratory for Medical Genomics, Rui-Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Department of Hematology, Rui-Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Shanghai Institute of Hematology, Rui-Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Collaborative Innovation Center of Hematology, Rui-Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; and
| | - Wei-Yue Fang
- State Key Laboratory for Medical Genomics, Rui-Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Department of Hematology, Rui-Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Shanghai Institute of Hematology, Rui-Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Collaborative Innovation Center of Hematology, Rui-Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; and
| | - Jie-Hui Shan
- State Key Laboratory for Medical Genomics, Rui-Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Department of Hematology, Rui-Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Shanghai Institute of Hematology, Rui-Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Collaborative Innovation Center of Hematology, Rui-Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; and
| | - Yue-Ying Wang
- State Key Laboratory for Medical Genomics, Rui-Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Shanghai Institute of Hematology, Rui-Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Collaborative Innovation Center of Hematology, Rui-Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; and
| | - Jiang Zhu
- State Key Laboratory for Medical Genomics, Rui-Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Shanghai Institute of Hematology, Rui-Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Collaborative Innovation Center of Hematology, Rui-Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; and
| | - Zhu Chen
- State Key Laboratory for Medical Genomics, Rui-Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Shanghai Institute of Hematology, Rui-Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Collaborative Innovation Center of Hematology, Rui-Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; and
| | - Jiong Hu
- State Key Laboratory for Medical Genomics, Rui-Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Department of Hematology, Rui-Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Shanghai Institute of Hematology, Rui-Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Collaborative Innovation Center of Hematology, Rui-Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; and
| |
Collapse
|
202
|
Koyama M, Cheong M, Markey KA, Gartlan KH, Kuns RD, Locke KR, Lineburg KE, Teal BE, Leveque-El Mouttie L, Bunting MD, Vuckovic S, Zhang P, Teng MWL, Varelias A, Tey SK, Wockner LF, Engwerda CR, Smyth MJ, Belz GT, McColl SR, MacDonald KPA, Hill GR. Donor colonic CD103+ dendritic cells determine the severity of acute graft-versus-host disease. ACTA ACUST UNITED AC 2015; 212:1303-21. [PMID: 26169940 PMCID: PMC4516799 DOI: 10.1084/jem.20150329] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2015] [Accepted: 06/26/2015] [Indexed: 12/23/2022]
Abstract
Koyama et al. show that GVHD markedly enhances alloantigen presentation within the mesenteric lymph nodes, mediated by donor CD103+CD11b− DCs that migrate from the colon under the influence of CCR7. This antigen presentation imprints gut-homing integrin signatures on donor T cells, leading to their migration to the GI tract where they mediate fulminant disease. The primacy of the gastrointestinal (GI) tract in dictating the outcome of graft-versus-host disease (GVHD) is broadly accepted; however, the mechanisms controlling this effect are poorly understood. Here, we demonstrate that GVHD markedly enhances alloantigen presentation within the mesenteric lymph nodes (mLNs), mediated by donor CD103+CD11b− dendritic cells (DCs) that migrate from the colon under the influence of CCR7. Expansion and differentiation of donor T cells specifically within the mLNs is driven by profound levels of alloantigen, IL-12, and IL-6 promoted by Toll-like receptor (TLR) and receptor for advanced glycation end products (RAGE) signals. Critically, alloantigen presentation in the mLNs imprints gut-homing integrin signatures on donor T cells, leading to their emigration into the GI tract where they mediate fulminant disease. These data identify a critical, anatomically distinct, donor DC subset that amplifies GVHD. We thus highlight multiple therapeutic targets and the ability of GVHD, once initiated by recipient antigen-presenting cells, to generate a profound, localized, and lethal feed-forward cascade of donor DC–mediated indirect alloantigen presentation and cytokine secretion within the GI tract.
Collapse
Affiliation(s)
- Motoko Koyama
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland 4006, Australia
| | - Melody Cheong
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland 4006, Australia
| | - Kate A Markey
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland 4006, Australia
| | - Kate H Gartlan
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland 4006, Australia
| | - Rachel D Kuns
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland 4006, Australia
| | - Kelly R Locke
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland 4006, Australia
| | - Katie E Lineburg
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland 4006, Australia
| | - Bianca E Teal
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland 4006, Australia
| | | | - Mark D Bunting
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland 4006, Australia
| | - Slavica Vuckovic
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland 4006, Australia
| | - Ping Zhang
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland 4006, Australia
| | - Michele W L Teng
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland 4006, Australia
| | - Antiopi Varelias
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland 4006, Australia
| | - Siok-Keen Tey
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland 4006, Australia The Royal Brisbane and Women's Hospital, Brisbane, Queensland 4029, Australia
| | - Leesa F Wockner
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland 4006, Australia
| | | | - Mark J Smyth
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland 4006, Australia
| | - Gabrielle T Belz
- Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria 3052, Australia
| | - Shaun R McColl
- The University of Adelaide, Adelaide, South Australia 5005, Australia
| | - Kelli P A MacDonald
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland 4006, Australia
| | - Geoffrey R Hill
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland 4006, Australia The Royal Brisbane and Women's Hospital, Brisbane, Queensland 4029, Australia
| |
Collapse
|
203
|
Intestinal barrier loss as a critical pathogenic link between inflammatory bowel disease and graft-versus-host disease. Mucosal Immunol 2015; 8:720-30. [PMID: 25943273 DOI: 10.1038/mi.2015.40] [Citation(s) in RCA: 100] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2015] [Accepted: 04/08/2015] [Indexed: 02/04/2023]
Abstract
Compromised intestinal barrier function is a prominent feature of inflammatory bowel disease (IBD). However, links between intestinal barrier loss and disease extend much further, including documented associations with celiac disease, type I diabetes, rheumatoid arthritis, and multiple sclerosis. Intestinal barrier loss has also been proposed to have a critical role in the pathogenesis of graft-versus-host disease (GVHD), a serious, potentially fatal consequence of hematopoietic stem cell transplantation. Experimental evidence has begun to support this view, as barrier loss and its role in initiating and establishing a pathogenic inflammatory cycle in GVHD is emerging. Here we discuss similarities between IBD and GVHD, mechanisms of intestinal barrier loss in these diseases, and the crosstalk between barrier loss and the immune system, with a special focus on natural killer (NK) cells. Unanswered questions and future research directions on the topic are discussed along with implications for treatment.
Collapse
|
204
|
Lim JY, Lee YK, Lee SE, Ju JM, Park G, Choi EY, Min CK. Attenuation of Hepatic Graft-versus-host Disease in Allogeneic Recipients of MyD88-deficient Donor Bone Marrow. Immune Netw 2015; 15:125-34. [PMID: 26140044 PMCID: PMC4486775 DOI: 10.4110/in.2015.15.3.125] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2015] [Revised: 05/22/2015] [Accepted: 05/30/2015] [Indexed: 12/28/2022] Open
Abstract
Acute graft-versus-host-disease (GVHD) is characterized by selective damage to the liver, the skin, and the gastrointestinal tract. Following allogeneic hematopoietic stem cell transplantation, donor bone marrow (BM) cells repopulate the immune system of the recipient. We previously demonstrated that the acute intestinal GVHD (iGVHD) mortality rate was higher in MyD88-deficient BM recipients than that in the control BM recipients. In the present study, the role of MyD88 (expressed by donor BM) in the pathophysiology of hepatic GVHD (hGVHD) was examined. Unlike iGVHD, transplantation with MyD88-deficient T-cell depleted (TCD) BM attenuated hGVHD severity and was associated with low infiltration of T cells into the liver of the recipients. Moreover, GVHD hosts, transplanted with MyD88-deficient TCD BM, exhibited markedly reduced expansion of CD11b+Gr-1+ myeloid-derived suppressor cells (MDSC) in the liver. Adoptive injection of the MDSC from wild type mice, but not MyD88-deficient mice, enhanced hepatic T cell infiltration in the MyD88-deficient TCD BM recipients. Pre-treatment of BM donors with LPS increased MDSC levels in the liver of allogeneic wild type BM recipients. In conclusion, hGVHD and iGVHD may occur through various mechanisms based on the presence of MyD88 in the non-T cell compartment of the allograft.
Collapse
Affiliation(s)
- Ji-Young Lim
- Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul 137-701, Korea
| | - Young-Kwan Lee
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 110-799, Korea
| | - Sung-Eun Lee
- Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul 137-701, Korea
| | - Ji-Min Ju
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 110-799, Korea
| | - Gyeongsin Park
- Department of Pathology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul 137-701, Korea
| | - Eun Young Choi
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 110-799, Korea
| | - Chang-Ki Min
- Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul 137-701, Korea
| |
Collapse
|
205
|
Sun Y, Oravecz-Wilson K, Mathewson N, Wang Y, McEachin R, Liu C, Toubai T, Wu J, Rossi C, Braun T, Saunders T, Reddy P. Mature T cell responses are controlled by microRNA-142. J Clin Invest 2015; 125:2825-40. [PMID: 26098216 DOI: 10.1172/jci78753] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2015] [Accepted: 05/14/2015] [Indexed: 12/16/2022] Open
Abstract
T cell proliferation is critical for immune responses; however, the molecular mechanisms that mediate the proliferative response are poorly understood. MicroRNAs (miRs) regulate various molecular processes, including development and function of the immune system. Here, utilizing multiple complementary genetic and molecular approaches, we investigated the contribution of a hematopoietic-specific miR, miR-142, in regulating T cell responses. T cell development was not affected in animals with a targeted deletion of Mir142; however, T cell proliferation was markedly reduced following stimulation both in vitro and in multiple murine models of graft-versus-host disease (GVHD). miR-142-deficient T cells demonstrated substantial cell-cycling defects, and microarray and bioinformatics analyses revealed upregulation of genes involved in cell cycling. Moreover, 2 predicted miR-142 target genes, the atypical E2F transcription factors E2f7 and E2f8, were most highly upregulated in miR-142-deficient cells. Clustered regularly interspaced short palindromic repeat interference-mediated (CRISPRi-mediated) silencing of E2F7 and E2F8 in miR-142-deficient T cells ameliorated cell-cycling defects and reduced GVHD, and overexpression of these factors in WT T cells inhibited the proliferative response. Together, these results identify a link between hematopoietic-specific miR-142 and atypical E2F transcription factors in the regulation of mature T cell cycling and suggest that targeting this interaction may be relevant for mitigating GVHD.
Collapse
|
206
|
Li HW, Andreola G, Carlson AL, Shao S, Lin CP, Zhao G, Sykes M. Rapid Functional Decline of Activated and Memory Graft-versus-Host-Reactive T Cells Encountering Host Antigens in the Absence of Inflammation. THE JOURNAL OF IMMUNOLOGY 2015; 195:1282-92. [PMID: 26085679 DOI: 10.4049/jimmunol.1401511] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/12/2014] [Accepted: 05/22/2015] [Indexed: 01/04/2023]
Abstract
Inflammation in the priming host environment has critical effects on the graft-versus-host (GVH) responses mediated by naive donor T cells. However, it is unclear how a quiescent or inflammatory environment impacts the activity of GVH-reactive primed T and memory cells. We show in this article that GVH-reactive primed donor T cells generated in irradiated recipients had diminished ability compared with naive T cells to increase donor chimerism when transferred to quiescent mixed allogeneic chimeras. GVH-reactive primed T cells showed marked loss of cytotoxic function and activation, and delayed but not decreased proliferation or accumulation in lymphoid tissues when transferred to quiescent mixed chimeras compared with freshly irradiated secondary recipients. Primed CD4 and CD8 T cells provided mutual help to sustain these functions in both subsets. CD8 help for CD4 cells was largely IFN-γ dependent. TLR stimulation after transfer of GVH-reactive primed T cells to mixed chimeras restored their cytotoxic effector function and permitted the generation of more effective T cell memory in association with reduced PD-1 expression on CD4 memory cells. Our data indicate that an inflammatory host environment is required for the maintenance of GVH-reactive primed T cell functions and the generation of memory T cells that can rapidly acquire effector functions. These findings have important implications for graft-versus-host disease and T cell-mediated immunotherapies.
Collapse
Affiliation(s)
- Hao Wei Li
- Columbia Center for Translational Immunology, Columbia University Medical Center, New York, NY 10032; Transplantation Biology Research Center, Massachusetts General Hospital/Harvard Medical School, Boston, MA 02129
| | - Giovanna Andreola
- Transplantation Biology Research Center, Massachusetts General Hospital/Harvard Medical School, Boston, MA 02129
| | - Alicia L Carlson
- Advanced Microscopy Program, Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA 02114; and Center for Systems Biology, Massachusetts General Hospital, Boston, MA 02114
| | - Steven Shao
- Columbia Center for Translational Immunology, Columbia University Medical Center, New York, NY 10032
| | - Charles P Lin
- Advanced Microscopy Program, Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA 02114; and Center for Systems Biology, Massachusetts General Hospital, Boston, MA 02114
| | - Guiling Zhao
- Columbia Center for Translational Immunology, Columbia University Medical Center, New York, NY 10032; Transplantation Biology Research Center, Massachusetts General Hospital/Harvard Medical School, Boston, MA 02129
| | - Megan Sykes
- Columbia Center for Translational Immunology, Columbia University Medical Center, New York, NY 10032; Transplantation Biology Research Center, Massachusetts General Hospital/Harvard Medical School, Boston, MA 02129;
| |
Collapse
|
207
|
Espinosa-Carrasco G, Villard M, Le Saout C, Louis-Plence P, Vicente R, Hernandez J. Systemic LPS Translocation Activates Cross-Presenting Dendritic Cells but Is Dispensable for the Breakdown of CD8+ T Cell Peripheral Tolerance in Irradiated Mice. PLoS One 2015; 10:e0130041. [PMID: 26075613 PMCID: PMC4468093 DOI: 10.1371/journal.pone.0130041] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2015] [Accepted: 05/15/2015] [Indexed: 11/26/2022] Open
Abstract
Lymphodepletion is currently used to enhance the efficacy of cytotoxic T lymphocyte adoptive transfer immunotherapy against cancer. This beneficial effect of conditioning regimens is due, at least in part, to promoting the breakdown of peripheral CD8+ T cell tolerance. Lymphodepletion by total body irradiation induces systemic translocation of commensal bacteria LPS from the gastrointestinal tract. Since LPS is a potent activator of the innate immune system, including antigen presenting dendritic cells, we hypothesized that LPS translocation could be required for the breakdown of peripheral tolerance observed in irradiated mice. To address this issue, we have treated irradiated mice with antibiotics in order to prevent LPS translocation and utilized them in T cell adoptive transfer experiments. Surprisingly, we found that despite of completely blocking LPS translocation into the bloodstream, antibiotic treatment did not prevent the breakdown of peripheral tolerance. Although irradiation induced the activation of cross-presenting CD8+ dendritic cells in the lymphoid tissue, LPS could not solely account for this effect. Activation of dendritic cells by mechanisms other than LPS translocation is sufficient to promote the differentiation of potentially autoreactive CD8+ T cells into effectors in irradiated mice. Our data indicate that LPS translocation is dispensable for the breakdown of CD8+ T cell tolerance in irradiated mice.
Collapse
Affiliation(s)
- Gabriel Espinosa-Carrasco
- Inserm U1183, Institute for Regenerative Medicine and Biotherapy, Montpellier, F-34295, France
- Université Montpellier, UFR de Médecine, Montpellier, F-34000, France
| | - Marine Villard
- Inserm U1183, Institute for Regenerative Medicine and Biotherapy, Montpellier, F-34295, France
- Université Montpellier, UFR de Médecine, Montpellier, F-34000, France
| | - Cecile Le Saout
- CMRS/Laboratory of Immunoregulation, NIAID, NIH, Bethesda, Maryland, United States of America
| | - Pascale Louis-Plence
- Inserm U1183, Institute for Regenerative Medicine and Biotherapy, Montpellier, F-34295, France
- Université Montpellier, UFR de Médecine, Montpellier, F-34000, France
| | - Rita Vicente
- Inserm U1183, Institute for Regenerative Medicine and Biotherapy, Montpellier, F-34295, France
- Université Montpellier, UFR de Médecine, Montpellier, F-34000, France
| | - Javier Hernandez
- Inserm U1183, Institute for Regenerative Medicine and Biotherapy, Montpellier, F-34295, France
- Université Montpellier, UFR de Médecine, Montpellier, F-34000, France
- * E-mail:
| |
Collapse
|
208
|
Chen Y, Zhao Y, Cheng Q, Wu D, Liu H. The Role of Intestinal Microbiota in Acute Graft-versus-Host Disease. J Immunol Res 2015; 2015:145859. [PMID: 26090477 PMCID: PMC4452092 DOI: 10.1155/2015/145859] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2014] [Revised: 02/16/2015] [Accepted: 02/28/2015] [Indexed: 01/16/2023] Open
Abstract
The mammalian intestinal microbiota is a complex ecosystem that plays an important role in host immune responses. Recent studies have demonstrated that alterations in intestinal microbiota composition are linked to multiple inflammatory diseases in humans, including acute graft-versus-host disease (aGVHD). aGVHD is one of the major obstacles in allogeneic hematopoietic stem cell transplantation (allo-HSCT), characterized by tissue damage in the gastrointestinal (GI) tract, liver, lung, and skin. Here, we review the current understanding of the role of intestinal microbiota in the control of immune responses during aGVHD. Additionally, the possibility of using probiotic strains for potential treatment or prevention of aGVHD will be discussed.
Collapse
Affiliation(s)
- Yuanyuan Chen
- Laboratory of Cellular and Molecular Tumor Immunology, Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou 215123, China
| | - Ye Zhao
- Cyrus Tang Hematology Center, Department of Hematology, Jiangsu Institute of Hematology, Collaborative Innovation Center of Hematology, The First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Qiao Cheng
- Laboratory of Cellular and Molecular Tumor Immunology, Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou 215123, China
| | - Depei Wu
- Cyrus Tang Hematology Center, Department of Hematology, Jiangsu Institute of Hematology, Collaborative Innovation Center of Hematology, The First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Haiyan Liu
- Laboratory of Cellular and Molecular Tumor Immunology, Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou 215123, China
- Cyrus Tang Hematology Center, Department of Hematology, Jiangsu Institute of Hematology, Collaborative Innovation Center of Hematology, The First Affiliated Hospital of Soochow University, Suzhou 215006, China
| |
Collapse
|
209
|
Beyersdorf N, Werner S, Wolf N, Hünig T, Kerkau T. In vitro polyclonal activation of conventional T cells with a CD28 superagonist protects mice from acute graft versus host disease. Eur J Immunol 2015; 45:1997-2007. [DOI: 10.1002/eji.201445317] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2014] [Revised: 02/27/2015] [Accepted: 04/21/2015] [Indexed: 12/17/2022]
Affiliation(s)
- Niklas Beyersdorf
- Institute for Virology and Immunobiology; University of Würzburg; Würzburg Germany
| | - Sandra Werner
- Institute for Virology and Immunobiology; University of Würzburg; Würzburg Germany
| | - Nelli Wolf
- Institute for Virology and Immunobiology; University of Würzburg; Würzburg Germany
| | - Thomas Hünig
- Institute for Virology and Immunobiology; University of Würzburg; Würzburg Germany
| | - Thomas Kerkau
- Institute for Virology and Immunobiology; University of Würzburg; Würzburg Germany
| |
Collapse
|
210
|
State-of-the-art acute and chronic GVHD treatment. Int J Hematol 2015; 101:452-66. [DOI: 10.1007/s12185-015-1785-1] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2015] [Accepted: 03/23/2015] [Indexed: 01/09/2023]
|
211
|
Liu Y, Wu Y, Wang Y, Cai Y, Hu B, Bao G, Fang H, Zhao L, Ma S, Cheng Q, Song Y, Liu Y, Zhu Z, Chang H, Yu X, Sun A, Zhang Y, Vignali DAA, Wu D, Liu H. IL-35 mitigates murine acute graft-versus-host disease with retention of graft-versus-leukemia effects. Leukemia 2015; 29:939-46. [PMID: 25363669 PMCID: PMC4391991 DOI: 10.1038/leu.2014.310] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2014] [Revised: 10/13/2014] [Accepted: 10/16/2014] [Indexed: 01/09/2023]
Abstract
IL-35 is a newly discovered inhibitory cytokine secreted by regulatory T cells (Tregs) and may have therapeutic potential in several inflammatory disorders. Acute graft-versus-host disease (aGVHD) is a major complication of allogeneic hematopoietic stem cell transplantation and caused by donor T cells and inflammatory cytokines. The role of IL-35 in aGVHD is still unknown. Here we demonstrate that IL-35 overexpression suppresses CD4(+) effector T-cell activation, leading to a reduction in alloreactive T-cell responses and aGVHD severity. It also leads to the expansion of CD4(+)Foxp3(+) Tregs in the aGVHD target organs. Furthermore, IL-35 overexpression results in a selective decrease in the frequency of Th1 cells and an increase of IL-10-producing CD4(+) T cells in aGVHD target tissues. Serum levels of TNF-α, IFN-γ, IL-6, IL-22 and IL-23 decrease and IL-10 increases in response to IL-35. Most importantly, IL-35 preserves graft-versus-leukemia effect. Finally, aGVHD grade 2-4 patients have decreased serum IL-35 levels comparing with time-matched patients with aGVHD grade 0-1. Our findings indicate that IL-35 has an important role in reducing aGVHD through promoting the expansion of Tregs and repressing Th1 responses, and should be investigated as the therapeutic strategy for aGVHD.
Collapse
Affiliation(s)
- Yuejun Liu
- Laboratory of Cellular and Molecular Tumor Immunology, Cyrus Tang Hematology Center, Department of Hematology, Jiangsu Institute of Hematology, Collaborative Innovation Center of Hematology, the First Affiliated Hospital of Soochow University, Suzhou 215006, People's Republic of China
| | - Yan Wu
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou 215123, P. R. China
| | - Ying Wang
- Laboratory of Cellular and Molecular Tumor Immunology, Cyrus Tang Hematology Center, Department of Hematology, Jiangsu Institute of Hematology, Collaborative Innovation Center of Hematology, the First Affiliated Hospital of Soochow University, Suzhou 215006, People's Republic of China
| | - Yifeng Cai
- Laboratory of Cellular and Molecular Tumor Immunology, Cyrus Tang Hematology Center, Department of Hematology, Jiangsu Institute of Hematology, Collaborative Innovation Center of Hematology, the First Affiliated Hospital of Soochow University, Suzhou 215006, People's Republic of China
| | - Bo Hu
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou 215123, P. R. China
| | - Guangming Bao
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou 215123, P. R. China
| | - Hongying Fang
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou 215123, P. R. China
| | - Lixiang Zhao
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou 215123, P. R. China
| | - Shoubao Ma
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou 215123, P. R. China
| | - Qiao Cheng
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou 215123, P. R. China
| | - Yuan Song
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou 215123, P. R. China
| | - Yonghao Liu
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou 215123, P. R. China
| | - Ziling Zhu
- Laboratory of Cellular and Molecular Tumor Immunology, Cyrus Tang Hematology Center, Department of Hematology, Jiangsu Institute of Hematology, Collaborative Innovation Center of Hematology, the First Affiliated Hospital of Soochow University, Suzhou 215006, People's Republic of China
| | - Huirong Chang
- Laboratory of Cellular and Molecular Tumor Immunology, Cyrus Tang Hematology Center, Department of Hematology, Jiangsu Institute of Hematology, Collaborative Innovation Center of Hematology, the First Affiliated Hospital of Soochow University, Suzhou 215006, People's Republic of China
| | - Xiao Yu
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou 215123, P. R. China
| | - Aining Sun
- Laboratory of Cellular and Molecular Tumor Immunology, Cyrus Tang Hematology Center, Department of Hematology, Jiangsu Institute of Hematology, Collaborative Innovation Center of Hematology, the First Affiliated Hospital of Soochow University, Suzhou 215006, People's Republic of China
| | - Yi Zhang
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Dario A. A. Vignali
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Depei Wu
- Laboratory of Cellular and Molecular Tumor Immunology, Cyrus Tang Hematology Center, Department of Hematology, Jiangsu Institute of Hematology, Collaborative Innovation Center of Hematology, the First Affiliated Hospital of Soochow University, Suzhou 215006, People's Republic of China
| | - Haiyan Liu
- Laboratory of Cellular and Molecular Tumor Immunology, Cyrus Tang Hematology Center, Department of Hematology, Jiangsu Institute of Hematology, Collaborative Innovation Center of Hematology, the First Affiliated Hospital of Soochow University, Suzhou 215006, People's Republic of China
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou 215123, P. R. China
| |
Collapse
|
212
|
Nakasone H, Sahaf B, Miklos DB. Therapeutic benefits targeting B-cells in chronic graft-versus-host disease. Int J Hematol 2015; 101:438-51. [PMID: 25812839 DOI: 10.1007/s12185-015-1782-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Accepted: 03/16/2015] [Indexed: 12/21/2022]
Abstract
Allogeneic hematopoietic cell transplantation (allo-HCT) can be a curative strategy for hematological diseases, and the indications for allo-HCT have broadened widely due to recent progress in supportive strategies. However, patients must overcome various complications and chronic graft-versus-host disease (cGVHD) remains the most common allo-HCT cause of long-term morbidity and mortality. cGVHD is difficult to biologically assess due to the heterogeneity of cGVHD symptoms, and the pathogenesis of cGVHD has yet to be established. Recent experimental model progress has suggested that B-cells play a critical role in cGVHD development. Consistent with these experimental results, some clinical studies investigating B-cell depletion and modulation of B-cell signaling pathways have decreased cGVHD incidence and provided some therapeutic benefit. However, randomized control studies are necessary to confirm the efficacy of B-cell targeting drugs for cGVHD. Here, we review the pathophysiology of cGVHD, especially focusing on the role of B-cell immunity, and discuss the efficacy of both B-cell depletion and modulation of B-cell signaling pathways in human cGVHD prevention, initial treatment, and salvage treatment.
Collapse
Affiliation(s)
- Hideki Nakasone
- Division of Blood and Marrow Transplantation, Stanford University School of Medicine, 269 West Campus Dr., CCSR #2205, Stanford, CA, 94305, USA
| | | | | |
Collapse
|
213
|
Shono Y, Docampo MD, Peled JU, Perobelli SM, Jenq RR. Intestinal microbiota-related effects on graft-versus-host disease. Int J Hematol 2015; 101:428-37. [PMID: 25812838 DOI: 10.1007/s12185-015-1781-5] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2015] [Accepted: 03/16/2015] [Indexed: 12/18/2022]
Abstract
Allogeneic hematopoietic stem cell transplantation (allo-HSCT) is an increasingly important treatment for conditions including hematopoietic malignancies and inherited hematopoietic disorders, and is considered to be the most effective form of tumor immunotherapy available to date. However, graft-versus-host disease (GVHD) remains a major source of morbidity and mortality following allo-HSCT, and understanding the mechanisms of GVHD has been highlighted as a key research priority. During development of GVHD, activation of various immune cells, especially donor T cells, leads to damage of target organs including skin, liver, hematopoietic system, and of particular clinical importance, gut. In addition to histocompatibility complex differences between the donor and recipient, pretransplant conditioning with chemotherapy and irradiation also contributes to GVHD by damaging the gut, resulting in systemic exposure to microbial products normally confined to the intestinal lumen. The intestinal microbiota is a modulator of gastrointestinal immune homeostasis. It also promotes the maintenance of epithelial cells. Recent reports provide growing evidence of the impact of intestinal microbiota on GVHD pathophysiology. This review summarizes current knowledge of changes and effects of intestinal microbiota in the setting of allo-HSCT. We will also discuss potential future strategies of intestinal microbiota manipulation that might be advantageous in decreasing allo-HSCT-related morbidity and mortality.
Collapse
Affiliation(s)
- Yusuke Shono
- Department of Immunology, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA,
| | | | | | | | | |
Collapse
|
214
|
Yuan L, Chen X, Qian L, Shen J, Cai J. Administration of hydrogen-rich saline in mice with allogeneic hematopoietic stem-cell transplantation. Med Sci Monit 2015; 21:749-754. [PMID: 25763677 PMCID: PMC4367861 DOI: 10.12659/msm.891338] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2014] [Accepted: 10/21/2014] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Hydrogen, as a novel antioxidant, has been shown to selectively reduce the level of hydroxyl radicals and alleviate acute oxidative stress in many animal experiments. Hydrogen-rich saline provides a high concentration of hydrogen that can be easily and safely applied. Allogeneic hematopoietic stem-cell transplantation (HSCT) has been the most curative therapy for hematological malignancies. However, acute graft-versus-host disease (aGVHD) is the main cause of death in post-transplantation patients. In this study, we examined whether hydrogen-rich saline would show favorable effects on acute GVHD in mice. MATERIAL AND METHODS After lethal irradiation, BALB/c mice received bone marrow transplantation from C57BL/6 mice. Hydrogen-rich saline (5 ml/kg) was given to recipient mice in the hydrogen group once a day by intraperitoneal injection, and saline (5 ml/kg) was given to recipient mice in the saline group. Survival rates were monitored, clinical and pathological scores of aGVHD were determined after bone marrow transplantation (BMT), and the serum cytokine levels were examined on the 7th day after BMT. RESULTS This study proves that hydrogen-rich saline increased the survival rate, reduced clinical and histopathological scores of aGVHD, promoted the recovery of white blood cells, reduced the serum cytokine levels, and reversed tissue damage after transplantation in mice. CONCLUSIONS Hydrogen has potential as an effective and safe therapeutic agent in aGVHD.
Collapse
Affiliation(s)
- Lijuan Yuan
- Department of Hematology, Navy General Hospital, Beijing, P.R. China
- Postgraduate School, Anhui Medical University, Hefei, Anhui, P.R. China
| | - Xiaoping Chen
- Department of Hematology, Navy General Hospital, Beijing, P.R. China
| | - Liren Qian
- Department of Hematology, Navy General Hospital, Beijing, P.R. China
| | - Jianliang Shen
- Department of Hematology, Navy General Hospital, Beijing, P.R. China
| | - Jianming Cai
- Department of Radiation Medicine, Faculty of Naval Medicine, 2 Military Medical University, Shanghai, P.R. China
| |
Collapse
|
215
|
Kuba A, Raida L, Mrazek F, Schneiderova P, Kriegova E, Furst T, Furstova J, Faber E, Ambruzova Z, Papajik T. ATM gene single nucleotide polymorphisms predict regimen-related gastrointestinal toxicity in patients allografted after reduced conditioning. Biol Blood Marrow Transplant 2015; 21:1136-40. [PMID: 25759145 DOI: 10.1016/j.bbmt.2015.02.021] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Accepted: 02/23/2015] [Indexed: 10/23/2022]
Abstract
Polymorphisms of genes involved in innate and adaptive immunity have become an object of major interest in regard to hematopoietic stem cell transplantation (HSCT) complications. Regimen-related gastrointestinal toxicity (RR-GIT) is the dominant complication during the pre-engraftment period and has been linked to increased risk of graft-versus-host disease (GVHD) development. According to our hypothesis, functional variants of genes participating in DNA damage response (DDR) may have an impact on the extent of tissue damage caused by the conditioning regimen. In our single-center study, we analyzed 62 patients who underwent HSCT from HLA-identical donors after reduced conditioning. The patients were genotyped for 5 single nucleotide polymorphisms (SNPs, rs4585 T/G, rs189037 A/G, rs227092 T/G, rs228590 C/T, and rs664677 T/C) of the ATM gene-the essential member of the DDR pathways, using allele-specific matrix-assisted laser desorption/ionization, time-of-flight (MALDI-TOF) mass spectrometry assay. Because of almost absolute linkage disequilibrium observed among all 5 SNPs, association of 2 major ATM haplotypes (ATM1/ATM2) with RR-GIT and acute GVHD (aGVHD) was analyzed. Importantly, the univariate and multivariate analysis showed that patients homozygous for ATM2 haplotype (rs4585*T, rs189037*A, rs227092*T, rs228590*C, and rs664677*T) are more likely to suffer from high-grade RR-GIT than ATM1 homozygous patients. The association with aGVHD was not significant. To our knowledge, this is the first report showing the ATM gene variability in relation to RR-GIT in the allogeneic HSCT setting.
Collapse
Affiliation(s)
- Adam Kuba
- Department of Hemato-Oncology, University Hospital and Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czech Republic.
| | - Ludek Raida
- Department of Hemato-Oncology, University Hospital and Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czech Republic
| | - Frantisek Mrazek
- Department of Immunology, University Hospital and Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czech Republic
| | - Petra Schneiderova
- Department of Immunology, University Hospital and Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czech Republic
| | - Eva Kriegova
- Department of Immunology, University Hospital and Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czech Republic
| | - Tomas Furst
- Department of Mathematical Analysis and Applications of Mathematics, Faculty of Science, Palacky University, Olomouc, Czech Republic
| | - Jana Furstova
- Department of Mathematical Analysis and Applications of Mathematics, Faculty of Science, Palacky University, Olomouc, Czech Republic
| | - Edgar Faber
- Department of Hemato-Oncology, University Hospital and Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czech Republic
| | - Zuzana Ambruzova
- Department of Immunology, University Hospital and Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czech Republic
| | - Tomas Papajik
- Department of Hemato-Oncology, University Hospital and Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czech Republic
| |
Collapse
|
216
|
Jaime-Pérez JC, Villarreal-Villarreal CD, Salazar-Riojas R, Méndez-Ramírez N, Vázquez-Garza E, Gómez-Almaguer D. Increased Bacterial Infections after Transfusion of Leukoreduced Non-Irradiated Blood Products in Recipients of Allogeneic Stem Cell Transplants after Reduced-Intensity Conditioning. Biol Blood Marrow Transplant 2015; 21:526-30. [DOI: 10.1016/j.bbmt.2014.12.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2014] [Accepted: 12/01/2014] [Indexed: 10/24/2022]
|
217
|
Nalle SC, Kwak HA, Edelblum KL, Joseph NE, Singh G, Khramtsova GF, Mortenson ED, Savage PA, Turner JR. Recipient NK cell inactivation and intestinal barrier loss are required for MHC-matched graft-versus-host disease. Sci Transl Med 2015; 6:243ra87. [PMID: 24990882 DOI: 10.1126/scitranslmed.3008941] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Previous studies have shown a correlation between pretransplant conditioning intensity, intestinal barrier loss, and graft-versus-host disease (GVHD) severity. However, because irradiation and other forms of pretransplant conditioning have pleiotropic effects, the precise role of intestinal barrier loss in GVHD pathogenesis remains unclear. We developed GVHD models that allowed us to isolate the specific contributions of distinct pretransplant variables. Intestinal damage was required for the induction of minor mismatch [major histocompatibility complex (MHC)-matched] GVHD, but was not necessary for major mismatch GVHD, demonstrating fundamental pathogenic distinctions between these forms of disease. Moreover, recipient natural killer (NK) cells prevented minor mismatch GVHD by limiting expansion and target organ infiltration of alloreactive T cells via a perforin-dependent mechanism, revealing an immunoregulatory function of MHC-matched recipient NK cells in GVHD. Minor mismatch GVHD required MyD88-mediated Toll-like receptor 4 (TLR4) signaling on donor cells, and intestinal damage could be bypassed by parenteral lipopolysaccharide (LPS) administration, indicating a critical role for the influx of bacterial components triggered by intestinal barrier loss. In all, the data demonstrate that pretransplant conditioning plays a dual role in promoting minor mismatch GVHD by both depleting recipient NK cells and inducing intestinal barrier loss.
Collapse
Affiliation(s)
- Sam C Nalle
- Department of Pathology, The University of Chicago, Chicago, IL 60637, USA
| | - H Aimee Kwak
- Department of Pathology, The University of Chicago, Chicago, IL 60637, USA
| | - Karen L Edelblum
- Department of Pathology, The University of Chicago, Chicago, IL 60637, USA
| | - Nora E Joseph
- Department of Pathology, The University of Chicago, Chicago, IL 60637, USA
| | - Gurminder Singh
- Department of Pathology, The University of Chicago, Chicago, IL 60637, USA
| | | | - Eric D Mortenson
- Department of Pathology, The University of Chicago, Chicago, IL 60637, USA
| | - Peter A Savage
- Department of Pathology, The University of Chicago, Chicago, IL 60637, USA.
| | - Jerrold R Turner
- Department of Pathology, The University of Chicago, Chicago, IL 60637, USA.
| |
Collapse
|
218
|
Docampo MD, Auletta JJ, Jenq RR. Emerging Influence of the Intestinal Microbiota during Allogeneic Hematopoietic Cell Transplantation: Control the Gut and the Body Will Follow. Biol Blood Marrow Transplant 2015; 21:1360-6. [PMID: 25708215 DOI: 10.1016/j.bbmt.2015.02.016] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2014] [Accepted: 02/16/2015] [Indexed: 02/07/2023]
Abstract
The intestinal microbiota has many critical roles in maintaining gastrointestinal epithelial and gastrointestinal systemic immune homeostasis. This review provides insight into how allogeneic hematopoietic cell transplantation (HCT) and its associated complications and supportive care therapies affect the microbiota. Additionally, the review discusses how preservation and restoration of the microbiota might be advantageous in decreasing HCT-related morbidity and mortality.
Collapse
Affiliation(s)
- Melissa D Docampo
- Department of Immunology, Weill Cornell Graduate School of Medical Sciences, New York, New York.
| | - Jeffery J Auletta
- Host Defense Program, Hematology/Oncology/BMT and Infectious Diseases, Nationwide Children's Hospital and Department of Pediatrics, The Ohio State University College of Medicine, Columbus, Ohio
| | - Robert R Jenq
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan-Kettering Cancer Center and Weill Cornell Medical College, New York, New York
| |
Collapse
|
219
|
Lung parenchyma-derived IL-6 promotes IL-17A-dependent acute lung injury after allogeneic stem cell transplantation. Blood 2015; 125:2435-44. [PMID: 25673640 DOI: 10.1182/blood-2014-07-590232] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2014] [Accepted: 01/29/2015] [Indexed: 11/20/2022] Open
Abstract
Idiopathic pneumonia syndrome (IPS) is a relatively common, frequently fatal clinical entity, characterized by noninfectious acute lung inflammation following allogeneic stem cell transplantation (SCT), the mechanisms of which are unclear. In this study, we demonstrate that immune suppression with cyclosporin after SCT limits T-helper cell (Th) 1 differentiation and interferon-γ secretion by donor T cells, which is critical for inhibiting interleukin (IL)-6 generation from lung parenchyma during an alloimmune response. Thereafter, local IL-6 secretion induces donor alloantigen-specific Th17 cells to preferentially expand within the lung, and blockade of IL-17A or transplantation of grafts lacking the IL-17 receptor prevents disease. Studies using IL-6(-/-) recipients or IL-6 blockade demonstrate that IL-6 is the critical driver of donor Th17 differentiation within the lung. Importantly, IL-6 is also dysregulated in patients undergoing clinical SCT and is present at very high levels in the plasma of patients with IPS compared with SCT recipients without complications. Furthermore, at the time of diagnosis, plasma IL-6 levels were higher in a subset of IPS patients who were nonresponsive to steroids and anti-tumor necrosis factor therapy. In sum, pulmonary-derived IL-6 promotes IPS via the induction of Th17 differentiation, and strategies that target these cytokines represent logical therapeutic approaches for IPS.
Collapse
|
220
|
Kornblit B, Enevold C, Wang T, Spellman S, Haagenson M, Lee SJ, Müller K. Toll-like receptor polymorphisms in allogeneic hematopoietic cell transplantation. Biol Blood Marrow Transplant 2015; 21:259-65. [PMID: 25464115 PMCID: PMC4297590 DOI: 10.1016/j.bbmt.2014.09.016] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2014] [Accepted: 09/24/2014] [Indexed: 12/20/2022]
Abstract
To assess the impact of the genetic variation in toll-like receptors (TLRs) on outcome after allogeneic myeloablative conditioning hematopoietic cell transplantation (HCT), we investigated 29 single nucleotide polymorphisms across 10 TLRs in 816 patients and donors. Only donor genotype of TLR8 rs3764879, which is located on the X chromosome, was significantly associated with outcome at the Bonferroni-corrected level P ≤ .001. Male hemizygosity and female homozygosity for the minor allele were significantly associated with disease-free survival (hazard ratio [HR], 1.47 [95% confidence interval {CI}, 1.16 to 1.85]; P = .001). Further analysis stratified by donor sex due to confounding by sex was suggestive for associations with overall survival (male donor: HR, 1.41 [95% CI, 1.09 to 1.83], P = .010; female donor: HR, 2.78 [95% CI, 1.43 to 5.41], P = .003), disease-free survival (male donor: HR, 1.45 [95% CI, 1.12 to 1.87], P = .005; female donor: HR, 2.34 [95% CI, 1.18 to 4.65], P = .015), and treatment-related mortality (male donor: HR, 1.49 [95% CI, 1.09 to 2.04], P = .012; female donor: HR, 3.12 [95% CI, 1.44 to 6.74], P = .004). In conclusion, our findings suggest that the minor allele of TLR8 rs3764879 of the donor is associated with outcome after myeloablative conditioned allogeneic HCT.
Collapse
Affiliation(s)
- Brian Kornblit
- The Laboratory for Allogeneic Hematopoietic Cell Transplantation, Department of Hematology, Rigshospitalet, Copehnhagen, Denmark.
| | - Christian Enevold
- Department of Infectious Diseases and Rheumatology, Institute for Inflammation Research, Rigshospitalet, Copenhagen, Denmark
| | - Tao Wang
- Division of Biostatistics, Institute for Health and Society, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Stephen Spellman
- Center for International Blood and Marrow Transplant Research, Minneapolis, Minnesota
| | - Mike Haagenson
- Center for International Blood and Marrow Transplant Research, Minneapolis, Minnesota
| | - Stephanie J Lee
- Clinical Transplant Research, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Klaus Müller
- Department of Infectious Diseases and Rheumatology, Institute for Inflammation Research, Rigshospitalet, Copenhagen, Denmark; Department of Pediatrics and Adolescent Medicine, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| |
Collapse
|
221
|
Ramadan A, Paczesny S. Various forms of tissue damage and danger signals following hematopoietic stem-cell transplantation. Front Immunol 2015; 6:14. [PMID: 25674088 PMCID: PMC4309199 DOI: 10.3389/fimmu.2015.00014] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2014] [Accepted: 01/08/2015] [Indexed: 12/13/2022] Open
Abstract
Hematopoietic stem-cell transplantation (HSCT) is the most potent curative therapy for many malignant and non-malignant disorders. Unfortunately, a major complication of HSCT is graft-versus-host disease (GVHD), which is mediated by tissue damage resulting from the conditioning regimens before the transplantation and the alloreaction of dual immune components (activated donor T-cells and recipient’s antigen-presenting cells). This tissue damage leads to the release of alarmins and the triggering of pathogen-recognition receptors that activate the innate immune system and subsequently the adaptive immune system. Alarmins, which are of endogenous origin, together with the exogenous pathogen-associated molecular patterns (PAMPs) elicit similar responses of danger signals and represent the group of damage-associated molecular patterns (DAMPs). Effector cells of innate and adaptive immunity that are activated by PAMPs or alarmins can secrete other alarmins and amplify the immune responses. These complex interactions and loops between alarmins and PAMPs are particularly potent at inducing and then aggravating the GVHD reaction. In this review, we highlight the role of these tissue damaging molecules and their signaling pathways. Interestingly, some DAMPs and PAMPs are organ specific and GVHD-induced and have been shown to be interesting biomarkers. Some of these molecules may represent potential targets for novel therapeutic approaches.
Collapse
Affiliation(s)
- Abdulraouf Ramadan
- Department of Pediatrics, Melvin and Bren Simon Cancer Center, Indiana University , Indianapolis, IN , USA ; Department of Microbiology and Immunology, Indiana University , Indianapolis, IN , USA
| | - Sophie Paczesny
- Department of Pediatrics, Melvin and Bren Simon Cancer Center, Indiana University , Indianapolis, IN , USA ; Department of Microbiology and Immunology, Indiana University , Indianapolis, IN , USA
| |
Collapse
|
222
|
|
223
|
Theiss-Suennemann J, Jörß K, Messmann JJ, Reichardt SD, Montes-Cobos E, Lühder F, Tuckermann JP, AWolff H, Dressel R, Gröne HJ, Strauß G, Reichardt HM. Glucocorticoids attenuate acute graft-versus-host disease by suppressing the cytotoxic capacity of CD8(+) T cells. J Pathol 2015; 235:646-55. [PMID: 25358639 DOI: 10.1002/path.4475] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2014] [Revised: 10/09/2014] [Accepted: 10/29/2014] [Indexed: 01/15/2023]
Abstract
Glucocorticoids (GCs) are released from the adrenal gland during inflammation and help to keep immune responses at bay. Owing to their potent anti-inflammatory activity, GCs also play a key role in controlling acute graft-versus-host disease (aGvHD). Here we demonstrate that mice lacking the glucocorticoid receptor (GR) in T cells develop fulminant disease after allogeneic bone marrow transplantation. In a fully MHC-mismatched model, transfer of GR-deficient T cells resulted in severe aGvHD symptoms and strongly decreased survival times. Histopathological features were aggravated and infiltration of CD8(+) T cells into the jejunum was increased when the GR was not expressed. Furthermore, serum levels of IL-2, IFNγ, and IL-17 were elevated and the cytotoxicity of CD8(+) T cells was enhanced after transfer of GR-deficient T cells. Short-term treatment with dexamethasone reduced cytokine secretion but neither impacted disease severity nor the CTLs' cytolytic capacity. Importantly, in an aGvHD model in which disease development exclusively depends on the presence of CD8(+) T cells in the transplant, transfer of GR-deficient T cells aggravated clinical symptoms and reduced survival times as well. Taken together, our findings highlight that suppression of CD8(+) T-cell function is a crucial mechanism in the control of aGvHD by endogenous GCs.
Collapse
Affiliation(s)
- Jennifer Theiss-Suennemann
- Institute for Cellular and Molecular Immunology, University of Göttingen Medical School, Göttingen, Germany
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
224
|
Impact of conditioning intensity and TBI on acute GVHD after hematopoietic cell transplantation. Bone Marrow Transplant 2014; 50:559-65. [DOI: 10.1038/bmt.2014.293] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2014] [Revised: 10/28/2014] [Accepted: 11/14/2014] [Indexed: 01/24/2023]
|
225
|
Davis JE, Harvey M, Gherardin NA, Koldej R, Huntington N, Neeson P, Trapani JA, Ritchie DS. A radio-resistant perforin-expressing lymphoid population controls allogeneic T cell engraftment, activation, and onset of graft-versus-host disease in mice. Biol Blood Marrow Transplant 2014; 21:242-9. [PMID: 25459639 DOI: 10.1016/j.bbmt.2014.11.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2014] [Accepted: 11/03/2014] [Indexed: 10/24/2022]
Abstract
Immunosuppressive pretransplantation conditioning is essential for donor cell engraftment in allogeneic bone marrow transplantation (BMT). The role of residual postconditioning recipient immunity in determining engraftment is poorly understood. We examined the role of recipient perforin in the kinetics of donor cell engraftment. MHC-mismatched BMT mouse models demonstrated that both the rate and proportion of donor lymphoid cell engraftment and expansion of effector memory donor T cells in both spleen and BM were significantly increased within 5 to 7 days post-BMT in perforin-deficient (pfn(-/-)) recipients, compared with wild-type. In wild-type recipients, depletion of natural killer (NK) cells before BMT enhanced donor lymphoid cell engraftment to that seen in pfn(-/-) recipients. This demonstrated that a perforin-dependent, NK-mediated, host-versus-graft (HVG) effect limits the rate of donor engraftment and T cell activation. Radiation-resistant natural killer T (NKT) cells survived in the BM of lethally irradiated mice and may drive NK cell activation, resulting in the HVG effect. Furthermore, reduced pretransplant irradiation doses in pfn(-/-) recipients permitted long-term donor lymphoid cell engraftment. These findings suggest that suppression of perforin activity or selective depletion of recipient NK cells before BMT could be used to improve donor stem cell engraftment, in turn allowing for the reduction of pretransplant conditioning.
Collapse
Affiliation(s)
- Joanne E Davis
- ACRF Translational Research Laboratory, The Department of Research, The Royal Melbourne Hospital, Melbourne, Victoria, Australia; Haematology and Immunology Translational Research Laboratory, Cancer Immunology Research Department, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia; Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Victoria, Australia; Department of Medicine, University of Melbourne, Melbourne, Victoria, Australia.
| | - Michael Harvey
- Haematology and Immunology Translational Research Laboratory, Cancer Immunology Research Department, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Nicholas A Gherardin
- Haematology and Immunology Translational Research Laboratory, Cancer Immunology Research Department, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia; Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Parkville, Victoria, Australia
| | - Rachel Koldej
- ACRF Translational Research Laboratory, The Department of Research, The Royal Melbourne Hospital, Melbourne, Victoria, Australia; Haematology and Immunology Translational Research Laboratory, Cancer Immunology Research Department, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia; Department of Medicine, University of Melbourne, Melbourne, Victoria, Australia
| | - Nicholas Huntington
- Molecular Immunology Division, Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia; Department of Medical Biology, University of Melbourne, Melbourne, Victoria, Australia
| | - Paul Neeson
- ACRF Translational Research Laboratory, The Department of Research, The Royal Melbourne Hospital, Melbourne, Victoria, Australia; Haematology and Immunology Translational Research Laboratory, Cancer Immunology Research Department, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia; Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Victoria, Australia
| | - Joseph A Trapani
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Victoria, Australia
| | - David S Ritchie
- ACRF Translational Research Laboratory, The Department of Research, The Royal Melbourne Hospital, Melbourne, Victoria, Australia; Haematology and Immunology Translational Research Laboratory, Cancer Immunology Research Department, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia; Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Victoria, Australia; Department of Medicine, University of Melbourne, Melbourne, Victoria, Australia; Clinical Haematology and Bone Marrow Transplantation Service, Department of Clinical Oncology and Haematology, The Royal Melbourne Hospital, Melbourne, Victoria, Australia
| |
Collapse
|
226
|
Leveque L, Le Texier L, Lineburg KE, Hill GR, MacDonald KPA. Autophagy and haematopoietic stem cell transplantation. Immunol Cell Biol 2014; 93:43-50. [DOI: 10.1038/icb.2014.95] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2014] [Revised: 10/07/2014] [Accepted: 10/08/2014] [Indexed: 12/17/2022]
Affiliation(s)
- Lucie Leveque
- Department of Immunology, QIMR Berghofer Medical Research InstituteBrisbaneAustralia
| | - Laetitia Le Texier
- Department of Immunology, QIMR Berghofer Medical Research InstituteBrisbaneAustralia
| | - Katie E Lineburg
- Department of Immunology, QIMR Berghofer Medical Research InstituteBrisbaneAustralia
| | - Geoffrey R Hill
- Department of Immunology, QIMR Berghofer Medical Research InstituteBrisbaneAustralia
| | - Kelli PA MacDonald
- Department of Immunology, QIMR Berghofer Medical Research InstituteBrisbaneAustralia
| |
Collapse
|
227
|
Glick GD, Rossignol R, Lyssiotis CA, Wahl D, Lesch C, Sanchez B, Liu X, Hao LY, Taylor C, Hurd A, Ferrara JLM, Tkachev V, Byersdorfer CA, Boros L, Opipari AW. Anaplerotic metabolism of alloreactive T cells provides a metabolic approach to treat graft-versus-host disease. J Pharmacol Exp Ther 2014; 351:298-307. [PMID: 25125579 PMCID: PMC4201277 DOI: 10.1124/jpet.114.218099] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2014] [Accepted: 08/13/2014] [Indexed: 01/19/2023] Open
Abstract
T-cell activation requires increased ATP and biosynthesis to support proliferation and effector function. Most models of T-cell activation are based on in vitro culture systems and posit that aerobic glycolysis is employed to meet increased energetic and biosynthetic demands. By contrast, T cells activated in vivo by alloantigens in graft-versus-host disease (GVHD) increase mitochondrial oxygen consumption, fatty acid uptake, and oxidation, with small increases of glucose uptake and aerobic glycolysis. Here we show that these differences are not a consequence of alloactivation, because T cells activated in vitro either in a mixed lymphocyte reaction to the same alloantigens used in vivo or with agonistic anti-CD3/anti-CD28 antibodies increased aerobic glycolysis. Using targeted metabolic (13)C tracer fate associations, we elucidated the metabolic pathway(s) employed by alloreactive T cells in vivo that support this phenotype. We find that glutamine (Gln)-dependent tricarboxylic acid cycle anaplerosis is increased in alloreactive T cells and that Gln carbon contributes to ribose biosynthesis. Pharmacological modulation of oxidative phosphorylation rapidly reduces anaplerosis in alloreactive T cells and improves GVHD. On the basis of these data, we propose a model of T-cell metabolism that is relevant to activated lymphocytes in vivo, with implications for the discovery of new drugs for immune disorders.
Collapse
Affiliation(s)
- Gary D Glick
- Lycera Corporation, Ann Arbor, Michigan (G.D.G., C.L., B.S., X.L., L.-Y.H., C.T., A.H., A.W.O.); Departments of Chemical Biology (G.D.G., D.W.), Chemistry (G.D.G.), Pediatrics and Communicable Disease (J.L.M.F., V.T., C.A.B.), and Obstetrics and Gynecology (A.W.O.), University of Michigan, Ann Arbor, Michigan; Université Victor Segalen, Bordeaux, France (R.R.); Department of Medicine, Weill Cornell Medical College, New York, New York (C.A.L.); and SIDMAP, Los Angeles, California (L.B.)
| | - Rodrigue Rossignol
- Lycera Corporation, Ann Arbor, Michigan (G.D.G., C.L., B.S., X.L., L.-Y.H., C.T., A.H., A.W.O.); Departments of Chemical Biology (G.D.G., D.W.), Chemistry (G.D.G.), Pediatrics and Communicable Disease (J.L.M.F., V.T., C.A.B.), and Obstetrics and Gynecology (A.W.O.), University of Michigan, Ann Arbor, Michigan; Université Victor Segalen, Bordeaux, France (R.R.); Department of Medicine, Weill Cornell Medical College, New York, New York (C.A.L.); and SIDMAP, Los Angeles, California (L.B.)
| | - Costas A Lyssiotis
- Lycera Corporation, Ann Arbor, Michigan (G.D.G., C.L., B.S., X.L., L.-Y.H., C.T., A.H., A.W.O.); Departments of Chemical Biology (G.D.G., D.W.), Chemistry (G.D.G.), Pediatrics and Communicable Disease (J.L.M.F., V.T., C.A.B.), and Obstetrics and Gynecology (A.W.O.), University of Michigan, Ann Arbor, Michigan; Université Victor Segalen, Bordeaux, France (R.R.); Department of Medicine, Weill Cornell Medical College, New York, New York (C.A.L.); and SIDMAP, Los Angeles, California (L.B.)
| | - Daniel Wahl
- Lycera Corporation, Ann Arbor, Michigan (G.D.G., C.L., B.S., X.L., L.-Y.H., C.T., A.H., A.W.O.); Departments of Chemical Biology (G.D.G., D.W.), Chemistry (G.D.G.), Pediatrics and Communicable Disease (J.L.M.F., V.T., C.A.B.), and Obstetrics and Gynecology (A.W.O.), University of Michigan, Ann Arbor, Michigan; Université Victor Segalen, Bordeaux, France (R.R.); Department of Medicine, Weill Cornell Medical College, New York, New York (C.A.L.); and SIDMAP, Los Angeles, California (L.B.)
| | - Charles Lesch
- Lycera Corporation, Ann Arbor, Michigan (G.D.G., C.L., B.S., X.L., L.-Y.H., C.T., A.H., A.W.O.); Departments of Chemical Biology (G.D.G., D.W.), Chemistry (G.D.G.), Pediatrics and Communicable Disease (J.L.M.F., V.T., C.A.B.), and Obstetrics and Gynecology (A.W.O.), University of Michigan, Ann Arbor, Michigan; Université Victor Segalen, Bordeaux, France (R.R.); Department of Medicine, Weill Cornell Medical College, New York, New York (C.A.L.); and SIDMAP, Los Angeles, California (L.B.)
| | - Brian Sanchez
- Lycera Corporation, Ann Arbor, Michigan (G.D.G., C.L., B.S., X.L., L.-Y.H., C.T., A.H., A.W.O.); Departments of Chemical Biology (G.D.G., D.W.), Chemistry (G.D.G.), Pediatrics and Communicable Disease (J.L.M.F., V.T., C.A.B.), and Obstetrics and Gynecology (A.W.O.), University of Michigan, Ann Arbor, Michigan; Université Victor Segalen, Bordeaux, France (R.R.); Department of Medicine, Weill Cornell Medical College, New York, New York (C.A.L.); and SIDMAP, Los Angeles, California (L.B.)
| | - Xikui Liu
- Lycera Corporation, Ann Arbor, Michigan (G.D.G., C.L., B.S., X.L., L.-Y.H., C.T., A.H., A.W.O.); Departments of Chemical Biology (G.D.G., D.W.), Chemistry (G.D.G.), Pediatrics and Communicable Disease (J.L.M.F., V.T., C.A.B.), and Obstetrics and Gynecology (A.W.O.), University of Michigan, Ann Arbor, Michigan; Université Victor Segalen, Bordeaux, France (R.R.); Department of Medicine, Weill Cornell Medical College, New York, New York (C.A.L.); and SIDMAP, Los Angeles, California (L.B.)
| | - Ling-Yang Hao
- Lycera Corporation, Ann Arbor, Michigan (G.D.G., C.L., B.S., X.L., L.-Y.H., C.T., A.H., A.W.O.); Departments of Chemical Biology (G.D.G., D.W.), Chemistry (G.D.G.), Pediatrics and Communicable Disease (J.L.M.F., V.T., C.A.B.), and Obstetrics and Gynecology (A.W.O.), University of Michigan, Ann Arbor, Michigan; Université Victor Segalen, Bordeaux, France (R.R.); Department of Medicine, Weill Cornell Medical College, New York, New York (C.A.L.); and SIDMAP, Los Angeles, California (L.B.)
| | - Clarke Taylor
- Lycera Corporation, Ann Arbor, Michigan (G.D.G., C.L., B.S., X.L., L.-Y.H., C.T., A.H., A.W.O.); Departments of Chemical Biology (G.D.G., D.W.), Chemistry (G.D.G.), Pediatrics and Communicable Disease (J.L.M.F., V.T., C.A.B.), and Obstetrics and Gynecology (A.W.O.), University of Michigan, Ann Arbor, Michigan; Université Victor Segalen, Bordeaux, France (R.R.); Department of Medicine, Weill Cornell Medical College, New York, New York (C.A.L.); and SIDMAP, Los Angeles, California (L.B.)
| | - Alexander Hurd
- Lycera Corporation, Ann Arbor, Michigan (G.D.G., C.L., B.S., X.L., L.-Y.H., C.T., A.H., A.W.O.); Departments of Chemical Biology (G.D.G., D.W.), Chemistry (G.D.G.), Pediatrics and Communicable Disease (J.L.M.F., V.T., C.A.B.), and Obstetrics and Gynecology (A.W.O.), University of Michigan, Ann Arbor, Michigan; Université Victor Segalen, Bordeaux, France (R.R.); Department of Medicine, Weill Cornell Medical College, New York, New York (C.A.L.); and SIDMAP, Los Angeles, California (L.B.)
| | - James L M Ferrara
- Lycera Corporation, Ann Arbor, Michigan (G.D.G., C.L., B.S., X.L., L.-Y.H., C.T., A.H., A.W.O.); Departments of Chemical Biology (G.D.G., D.W.), Chemistry (G.D.G.), Pediatrics and Communicable Disease (J.L.M.F., V.T., C.A.B.), and Obstetrics and Gynecology (A.W.O.), University of Michigan, Ann Arbor, Michigan; Université Victor Segalen, Bordeaux, France (R.R.); Department of Medicine, Weill Cornell Medical College, New York, New York (C.A.L.); and SIDMAP, Los Angeles, California (L.B.)
| | - Victor Tkachev
- Lycera Corporation, Ann Arbor, Michigan (G.D.G., C.L., B.S., X.L., L.-Y.H., C.T., A.H., A.W.O.); Departments of Chemical Biology (G.D.G., D.W.), Chemistry (G.D.G.), Pediatrics and Communicable Disease (J.L.M.F., V.T., C.A.B.), and Obstetrics and Gynecology (A.W.O.), University of Michigan, Ann Arbor, Michigan; Université Victor Segalen, Bordeaux, France (R.R.); Department of Medicine, Weill Cornell Medical College, New York, New York (C.A.L.); and SIDMAP, Los Angeles, California (L.B.)
| | - Craig A Byersdorfer
- Lycera Corporation, Ann Arbor, Michigan (G.D.G., C.L., B.S., X.L., L.-Y.H., C.T., A.H., A.W.O.); Departments of Chemical Biology (G.D.G., D.W.), Chemistry (G.D.G.), Pediatrics and Communicable Disease (J.L.M.F., V.T., C.A.B.), and Obstetrics and Gynecology (A.W.O.), University of Michigan, Ann Arbor, Michigan; Université Victor Segalen, Bordeaux, France (R.R.); Department of Medicine, Weill Cornell Medical College, New York, New York (C.A.L.); and SIDMAP, Los Angeles, California (L.B.)
| | - Laszlo Boros
- Lycera Corporation, Ann Arbor, Michigan (G.D.G., C.L., B.S., X.L., L.-Y.H., C.T., A.H., A.W.O.); Departments of Chemical Biology (G.D.G., D.W.), Chemistry (G.D.G.), Pediatrics and Communicable Disease (J.L.M.F., V.T., C.A.B.), and Obstetrics and Gynecology (A.W.O.), University of Michigan, Ann Arbor, Michigan; Université Victor Segalen, Bordeaux, France (R.R.); Department of Medicine, Weill Cornell Medical College, New York, New York (C.A.L.); and SIDMAP, Los Angeles, California (L.B.)
| | - Anthony W Opipari
- Lycera Corporation, Ann Arbor, Michigan (G.D.G., C.L., B.S., X.L., L.-Y.H., C.T., A.H., A.W.O.); Departments of Chemical Biology (G.D.G., D.W.), Chemistry (G.D.G.), Pediatrics and Communicable Disease (J.L.M.F., V.T., C.A.B.), and Obstetrics and Gynecology (A.W.O.), University of Michigan, Ann Arbor, Michigan; Université Victor Segalen, Bordeaux, France (R.R.); Department of Medicine, Weill Cornell Medical College, New York, New York (C.A.L.); and SIDMAP, Los Angeles, California (L.B.)
| |
Collapse
|
228
|
Haploidentical stem cell transplantation for acute leukemia patients who experienced early relapse within one year after the first transplantation. Transplant Proc 2014; 46:3611-5. [PMID: 25240310 DOI: 10.1016/j.transproceed.2014.04.011] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2013] [Revised: 04/14/2014] [Accepted: 04/30/2014] [Indexed: 11/24/2022]
Abstract
To assess the safety and efficacy of allogeneic stem cell transplantation from haploidentical related donors (haplo-SCT) as 2nd transplantation for patients with early relapsed disease, we retrospectively evaluated 7 consecutive patients (median age, 42 years; range, 29-63 years) who experienced relapse within 1 year of the 1st transplantation and received haplo-SCT as a 2nd transplantation. Among the 7 patients who received haplo-SCT, 2 who were in morphologically complete remission (CR) at transplantation were conditioned with a reduced-intensity regimen, and the 5 non-CR patients were conditioned with a myeloablative regimen. Both conditioning regimens included antithymocyte globulin. Graft-versus-host disease (GVHD) prophylaxis consisted of tacrolimus and methylprednisolone. Sustained neutrophil engraftment was achieved in all 7 patients. One patient developed severe acute GVHD. Notably, only 1 patient experienced relapse, and each patient achieved longer CR duration than after the 1st transplantation. Three of the 7 patients died from treatment-related causes: acute GVHD, post-transplantation lymphoproliferative disorder, and bacterial pneumonia. At the time of analysis, the 2-year overall survival rate of these 7 patients was 42.9%. This suggests that use of haploidentical related donors is a viable alternative for 2nd transplantation and should be confirmed in larger cohorts.
Collapse
|
229
|
van der Velden WJFM, Herbers AHE, Netea MG, Blijlevens NMA. Mucosal barrier injury, fever and infection in neutropenic patients with cancer: introducing the paradigm febrile mucositis. Br J Haematol 2014; 167:441-52. [PMID: 25196917 DOI: 10.1111/bjh.13113] [Citation(s) in RCA: 95] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Infection remains one of the most prominent complications after cytotoxic treatment for cancer. The connection between neutropenia and both infections and fever has long been designated as 'febrile neutropenia', but treatment with antimicrobial agents and haematopoietic growth factors has failed to significantly reduce its incidence. Moreover, emerging antimicrobial resistance is becoming a concern that necessitates the judicious use of available antimicrobial agents. In addition to neutropenia, patients who receive cytotoxic therapy experience mucosal barrier injury (MBI) or 'mucositis'. MBI creates a port-de-entrée for resident micro-organisms to cause blood stream infections and contributes directly to the occurrence of fever by disrupting the highly regulated host-microbe interactions, which, even in the absence of an infection, can result in strong inflammatory reactions. Indeed, MBI has been shown to be a pivotal factor in the occurrence of inflammatory complications after cytotoxic therapy. Hence, the concept 'febrile neutropenia' alone may no longer suffice and a new concept 'febrile mucositis' should be recognized as the two are at least complementary. This review we summarizes the existing evidence for both paradigms and proposes new therapeutic approaches to tackle the perturbed host-microbe interactions arising from cytotoxic therapy-induced tissue damage in order to reduce fever in neutropenic patients with cancer.
Collapse
|
230
|
Cruz A, Mendes ÉA, de Andrade MVM, do Nascimento VC, Cartelle CT, Arantes RME, Melo JRDC, Gazzinelli RT, Ropert C. Mast cells are crucial in the resistance against Toxoplasma gondii oral infection. Eur J Immunol 2014; 44:2949-54. [PMID: 25091816 DOI: 10.1002/eji.201344185] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2013] [Revised: 06/19/2014] [Accepted: 07/30/2014] [Indexed: 11/06/2022]
Abstract
During oral infection, mucosal immunity assumes a predominant role. Here, we addressed the role of mast cells (MCs), which are mainly located in mucosa during oral infection with Toxoplasma gondii, using MC-deficient (W/W(v) ) mice. We show that in the absence of MCs the resistance of W/W(v) mice to oral infection was considerably reduced. W/W(v) mice uniformly succumbed within 15 days of infection after administration of cysts of the ME49 strain of T. gondii. The rapid lethality of T. gondii in W/W(v) mice correlated with a delayed Th1-cell response, since IFN-γ and IL-12 levels peaked in the later phase of the infection. In vitro, BM-derived MCs were able to recognize parasite lysate in a MyD88-dependent way, reaffirming the role of this TLR adapter in immune responses to T. gondii. The importance of MCs in vivo was confirmed when W/W(v) mice reconstituted with BM-derived MCs from control mice retrieved an early strong Th1-cell response and specially a significant IL-12 production. In conclusion, MCs play an important role for the development of a protective immune response during oral infection with T. gondii.
Collapse
Affiliation(s)
- Aline Cruz
- School of Medicine, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil; Department of Biochemistry and Immunology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | | | | | | | | | | | | | | | | |
Collapse
|
231
|
Alexander KA, Flynn R, Lineburg KE, Kuns RD, Teal BE, Olver SD, Lor M, Raffelt NC, Koyama M, Leveque L, Le Texier L, Melino M, Markey KA, Varelias A, Engwerda C, Serody JS, Janela B, Ginhoux F, Clouston AD, Blazar BR, Hill GR, MacDonald KPA. CSF-1-dependant donor-derived macrophages mediate chronic graft-versus-host disease. J Clin Invest 2014; 124:4266-80. [PMID: 25157821 DOI: 10.1172/jci75935] [Citation(s) in RCA: 176] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2014] [Accepted: 07/10/2014] [Indexed: 12/23/2022] Open
Abstract
Chronic GVHD (cGVHD) is the major cause of late, nonrelapse death following stem cell transplantation and characteristically develops in organs such as skin and lung. Here, we used multiple murine models of cGVHD to investigate the contribution of macrophage populations in the development of cGVHD. Using an established IL-17-dependent sclerodermatous cGVHD model, we confirmed that macrophages infiltrating the skin are derived from donor bone marrow (F4/80+CSF-1R+CD206+iNOS-). Cutaneous cGVHD developed in a CSF-1/CSF-1R-dependent manner, as treatment of recipients after transplantation with CSF-1 exacerbated macrophage infiltration and cutaneous pathology. Additionally, recipients of grafts from Csf1r-/- mice had substantially less macrophage infiltration and cutaneous pathology as compared with those receiving wild-type grafts. Neither CCL2/CCR2 nor GM-CSF/GM-CSFR signaling pathways were required for macrophage infiltration or development of cGVHD. In a different cGVHD model, in which bronchiolitis obliterans is a prominent manifestation, F4/80+ macrophage infiltration was similarly noted in the lungs of recipients after transplantation, and lung cGVHD was also IL-17 and CSF-1/CSF-1R dependent. Importantly, depletion of macrophages using an anti-CSF-1R mAb markedly reduced cutaneous and pulmonary cGVHD. Taken together, these data indicate that donor macrophages mediate the development of cGVHD and suggest that targeting CSF-1 signaling after transplantation may prevent and treat cGVHD.
Collapse
|
232
|
Wu T, Zhao Y, Zhao Y. The roles of myeloid-derived suppressor cells in transplantation. Expert Rev Clin Immunol 2014; 10:1385-94. [DOI: 10.1586/1744666x.2014.948424] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
|
233
|
Trzonkowski P, Dukat-Mazurek A, Bieniaszewska M, Marek-Trzonkowska N, Dobyszuk A, Juścińska J, Dutka M, Myśliwska J, Hellmann A. Treatment of graft-versus-host disease with naturally occurring T regulatory cells. BioDrugs 2014; 27:605-14. [PMID: 23813436 PMCID: PMC3832760 DOI: 10.1007/s40259-013-0050-5] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
A significant body of evidence suggests that treatment with naturally occurring CD4+CD25+ T regulatory cells (Tregs) is an appropriate therapy for graft-versus-host disease (GvHD). GvHD is a major complication of bone marrow transplantation in which the transplanted immune system recognizes recipient tissues as a non-self and destroys them. In many cases, this condition significantly deteriorates the quality of life of the affected patients. It is also one of the most important causes of death after bone marrow transplantation. Tregs constitute a population responsible for dominant tolerance to self-tissues in the immune system. These cells prevent autoimmune and allergic reactions and decrease the risk of rejection of allotransplants. For these reasons, Tregs are considered as a cellular drug in GvHD. The results of the first clinical trials with these cells are already available. In this review we present important experimental facts which led to the clinical use of Tregs. We then critically evaluate specific requirements for Treg therapy in GvHD and therapies with Tregs currently under clinical investigation, including our experience and future perspectives on this kind of cellular treatment.
Collapse
Affiliation(s)
- Piotr Trzonkowski
- Department of Clinical Immunology and Transplantology, Medical University of Gdańsk, Ul. Dębinki 1, 80-211, Gdańsk, Poland,
| | | | | | | | | | | | | | | | | |
Collapse
|
234
|
Allogeneic hematopoietic cell transplantation for myelofibrosis using fludarabine-, intravenous busulfan- and low-dose TBI-based conditioning. Bone Marrow Transplant 2014; 49:1162-9. [PMID: 24978138 DOI: 10.1038/bmt.2014.131] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2014] [Revised: 05/08/2014] [Accepted: 05/12/2014] [Indexed: 12/31/2022]
Abstract
Graft failure is one of the major barriers to the success of allogeneic hematopoietic cell transplantation (HCT) in myelofibrosis (MF). We report our institutional experience with 27 MF patients who underwent HCT using fludarabine-, intravenous BU- and low-dose total body irradiation (FBT)-based reduced-intensity (n=20) or full-intensity (n=7) conditioning regimens. Eight patients had prior exposure to JAK1/2 inhibitor therapy; six patients received JAK1/2 inhibitors leading on to HCT and two patients received transplant at the failure of JAK1/2 inhibitor therapy. No adverse impact of JAK1/2 inhibitor therapy was observed on early post-transplant outcomes. All evaluable patients had neutrophil recovery, and no primary graft failure was observed. Cumulative incidence of grades II-IV acute GVHD at day 100 was 48% (95% confidence interval (CI), 29-67%) and chronic GVHD at 2 years was 66% (95% CI, 49-84%). Cumulative incidences of nonrelapse mortality (NRM), relapse and probability of OS at 2 years were: 43% (95% CI, 12-74%), 10% (95% CI, 0-39%) and 56% (95% CI, 28-77%), respectively. FBT-based conditioning regimen has a favorable impact on engraftment; however, further efforts are required to reduce NRM.
Collapse
|
235
|
Progressive proximal-to-distal reduction in expression of the tight junction complex in colonic epithelium of virally-suppressed HIV+ individuals. PLoS Pathog 2014; 10:e1004198. [PMID: 24968145 PMCID: PMC4072797 DOI: 10.1371/journal.ppat.1004198] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2013] [Accepted: 05/06/2014] [Indexed: 01/11/2023] Open
Abstract
Effective antiretroviral therapy (ART) dramatically reduces AIDS-related complications, yet the life expectancy of long-term ART-treated HIV-infected patients remains shortened compared to that of uninfected controls, due to increased risk of non-AIDS related morbidities. Many propose that these complications result from translocated microbial products from the gut that stimulate systemic inflammation--a consequence of increased intestinal paracellular permeability that persists in this population. Concurrent intestinal immunodeficiency and structural barrier deterioration are postulated to drive microbial translocation, and direct evidence of intestinal epithelial breakdown has been reported in untreated pathogenic SIV infection of rhesus macaques. To assess and characterize the extent of epithelial cell damage in virally-suppressed HIV-infected patients, we analyzed intestinal biopsy tissues for changes in the epithelium at the cellular and molecular level. The intestinal epithelium in the HIV gut is grossly intact, exhibiting no decreases in the relative abundance and packing of intestinal epithelial cells. We found no evidence for structural and subcellular localization changes in intestinal epithelial tight junctions (TJ), but observed significant decreases in the colonic, but not terminal ileal, transcript levels of TJ components in the HIV+ cohort. This result is confirmed by a reduction in TJ proteins in the descending colon of HIV+ patients. In the HIV+ cohort, colonic TJ transcript levels progressively decreased along the proximal-to-distal axis. In contrast, expression levels of the same TJ transcripts stayed unchanged, or progressively increased, from the proximal-to-distal gut in the healthy controls. Non-TJ intestinal epithelial cell-specific mRNAs reveal differing patterns of HIV-associated transcriptional alteration, arguing for an overall change in intestinal epithelial transcriptional regulation in the HIV colon. These findings suggest that persistent intestinal epithelial dysregulation involving a reduction in TJ expression is a mechanism driving increases in colonic permeability and microbial translocation in the ART-treated HIV-infected patient, and a possible immunopathogenic factor for non-AIDS related complications.
Collapse
|
236
|
Abstract
The last 6 decades have seen major advances in the understanding of immunologic diseases, driven by preclinical animal models. Indeed, bone marrow transplantation (BMT) has its genesis in rodent models dating back to the 1950s. Allogeneic BMT and its major complication, graft-versus-host disease (GVHD), represent a paradigm for the translation of preclinical concepts into clinical practice. The appreciation that GVHD can be thought of as a stepwise escalation in immune activation characterized by eventual massive target tissue apoptosis has allowed the design of rational approaches to better manage patients. Here, we describe the pathophysiology of GVHD as defined in preclinical models, focusing on the successes and failures of this research to instruct and translate clinical practice. We also provide a commentary on the limitations of these models so that they may be better appreciated and addressed in future studies. Notable preclinical successes include the definition of modern immune suppression, reductions in conditioning intensity, posttransplant cyclophosphamide, and the promotion of regulatory T-cell reconstitution. New strategies including naïve T-cell depletion, focused cytokine and chemokine inhibition, and the blockade of costimulation now also appear highly promising and very likely to translate into patients in the near future.
Collapse
|
237
|
Bone marrow T-cell infiltration during acute GVHD is associated with delayed B-cell recovery and function after HSCT. Blood 2014; 124:963-72. [PMID: 24833353 DOI: 10.1182/blood-2013-11-539031] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
B-cell immune dysfunction contributes to the risk of severe infections after allogeneic hematopoietic stem cell transplantation (allo-HSCT). Delayed B-cell regeneration is found in patients with systemic graft-versus-host disease (GVHD) and is often accompanied by bone marrow (BM) suppression. Little is known about human BM GVHD. We analyzed the reconstitution kinetics of B-cell subsets in adult leukemic patients within 6 months after allo-HSCT. B-cell deficiency already existed before transplant and was aggravated after transplant. Onset of B-cell reconstitution characterized by transitional B-cell recovery occurred either early (months 2-3) or late (from month 6 on) and correlated highly positively with reverse transcription-polymerase chain reaction quantified numbers of κ-deleting recombination excision circles (KRECs). Delayed recovery was associated with systemic acute GVHD and full-intensity conditioning therapy. Histological analysis of BM trephines revealed increased T-cell infiltration in late recovering patients, which was associated with reduced numbers of osteoblasts. Functionally, late recovering patients displayed less pneumococcal polysaccharide-specific immunoglobin M-producing B cells on ex vivo B-cell activation than early recovering patients. Our results provide evidence for acute BM GVHD in allo-HSCT patients with infiltrating donor T cells and osteoblast destruction. This is associated with delayed B-cell reconstitution and impaired antibody response. Herein, KREC appears suitable to monitor BM B-cell output after transplant.
Collapse
|
238
|
Johnston HF, Xu Y, Racine JJ, Cassady K, Ni X, Wu T, Chan A, Forman S, Zeng D. Administration of anti-CD20 mAb is highly effective in preventing but ineffective in treating chronic graft-versus-host disease while preserving strong graft-versus-leukemia effects. Biol Blood Marrow Transplant 2014; 20:1089-103. [PMID: 24796279 DOI: 10.1016/j.bbmt.2014.04.028] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2014] [Accepted: 04/25/2014] [Indexed: 12/15/2022]
Abstract
Chronic graft-versus-host disease (cGVHD) is an autoimmune-like syndrome, and donor B cells play important roles in augmenting its pathogenesis. B cell-depleting anti-CD20 mAb has been administered before or after cGVHD onset for preventing or treating cGVHD in the clinic. Although administration before onset appeared to be more effective, the effect is variable and sometimes minimal. Here, we used 2 mouse cGVHD models to evaluate the preventive and therapeutic effect of anti-CD20 mAb. With the model of DBA/2 donor to MHC-matched BALB/c recipient, 1 intravenous injection of anti-CD20 mAb (40 mg/kg) the following day or on day 7 after hematopoietic cell transplantation when serum autoantibodies were undetectable effectively prevented induction of cGVHD and preserved a strong graft-versus-leukemia (GVL) effect. The separation of GVL effect from GVHD was associated with a significant reduction of donor CD4(+) T cell proliferation and expansion and protection of host thymic medullary epithelial cells. Anti-CD20 mAb administration also prevented expansion of donor T cells and induction of cGVHD in another mouse model of C57BL/6 donor to MHC-mismatched BALB/c recipients. In contrast, administration of anti-CD20 mAb after GVHD onset was not able to effectively deplete donor B cells or ameliorate cGVHD in either model. These results indicate that administration of anti-CD20 mAb before signs of cGVHD can prevent induction of autoimmune-like cGVHD while preserving a GVL effect; there is little effect if administered after cGVHD onset. This provides new insights into clinical prevention and therapy of cGVHD with B cell-depleting reagents.
Collapse
Affiliation(s)
- Heather F Johnston
- Departments of Diabetes Research and Hematology/Hematopoietic Cell Transplantation, Beckman Research Institute at City of Hope National Medical Center, Duarte, California; Irell and Manella Graduate School of Biological Sciences, Beckman Research Institute at City of Hope National Medical Center, Duarte, California
| | - Yajing Xu
- Departments of Diabetes Research and Hematology/Hematopoietic Cell Transplantation, Beckman Research Institute at City of Hope National Medical Center, Duarte, California; Department of Hematology, Xiangya Hospital, Central South University, Changsha, China
| | - Jeremy J Racine
- Departments of Diabetes Research and Hematology/Hematopoietic Cell Transplantation, Beckman Research Institute at City of Hope National Medical Center, Duarte, California; Irell and Manella Graduate School of Biological Sciences, Beckman Research Institute at City of Hope National Medical Center, Duarte, California
| | - Kaniel Cassady
- Departments of Diabetes Research and Hematology/Hematopoietic Cell Transplantation, Beckman Research Institute at City of Hope National Medical Center, Duarte, California; Irell and Manella Graduate School of Biological Sciences, Beckman Research Institute at City of Hope National Medical Center, Duarte, California
| | - Xiong Ni
- Departments of Diabetes Research and Hematology/Hematopoietic Cell Transplantation, Beckman Research Institute at City of Hope National Medical Center, Duarte, California; Department of Hematology, Changhai Hospital, The Second Military Medical School, Shanghai, China
| | - Tao Wu
- Departments of Diabetes Research and Hematology/Hematopoietic Cell Transplantation, Beckman Research Institute at City of Hope National Medical Center, Duarte, California; Department of Hematology, Changhai Hospital, The Second Military Medical School, Shanghai, China
| | - Andrew Chan
- Department of Research Biology, Genentech, San Francisco, California
| | - Stephen Forman
- Departments of Diabetes Research and Hematology/Hematopoietic Cell Transplantation, Beckman Research Institute at City of Hope National Medical Center, Duarte, California; Irell and Manella Graduate School of Biological Sciences, Beckman Research Institute at City of Hope National Medical Center, Duarte, California
| | - Defu Zeng
- Departments of Diabetes Research and Hematology/Hematopoietic Cell Transplantation, Beckman Research Institute at City of Hope National Medical Center, Duarte, California; Irell and Manella Graduate School of Biological Sciences, Beckman Research Institute at City of Hope National Medical Center, Duarte, California.
| |
Collapse
|
239
|
Ju JM, Lee H, Oh K, Lee DS, Choi EY. Kinetics of IFN-γ and IL-17 Production by CD4 and CD8 T Cells during Acute Graft-versus-Host Disease. Immune Netw 2014; 14:89-99. [PMID: 24851098 PMCID: PMC4022783 DOI: 10.4110/in.2014.14.2.89] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2013] [Revised: 03/16/2014] [Accepted: 03/27/2014] [Indexed: 02/07/2023] Open
Abstract
Graft-versus-host disease (GVHD) is a fatal complication that occurs after allogeneic hematopoietic stem cell transplantation. To understand the dynamics of CD4 and CD8 T cell production of IFN-γ and IL-17 during GVHD progression, we established a GVHD model by transplanting T cell-depleted bone marrow (TCD-BM) and purified T cells from B6 mice into irradiated BALB.B, creating an MHC-matched but minor histocompatibility (H) antigen-mismatched transplantation (B6 → BALB.B GVHD). Transplantation-induced GVHD was confirmed by the presence of the appropriate compositional changes in the T cell compartments and innate immune cells in the blood and the systemic secretion of inflammatory cytokines. Using this B6 → BALB.B GVHD model, we showed that the production of IFN-γ and IL-17 by CD4 T cells preceded that by CD8 T cells in the spleen, mesenteric lymph node, liver, and lung in the BALB.B GVHD host, and Th1 differentiation predated Th17 differentiation in all organs during GVHD progression. Such changes in cytokine production were based on changes in cytokine gene expression by the T cells at different time points during GVHD development. These results demonstrate that both IFN-γ and IL-17 are produced by CD4 and CD8 T cells but with different kinetics during GVHD progression.
Collapse
Affiliation(s)
- Ji-Min Ju
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 110-799, Korea
| | - Hakmo Lee
- Biomedical Research Institute, Seoul National University Hospital, Seoul 110-799, Korea
| | - Keunhee Oh
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 110-799, Korea
| | - Dong-Sup Lee
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 110-799, Korea
| | - Eun Young Choi
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 110-799, Korea
| |
Collapse
|
240
|
Total body irradiation and cyclophosphamide plus antithymocyte globulin regimen is well tolerated and promotes stable engraftment as a preparative regimen before T cell-replete haploidentical transplantation for acute leukemia. Biol Blood Marrow Transplant 2014; 20:1176-82. [PMID: 24747336 DOI: 10.1016/j.bbmt.2014.04.012] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2014] [Accepted: 04/10/2014] [Indexed: 11/20/2022]
Abstract
We compared total body irradiation (TBI, 700 cGy)/cyclophosphamide (Cy, 3.6 g/m(2))/simustine (250 mg/m(2)) plus antithymocyte globulin (ATG) (TBI/Cy plus ATG) with cytarabine (8 g/m(2))/i.v. busulfan (Bu, 9.6 mg/kg)/Cy (3.6 g/m(2))/simustine (250 mg/m(2)) plus ATG (modified Bu/Cy plus ATG) as preparative therapy in T cell-replete haploidentical hematopoietic stem cell transplantation (haplo-HSCT) for acute leukemia. From August 2009 to August 2013, 38 consecutive patients using TBI/Cy plus ATG regimen for T cell-replete haplo-HSCT (TBI group) at our center were eligible, which contained 28 high-risk and 10 standard-risk patients. A nested case-control study was designed. Seventy-seven patients using modified Bu/Cy plus ATG regimen (Bu group) were randomly selected in a 1 to 3:1 ratio matching for age, disease and status, year of HSCT (±2 years), and length of follow-up. Only 1 graft failure occurred in the TBI group. The incidence and time of neutrophil and platelet engraftment were comparable between the 2 groups. Severe grades III/IV graft-versus-host disease was observed in 13.4% of Bu group and only 2.6% of TBI group (P = .083). More toxicity of the liver (37.7% versus 10.5%; P = .002) and more hemorrhagic cystitis occurred in the Bu group (49.3% versus 23.7%, P = .008). Diarrhea was more common in the TBI group (44.7% versus 22.1%; P = .031). No significant differences were found in the 2-year incidences of relapse (26.5% for TBI group versus 32.3% for Bu group, P = .742), 1-year transplant-related mortality (12.6% versus 16.2%, P = .862), 2-year overall survival (60.2% versus 57.0%, P = .937), and 2-year incidence of disease-free survival (57.9% versus 56.6%, P = .845) between the 2 groups. We conclude that the TBI/Cy plus ATG regimen seems to be feasible in T cell-replete haplo-HSCT, which promotes stable engraftment and a lower incidence of liver toxicity and hemorrhagic cystitis. However, longer follow-up is necessary to determine the late relapse rate and late toxicity.
Collapse
|
241
|
Park G, Choi YJ, Lee SE, Lim JY, Lee C, Choi EY, Min CK. A paradoxical pattern of indoleamine 2,3-dioxygenase expression in the colon tissues of patients with acute graft-versus-host disease. Exp Hematol 2014; 42:734-40. [PMID: 24732701 DOI: 10.1016/j.exphem.2014.04.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2013] [Revised: 03/31/2014] [Accepted: 04/04/2014] [Indexed: 10/25/2022]
Abstract
Indoleamine 2,3-dioxygenase (IDO) is a rate-limiting enzyme for tryptophan catabolism that plays an important role in the induction of immune tolerance. It is induced in the colon and exerts its effects there, regulating T-cell proliferation and survival. To address the role of IDO in acute graft-versus-host disease (AGVHD) after human allogeneic hematopoietic stem cell transplantation, we analyzed the relationship between IDO expression in colon tissues and clinical outcomes among 41 AGVHD patients who were diagnosed as gut AGVHD by a colon mucosal biopsy within 100 days posttransplantation. By in situ immunohistochemical analyses, IDO expression was measured in colon mucosal mononuclear cells (MNCs) and endothelial cells (ECs) in GVHD areas. High IDO expression in MNCs and low IDO expression in ECs had a trend toward a lower nonrelapse mortality (p = 0.157 and p = 0.062, respectively). Multivariate analysis showed that high MNC combined with low EC IDO expression (p = 0.046), as well as low disease risk (p = 0.012), are associated with lower nonrelapse mortality. Paradoxical upregulation of IDO expression in colon MNCs and ECs may represent a new predictive factor for prognosis in gut AGVHD after human allogeneic hematopoietic stem cell transplantation.
Collapse
Affiliation(s)
- Gyeongsin Park
- Department of Pathology, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, Korea; Cancer Research Institute, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Yeong-Jin Choi
- Department of Pathology, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, Korea
| | - Sung-Eun Lee
- Department of Internal Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, Korea
| | - Ji-Young Lim
- Department of Internal Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, Korea
| | - Chulbom Lee
- Department of Chemistry, College of Natural Science, Seoul National University, Seoul, Korea
| | - Eun Young Choi
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea
| | - Chang-Ki Min
- Department of Internal Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, Korea.
| |
Collapse
|
242
|
Abstract
Activation of sialic-acid-binding immunoglobulin-like lectin-G (Siglec-G) by noninfectious damage-associated molecular patterns controls innate immune responses. However, whether it also regulates T-cell-mediated adaptive immune responses is not known. Graft-versus-host reaction is a robust adaptive immune response caused by allogeneic hematopoietic cell transplantation that have been activated by antigen-presenting cells (APCs) in the context of damaged host tissues following allogeneic hematopoietic cell transplantation. The role of infectious and noninfectious pattern recognition receptor-mediated activation in the induction and aggravation of graft-versus-host disease (GVHD) is being increasingly appreciated. But the role of pathways that control innate immune responses to noninfectious stimuli in modulating GVHD has heretofore not been recognized. We report that Siglec-G expression on host APCs, specifically on hematopoietic cells, negatively regulates GVHD in multiple clinically relevant murine models. Mechanistic studies with various relevant Siglec-G and CD24 knockout mice and chimeric animals, along with rescue experiments with novel CD24 fusion protein demonstrate that enhancing the interaction between Siglec-G on host APCs with CD24 on donor T cells attenuates GVHD. Taken together, our data demonstrate that Siglec-G-CD24 axis, controls the severity of GVHD and suggest that enhancing this interaction may represent a novel strategy for mitigating GVHD.
Collapse
|
243
|
Jordan KKI, Christensen IJ, Heilmann C, Sengeløv H, Müller KG. Pretransplant C-reactive Protein as A Prognostic Marker in Allogeneic Stem Cell Transplantation. Scand J Immunol 2014; 79:206-13. [DOI: 10.1111/sji.12137] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2013] [Accepted: 11/25/2013] [Indexed: 01/30/2023]
Affiliation(s)
- K. K. I. Jordan
- Department of Pediatric and Adolescent Medicine 4072 and Institute for Inflammation Research 7541; National University Hospital; Rigshospitalet; Copenhagen Denmark
| | - I. J. Christensen
- The Finsen Laboratory; Rigshospitalet and Biotech Research and Innovation Center (BRIC); University of Copenhagen; Copenhagen Denmark
| | - C. Heilmann
- Department of Pediatric and Adolescent Medicine 4072 and Institute for Inflammation Research 7541; National University Hospital; Rigshospitalet; Copenhagen Denmark
| | - H. Sengeløv
- Department of Haematology 4042; National University Hospital; Rigshospitalet; Copenhagen Denmark
| | - K. G. Müller
- Department of Pediatric and Adolescent Medicine 4072 and Institute for Inflammation Research 7541; National University Hospital; Rigshospitalet; Copenhagen Denmark
| |
Collapse
|
244
|
Suh JH, Kanathezhath B, Shenvi S, Guo H, Zhou A, Tiwana A, Kuypers F, Ames BN, Walters MC. Thiol/redox metabolomic profiling implicates GSH dysregulation in early experimental graft versus host disease (GVHD). PLoS One 2014; 9:e88868. [PMID: 24558439 PMCID: PMC3928313 DOI: 10.1371/journal.pone.0088868] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2013] [Accepted: 01/15/2014] [Indexed: 11/18/2022] Open
Abstract
Graft-versus-host disease (GVHD) is a common complication of allogeneic bone marrow transplantation (BMT). Upregulation of inflammatory cytokines precedes the clinical presentation of GVHD and predicts its severity. In this report, thiol/redox metabolomics was used to identify metabolic perturbations associated with early preclinical (Day+4) and clinical (Day+10) stages of GVHD by comparing effects in Syngeneic (Syn; major histocompatibility complex- identical) and allogeneic transplant recipients (Allo BMT) in experimental models. While most metabolic changes were similar in both groups, plasma glutathione (GSH) was significantly decreased, and GSH disulfide (GSSG) was increased after allogeneic compared to syngeneic recipient and non-transplant controls. The early oxidation of the plasma GSH/GSSG redox couple was also observed irrespective of radiation conditioning treatment and was accompanied by significant rise in hepatic protein oxidative damage and ROS generation. Despite a significant rise in oxidative stress, compensatory increase in hepatic GSH synthesis was absent following Allo BMT. Early shifts in hepatic oxidative stress and plasma GSH loss preceded a statistically significant rise in TNF-α. To identify metabolomic biomarkers of hepatic GVHD injury, plasma metabolite concentrations analyzed at Day+10 were correlated with hepatic organ injury. GSSG (oxidized GSH) and β-alanine, were positively correlated, and plasma GSH cysteinylglycine, and branched chain amino acids were inversely correlated with hepatic injury. Although changes in plasma concentrations of cysteine, cystathionine (GSH precursors) and cysteinylglycine (a GSH catabolite) were not significant by univariate analysis, principal component analysis (PCA) indicated that accumulation of these metabolites after Allo BMT contributed significantly to early GVHD in contrast to Syn BMT. In conclusion, thiol/redox metabolomic profiling implicates that early dysregulation of host hepatic GSH metabolism and oxidative stress in sub-clinical GVHD before elevated TNF-α levels is associated with GVHD pathogenesis. Future studies will probe the mechanisms for these changes and examine the potential of antioxidant intervention strategies to modulate GVHD.
Collapse
Affiliation(s)
- Jung H. Suh
- Children’s Hospital Oakland Research Institute, Oakland, California, United States of America
- * E-mail: (JHS); (MCW)
| | - Bindu Kanathezhath
- Children’s Hospital and Research Center Oakland, Oakland, California, United States of America
- Division of Blood and Marrow Transplantation, Children’s Hospital and Research Center Oakland, Oakland, California, United States of America
| | - Swapna Shenvi
- Children’s Hospital Oakland Research Institute, Oakland, California, United States of America
| | - Hua Guo
- Children’s Hospital and Research Center Oakland, Oakland, California, United States of America
- Department of Pathology, Children’s Hospital and Research Center Oakland, Oakland, California, United States of America
| | - Alicia Zhou
- Children’s Hospital Oakland Research Institute, Oakland, California, United States of America
| | - Anureet Tiwana
- Children’s Hospital Oakland Research Institute, Oakland, California, United States of America
| | - Frans Kuypers
- Children’s Hospital Oakland Research Institute, Oakland, California, United States of America
| | - Bruce N. Ames
- Children’s Hospital Oakland Research Institute, Oakland, California, United States of America
| | - Mark C. Walters
- Children’s Hospital and Research Center Oakland, Oakland, California, United States of America
- Division of Blood and Marrow Transplantation, Children’s Hospital and Research Center Oakland, Oakland, California, United States of America
- * E-mail: (JHS); (MCW)
| |
Collapse
|
245
|
Marchetti G, Cozzi-Lepri A, Tincati C, Calcagno A, Ceccherini-Silberstein F, De Luca A, Antinori A, Castagna A, Puoti M, Monforte AD. Immune activation and microbial translocation in liver disease progression in HIV/hepatitis co-infected patients: results from the Icona Foundation study. BMC Infect Dis 2014; 14:79. [PMID: 24520976 PMCID: PMC3923986 DOI: 10.1186/1471-2334-14-79] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2013] [Accepted: 02/05/2014] [Indexed: 01/11/2023] Open
Abstract
Background We evaluated whether immune activation (IA) and microbial translocation (MT) might play a role in accelerating liver disease progression in HIV-HBV/HCV co-infected patients. Methods ART-naïve HIV/viral hepatitis co-infected patients from Icona with a CD4 cell count >200/μl and with a known date of prior HIV neg/pos tests and ≥1 plasma sample stored were included in the study. Plasma MT (LPS, sCD14) and IA (IL-6,TNFα) were measured using ELISA while activated CD8 + CD38 + HLA-DR + were measured by flow cytometry, with one measurement being performed for all patients and two measurements for a smaller group of subjects. The association between these biomarkers and the time to i) a single ALT >200 IU/l and ii) a Fib-4 >1.45 was also investigated. A standard survival analysis with robust standard errors was used for all evaluations. Follow-up was censored at patients’ last clinical follow-up. Results We studied 127 HIV-infected hepatitis viruses co-infected patients (118 HCV, 9 HBV). Overall median (IQR) CD4, VL, age were 596/μl (208–1303), 3.8 log10cp/mL (3–4.3), 34 years (22–56). While heightened TNF-α was associated with a 13-fold increased risk of Fib-4 > 1.45 (RH 13.05, 95% CI 2.43-70; p = 0.003), markers of MT did not show an association with liver illness. Interestingly, higher sCD14 was associated with a decreased risk of Fib-4 > 1.45, independently of other biomarkers considered (RH 0.20, 95% CI 0.04-0,9; p = 0.04). Conclusions In HIV/hepatitis virus co-infected ART-naive patients, higher TNF-α plasma levels were associated with a 13-fold increase in the risk of progression to a Fib-4 >1.45, suggesting that the pro-inflammatory status in HIV infection might hasten the course of HCV. In view of the fact that sCD14 may hinder the interaction between LPS and the phagocyte membrane CD14, we herewith propose a model which aims to demonstrate that high sCD14 levels might contribute to shelter liver function through the down-regulation of the inflammatory cascade.
Collapse
Affiliation(s)
- Giulia Marchetti
- Department of Health Sciences- Clinic of Infectious Diseases - "San Paolo" Hospital, University of Milan, via A, di Rudinì, 8-20142 Milan, Italy.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
246
|
Mercadante ACT, Perobelli SM, Alves APG, Gonçalves-Silva T, Mello W, Gomes-Santos AC, Miyoshi A, Azevedo V, Faria AMC, Bonomo A. Oral combined therapy with probiotics and alloantigen induces B cell-dependent long-lasting specific tolerance. THE JOURNAL OF IMMUNOLOGY 2014; 192:1928-37. [PMID: 24453248 DOI: 10.4049/jimmunol.1301034] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Allogeneic hematopietic stem cell transplantation (aHSCT) is widely used for the treatment of hematologic malignancies. Although aHSCT provides a good response against the malignant cells (graft-versus-leukemia [GVL]), it also leads to the development of graft-versus-host disease (GVHD), a severe disease with high mortality and morbidity rates. Therapy for GVHD is commonly based on nonspecific immunosupression of the transplanted recipient, resulting in the concomitant inhibition of the GVL effect. In this study, we propose an alternative approach to specifically suppress GVHD while sparing the GVL, based on oral treatment of transplant donors with recipient Ags, associated with the intake of probiotic Lactococcus lactis as tolerogenic adjuvant (combined therapy). We show that treatment of C57BL/6 donor mice with combined therapy before the transplant protects the recipients F1 (C57BL/6 × BAL/c) mice from clinical and pathological manifestations of disease, resulting in 100% survival rate. Importantly, the animals keep the immunological competence maintaining the GVL response as well as the response to third-party Ags. The protection is specific, long lasting and dependent on donor IL-10-sufficient B cells activity, which induces regulatory T cells in the host. These data suggest that combined therapy is a promising strategy for prevention of GVHD with preservation of GVL, opening new possibilities to treat human patients subjected to transplantation.
Collapse
Affiliation(s)
- Ana C T Mercadante
- Department of Experimental Medicine, Brazilian National Cancer Institute, Rio de Janeiro 20231-050, Brazil
| | | | | | | | | | | | | | | | | | | |
Collapse
|
247
|
Normanton M, Marti LC. Current data on IL-17 and Th17 cells and implications for graft versus host disease. EINSTEIN-SAO PAULO 2014; 11:237-46. [PMID: 23843069 PMCID: PMC4872902 DOI: 10.1590/s1679-45082013000200019] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2012] [Accepted: 04/27/2013] [Indexed: 12/21/2022] Open
Abstract
Human interleukin 17 was first described in 1995 as a new cytokine produced primarily by activated T CD4+ cells that stimulate the secretion of IL-6 and IL-8 by human fibroblasts, besides increasing the expression of ICAM-1. Various authors have reported that IL-17A has a role in the protection of organisms against extracellular bacteria and fungi due to the capacity of IL-17A to recruit neutrophils to the areas of infection, evidencing a pathological role in various models of autoimmune diseases, such as experimental autoimmune encephalitis and arthritis. The participation of IL-17A has also been described in the acute rejection of organ transplants and graft versus host disease. However, the greatest revolution in research with IL-17 happened in 2000, when it was proposed that IL-17 cannot be classified as Th1 or Th2, but rather, simply as a new lineage of IL-17-producing T-cells. These findings modified the previously established Th1/Th2 paradigm, leading to the definition of the CD3+ CD4+ Th17 cellular subtype and establishment of a new model to explain the origin of various immune events, as well as its implication in the graft versus host disease that is discussed in depth in this article.
Collapse
|
248
|
Optimization of the therapeutic efficacy of human umbilical cord blood-mesenchymal stromal cells in an NSG mouse xenograft model of graft-versus-host disease. Cytotherapy 2014; 16:298-308. [PMID: 24418403 DOI: 10.1016/j.jcyt.2013.10.012] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2013] [Revised: 08/28/2013] [Accepted: 10/22/2013] [Indexed: 01/10/2023]
Abstract
BACKGROUND AIMS Although in vitro studies have demonstrated the immunosuppressive capacity of mesenchymal stromal cells (MSCs), most in vivo studies on graft-versus-host disease (GVHD) have focused on prevention, and the therapeutic effect of MSCs is controversial. Moreover, optimal time intervals for infusing MSCs have not been established. METHODS We attempted to evaluate whether human umbilical cord blood-MSCs (hUCB-MSCs) could either prevent or treat GVHD in an NSG mouse xenograft model by injection of MSCs before or after in vivo clearance. Mice were infused with either a single dose or multiple doses of 5 × 10(5) hUCB-MSCs (3- or 7-day intervals) before or after GVHD onset. RESULTS Before onset, hUCB-MSCs significantly improved the survival rate only when repeatedly injected at 3-day intervals. In contrast, single or repeated injections after GVHD onset significantly increased the survival rate and effectively attenuated tissue damage and inflammation. Furthermore, the levels of prostaglandin E2 and transforming growth factor-β1 increased significantly, whereas the level of interferon-γ decreased significantly in all MSC treatment groups. CONCLUSIONS These data establish the optimal time intervals for preventing GVHD and show that hUCB-MSCs effectively attenuated symptoms and improved survival rate when administered after the onset of GVDH.
Collapse
|
249
|
Iwasaki T, Shibasaki S. Hepatocyte growth factor regulates immune reactions caused by transplantation and autoimmune diseases. YAKUGAKU ZASSHI 2013; 133:1159-67. [PMID: 24189557 DOI: 10.1248/yakushi.13-00212-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Hepatocyte growth factor (HGF) was first identified and cloned as a mitogenic protein for hepatocytes, and subsequent studies revealed that HGF has multiple biological effects on a wide variety of cells, including mitogenic, motogenic, morphogenic, anti-apoptotic, and angiogenic activities. It plays roles in organizing tissues during development and regeneration. HGF may be applied for the treatment of acute onset diseases such as fulminant hepatitis, myocardial infarction, acute renal failure, cerebral infarction, and chronic diseases like liver cirrhosis, chronic renal failure, pulmonary fibrosis, cardiomyopathy, and arteriosclerosis obliterans. HGF also has immunomodulatory activities and we previously demonstrated that its administration inhibited acute graft-versus-host disease (GVHD) after treatment with hematopoietic stem cell transplantation. We also demonstrated that HGF inhibited lupus nephritis induced by chronic GVHD and dermal sclerosis in systemic sclerosis using model mice. More than 7 hundred thousand patients suffer from rheumatoid arthritis (RA) in Japan. Although the prognosis of these patients has improved by the treatment of biological agents such as TNF-α and IL-6 blockers, there remain many for whom these agents have not proved beneficial. Recently, using RA model mice, we demonstrated that the HGF antagonist, NK4, can block disease progression of RA through its anti-angiogenic and immunomodulatory actions. In this review article, we discuss the possible roles of HGF signaling for the treatment of immunological reactions in transplantation and autoimmune diseases.
Collapse
Affiliation(s)
- Tsuyoshi Iwasaki
- Division of Pharmacotherapy, Department of Pharmacy, School of Pharmacy, Hyogo University of Health Sciences
| | | |
Collapse
|
250
|
Vadhan-Raj S, Goldberg JD, Perales MA, Berger DP, Brink MRM. Clinical applications of palifermin: amelioration of oral mucositis and other potential indications. J Cell Mol Med 2013; 17:1371-84. [PMID: 24251854 PMCID: PMC4117550 DOI: 10.1111/jcmm.12169] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2013] [Accepted: 09/16/2013] [Indexed: 12/25/2022] Open
Abstract
Mucositis is one of the most significant toxicities in cancer patients undergoing cytotoxic treatment. It can have a negative impact on both quality of life and health economics. Severe oral mucositis can contribute to hospitalization, need for narcotic analgesics, total parentral nutrition, suboptimal delivery of anti-neoplastic treatment, and morbidity and mortality. Palifermin, a recombinant derivative of human keratinocyte growth factor, is the first active agent approved by the FDA for the prevention of severe oral mucositis in patients undergoing haematopoietic stem cell transplantation (HSCT). Several studies have also shown significant reduction in the incidence, severity and/or duration of oral mucositis in other high-risk settings such as concurrent chemoradiotherapy (CT/RT) for patients with head and neck cancer, and use of mucotoxic chemotherapeutic agents such as doxorubicin in sarcoma and fluorouracil for the treatment of colorectal cancer. The reduction in mucositis has translated into amelioration of symptoms and improvement in daily functioning as measured by patient-reported outcome in multiple studies. The clinical response to palifermin appears to be related in part to epithelial proliferation and mucosal thickening. Palifermin also has other potential clinical applications including the acceleration of immune reconstitution and inhibition of graft-versus-host disease in patients undergoing HSCT, and mitigation of dysphagia in lung cancer patients treated with concurrent CT/RT. Palifermin is generally well tolerated with mild-to-moderate skin and oral adverse events. Future studies may expand the use of palifermin into other areas that would benefit from its cytoprotective and regenerative effects.
Collapse
Affiliation(s)
- Saroj Vadhan-Raj
- Department of Sarcoma Medical Oncology, Section of Cytokines and Supportive Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer CenterHouston, TX, USA
| | - Jenna D Goldberg
- Department of Medicine, Adult Bone Marrow Transplantation Service, Memorial Sloan-Kettering Cancer CenterNew York, NY, USA
- Weill Cornell Medical CollegeNew York, NY, USA
| | - Miguel-Angel Perales
- Department of Medicine, Adult Bone Marrow Transplantation Service, Memorial Sloan-Kettering Cancer CenterNew York, NY, USA
- Weill Cornell Medical CollegeNew York, NY, USA
| | - Dietmar P Berger
- Amgen Inc.Thousand Oaks, CA, USA
- Department of Oncology, Genentech Inc.South San Francisco, CA, USA
| | - Marcel RM Brink
- Department of Medicine, Adult Bone Marrow Transplantation Service, Memorial Sloan-Kettering Cancer CenterNew York, NY, USA
- Weill Cornell Medical CollegeNew York, NY, USA
| |
Collapse
|