1
|
Lyu F, Gong H, Wu X, Liu X, Lu Y, Wei X, Liu C, Shen Y, Wang Y, Lei L, Chen J, Ma S, Sun H, Yu D, Han J, Xu Y, Wu D. Dimethyl fumarate ameliorates chronic graft-versus-host disease by inhibiting Tfh differentiation via Nrf2. Leukemia 2025; 39:473-481. [PMID: 39580582 DOI: 10.1038/s41375-024-02475-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 10/26/2024] [Accepted: 11/13/2024] [Indexed: 11/25/2024]
Abstract
Chronic graft-versus-host disease (cGVHD), characterized by chronic tissue inflammation and fibrosis involving multiple organs, remains a major complication after allogeneic hematopoietic stem cell transplantation (allo-HSCT). Dimethyl fumarate (DMF) is an anti-inflammatory drug approved for the treatment of multiple sclerosis and psoriasis. We previously reported that DMF effectively inhibits acute GVHD (aGVHD) while preserving the graft-versus-leukemia effect. However, the role of DMF in cGVHD progression remains unknown. Here, we found that DMF administration significantly suppresses follicular helper T cell (Tfh) differentiation, and germinal center formation and alleviates disease severity in different murine cGVHD models. Mechanistically, DMF treatment downregulates IL-21 transcription by activation of Nrf2, thus orchestrating Tfh-related gene programs both in mice and humans. The inhibitory role of DMF on Tfh cell differentiation was diminished in Nrf2 deficient T cells. Importantly, the therapeutic potential of DMF in clinical cGVHD has been validated in human data whereby DMF effectively reduces IL-21 production and Tfh cell generation in peripheral blood mononuclear cells from active cGVHD patients and further attenuates xenograft GVHD. Collectively, our findings reveal that DMF potently inhibits cGVHD development by repressing Tfh cell differentiation via Nrf2, paving the way for the treatment of cGVHD in the clinic.
Collapse
Affiliation(s)
- Fulian Lyu
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
- Key Laboratory of Stem Cells and Biomedical Materials of Jiangsu Province and Chinese Ministry of Science and Technology, Suzhou, China
| | - Huanle Gong
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China.
- Key Laboratory of Stem Cells and Biomedical Materials of Jiangsu Province and Chinese Ministry of Science and Technology, Suzhou, China.
| | - Xiaojin Wu
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
- Key Laboratory of Stem Cells and Biomedical Materials of Jiangsu Province and Chinese Ministry of Science and Technology, Suzhou, China
| | - Xin Liu
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
- Key Laboratory of Stem Cells and Biomedical Materials of Jiangsu Province and Chinese Ministry of Science and Technology, Suzhou, China
| | - Yinghao Lu
- Department of Hematology, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
| | - Xiya Wei
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Chenchen Liu
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Yaoyao Shen
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
- Key Laboratory of Stem Cells and Biomedical Materials of Jiangsu Province and Chinese Ministry of Science and Technology, Suzhou, China
| | - Yuhang Wang
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Lei Lei
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
- Key Laboratory of Stem Cells and Biomedical Materials of Jiangsu Province and Chinese Ministry of Science and Technology, Suzhou, China
| | - Jia Chen
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
- Key Laboratory of Stem Cells and Biomedical Materials of Jiangsu Province and Chinese Ministry of Science and Technology, Suzhou, China
| | - Shoubao Ma
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Los Angeles, CA, USA
| | - Hongjian Sun
- Frazer Institute, Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia
| | - Di Yu
- Frazer Institute, Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia
- Ian Frazer Centre for Children's Immunotherapy Research, Child Health Research Centre, Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia
| | - JingJing Han
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China.
- Key Laboratory of Stem Cells and Biomedical Materials of Jiangsu Province and Chinese Ministry of Science and Technology, Suzhou, China.
| | - Yang Xu
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China.
- Key Laboratory of Stem Cells and Biomedical Materials of Jiangsu Province and Chinese Ministry of Science and Technology, Suzhou, China.
| | - Depei Wu
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China.
- Key Laboratory of Stem Cells and Biomedical Materials of Jiangsu Province and Chinese Ministry of Science and Technology, Suzhou, China.
| |
Collapse
|
2
|
Xu R, Zhang L, Pan H, Zhang Y. Retinoid X receptor heterodimers in hepatic function: structural insights and therapeutic potential. Front Pharmacol 2024; 15:1464655. [PMID: 39478961 PMCID: PMC11521896 DOI: 10.3389/fphar.2024.1464655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Accepted: 09/30/2024] [Indexed: 11/02/2024] Open
Abstract
Nuclear receptors (NRs) are key regulators of multiple physiological functions and pathological changes in the liver in response to a variety of extracellular signaling changes. Retinoid X receptor (RXR) is a special member of the NRs, which not only responds to cellular signaling independently, but also regulates multiple signaling pathways by forming heterodimers with various other NR. Therefore, RXR is widely involved in hepatic glucose metabolism, lipid metabolism, cholesterol metabolism and bile acid homeostasis as well as hepatic fibrosis. Specific activation of particular dimers regulating physiological and pathological processes may serve as important pharmacological targets. So here we describe the basic information and structural features of the RXR protein and its heterodimers, focusing on the role of RXR heterodimers in a number of physiological processes and pathological imbalances in the liver, to provide a theoretical basis for RXR as a promising drug target.
Collapse
Affiliation(s)
- Renjie Xu
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Linyue Zhang
- Department of Ultrasound, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hao Pan
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yong Zhang
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
3
|
Nguyen JT, Jessri M, Costa-da-Silva AC, Sharma R, Mays JW, Treister NS. Oral Chronic Graft-Versus-Host Disease: Pathogenesis, Diagnosis, Current Treatment, and Emerging Therapies. Int J Mol Sci 2024; 25:10411. [PMID: 39408739 PMCID: PMC11476840 DOI: 10.3390/ijms251910411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 09/16/2024] [Accepted: 09/23/2024] [Indexed: 10/20/2024] Open
Abstract
Chronic graft-versus-host disease (cGvHD) is a multisystem disorder that occurs in recipients of allogeneic hematopoietic (alloHCT) stem cell transplants and is characterized by both inflammatory and fibrotic manifestations. It begins with the recognition of host tissues by the non-self (allogeneic) graft and progresses to tissue inflammation, organ dysfunction and fibrosis throughout the body. Oral cavity manifestations of cGVHD include mucosal features, salivary gland dysfunction and fibrosis. This review synthesizes current knowledge on the pathogenesis, diagnosis and management of oral cGVHD, with a focus on emerging trends and novel therapeutics. Data from various clinical studies and expert consensus are integrated to provide a comprehensive overview.
Collapse
Affiliation(s)
- Joe T. Nguyen
- Nguyen Laboratory, Head and Neck Cancer Section, Surgical Oncology Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
- Oral Immunobiology Unit, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA; (A.C.C.-d.-S.); (R.S.); (J.W.M.)
| | - Maryam Jessri
- Metro North Hospital and Health Service, Queensland Health, Brisbane, QLD 4029, Australia;
- Department of Oral Medicine and Pathology, School of Dentistry, The University of Queensland, Herston, QLD 4072, Australia
| | - Ana C. Costa-da-Silva
- Oral Immunobiology Unit, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA; (A.C.C.-d.-S.); (R.S.); (J.W.M.)
| | - Rubina Sharma
- Oral Immunobiology Unit, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA; (A.C.C.-d.-S.); (R.S.); (J.W.M.)
| | - Jacqueline W. Mays
- Oral Immunobiology Unit, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA; (A.C.C.-d.-S.); (R.S.); (J.W.M.)
| | - Nathaniel S. Treister
- Division of Oral Medicine and Dentistry, Brigham and Women’s Hospital, Boston, MA 02115, USA
- Department of Oral Medicine, Infection and Immunity, Harvard School of Dental Medicine, Boston, MA 02114, USA
| |
Collapse
|
4
|
Lee SK, Park SS, Park S, Lee SE, Cho BS, Eom KS, Kim YJ, Kim HJ, Min CK, Cho SG, Lee JW, Lee S, Kim Y, Han JW, Yang H, Bae SH, Jang JW, Choi JY, Yoon SK, Lee DY, Lee SH, Yoon JH, Sung PS. The Impact of Histologic Portal T-Cell Density on the Clinical Outcomes in Hepatic Graft-versus-Host Disease and Autoimmune Liver Diseases. Diagnostics (Basel) 2024; 14:1745. [PMID: 39202234 PMCID: PMC11353783 DOI: 10.3390/diagnostics14161745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 08/05/2024] [Accepted: 08/08/2024] [Indexed: 09/03/2024] Open
Abstract
Hepatic graft-versus-host disease (GVHD) significantly impacts morbidity and mortality among allogeneic hematopoietic stem cell transplant recipients. However, the relationship between clinical and immunopathological phenotypes and their influence on clinical outcomes in hepatic GVHD is not well understood. In this study, we aimed to study the implications of portal T-cell infiltration on the clinical outcomes in hepatic GHVD and its similarities to autoimmune liver disease. We analyzed 78 patients with biopsy-confirmed hepatic GVHD (n = 38) or autoimmune liver disease (n = 40) between 2016 and 2021. The cholestatic variant was defined by an R-value < 2.0, based on the ratio of alanine aminotransferase to alkaline phosphatase. The primary outcome was the biochemical response at 4 (early) and 8-12 (late) weeks after corticosteroid treatment. In hepatic GVHD patients, the hepatitic variant (n = 19) showed greater CD3+ T-cell infiltration than the cholestatic variant (n = 19; p < 0.001). No significant differences were observed in the infiltration of CD20+, CD38+, or CD68+ cells. The hepatitic variant had significantly better early and late responses and higher liver-related event-free survival than the cholestatic variants (p < 0.05). Concerning autoimmune liver diseases, the autoimmune hepatitis (AIH) group had significantly more portal T-cell infiltration and better treatment responses than the primary biliary cholangitis (PBC) group. In conclusion, higher portal T-cell infiltration may be associated with better clinical outcomes in patients with hepatic GVHD. Additionally, this study highlights similarities in portal T-cell infiltration and treatment response patterns between AIH and the hepatitic variant, as well as PBC and the cholestatic variant.
Collapse
Affiliation(s)
- Soon Kyu Lee
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Incheon St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea;
- The Catholic University Liver Research Centre, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (J.W.H.); (H.Y.); (S.H.B.); (J.W.J.); (J.Y.C.); (S.K.Y.); (D.Y.L.)
| | - Sung-Soo Park
- Department of Hematology, Catholic Hematology Hospital and Leukemia Research Institute, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (S.-S.P.); (S.P.); (S.-E.L.); (B.-S.C.); (K.-S.E.); (Y.-J.K.); (H.-J.K.); (C.-K.M.); (S.-G.C.); (J.W.L.); (S.L.)
| | - Silvia Park
- Department of Hematology, Catholic Hematology Hospital and Leukemia Research Institute, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (S.-S.P.); (S.P.); (S.-E.L.); (B.-S.C.); (K.-S.E.); (Y.-J.K.); (H.-J.K.); (C.-K.M.); (S.-G.C.); (J.W.L.); (S.L.)
| | - Sung-Eun Lee
- Department of Hematology, Catholic Hematology Hospital and Leukemia Research Institute, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (S.-S.P.); (S.P.); (S.-E.L.); (B.-S.C.); (K.-S.E.); (Y.-J.K.); (H.-J.K.); (C.-K.M.); (S.-G.C.); (J.W.L.); (S.L.)
| | - Byung-Sik Cho
- Department of Hematology, Catholic Hematology Hospital and Leukemia Research Institute, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (S.-S.P.); (S.P.); (S.-E.L.); (B.-S.C.); (K.-S.E.); (Y.-J.K.); (H.-J.K.); (C.-K.M.); (S.-G.C.); (J.W.L.); (S.L.)
| | - Ki-Seong Eom
- Department of Hematology, Catholic Hematology Hospital and Leukemia Research Institute, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (S.-S.P.); (S.P.); (S.-E.L.); (B.-S.C.); (K.-S.E.); (Y.-J.K.); (H.-J.K.); (C.-K.M.); (S.-G.C.); (J.W.L.); (S.L.)
| | - Yoo-Jin Kim
- Department of Hematology, Catholic Hematology Hospital and Leukemia Research Institute, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (S.-S.P.); (S.P.); (S.-E.L.); (B.-S.C.); (K.-S.E.); (Y.-J.K.); (H.-J.K.); (C.-K.M.); (S.-G.C.); (J.W.L.); (S.L.)
| | - Hee-Je Kim
- Department of Hematology, Catholic Hematology Hospital and Leukemia Research Institute, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (S.-S.P.); (S.P.); (S.-E.L.); (B.-S.C.); (K.-S.E.); (Y.-J.K.); (H.-J.K.); (C.-K.M.); (S.-G.C.); (J.W.L.); (S.L.)
| | - Chang-Ki Min
- Department of Hematology, Catholic Hematology Hospital and Leukemia Research Institute, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (S.-S.P.); (S.P.); (S.-E.L.); (B.-S.C.); (K.-S.E.); (Y.-J.K.); (H.-J.K.); (C.-K.M.); (S.-G.C.); (J.W.L.); (S.L.)
| | - Seok-Goo Cho
- Department of Hematology, Catholic Hematology Hospital and Leukemia Research Institute, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (S.-S.P.); (S.P.); (S.-E.L.); (B.-S.C.); (K.-S.E.); (Y.-J.K.); (H.-J.K.); (C.-K.M.); (S.-G.C.); (J.W.L.); (S.L.)
| | - Jong Wook Lee
- Department of Hematology, Catholic Hematology Hospital and Leukemia Research Institute, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (S.-S.P.); (S.P.); (S.-E.L.); (B.-S.C.); (K.-S.E.); (Y.-J.K.); (H.-J.K.); (C.-K.M.); (S.-G.C.); (J.W.L.); (S.L.)
| | - Seok Lee
- Department of Hematology, Catholic Hematology Hospital and Leukemia Research Institute, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (S.-S.P.); (S.P.); (S.-E.L.); (B.-S.C.); (K.-S.E.); (Y.-J.K.); (H.-J.K.); (C.-K.M.); (S.-G.C.); (J.W.L.); (S.L.)
| | - Younghoon Kim
- Department of Hospital Pathology, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea;
| | - Ji Won Han
- The Catholic University Liver Research Centre, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (J.W.H.); (H.Y.); (S.H.B.); (J.W.J.); (J.Y.C.); (S.K.Y.); (D.Y.L.)
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Hyun Yang
- The Catholic University Liver Research Centre, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (J.W.H.); (H.Y.); (S.H.B.); (J.W.J.); (J.Y.C.); (S.K.Y.); (D.Y.L.)
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Eunpyeong St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Si Hyun Bae
- The Catholic University Liver Research Centre, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (J.W.H.); (H.Y.); (S.H.B.); (J.W.J.); (J.Y.C.); (S.K.Y.); (D.Y.L.)
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Eunpyeong St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Jeong Won Jang
- The Catholic University Liver Research Centre, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (J.W.H.); (H.Y.); (S.H.B.); (J.W.J.); (J.Y.C.); (S.K.Y.); (D.Y.L.)
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Jong Young Choi
- The Catholic University Liver Research Centre, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (J.W.H.); (H.Y.); (S.H.B.); (J.W.J.); (J.Y.C.); (S.K.Y.); (D.Y.L.)
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Seung Kew Yoon
- The Catholic University Liver Research Centre, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (J.W.H.); (H.Y.); (S.H.B.); (J.W.J.); (J.Y.C.); (S.K.Y.); (D.Y.L.)
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Dong Yeup Lee
- The Catholic University Liver Research Centre, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (J.W.H.); (H.Y.); (S.H.B.); (J.W.J.); (J.Y.C.); (S.K.Y.); (D.Y.L.)
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Sung Hak Lee
- Department of Hospital Pathology, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea;
| | - Jae-Ho Yoon
- Department of Hematology, Catholic Hematology Hospital and Leukemia Research Institute, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (S.-S.P.); (S.P.); (S.-E.L.); (B.-S.C.); (K.-S.E.); (Y.-J.K.); (H.-J.K.); (C.-K.M.); (S.-G.C.); (J.W.L.); (S.L.)
| | - Pil Soo Sung
- The Catholic University Liver Research Centre, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (J.W.H.); (H.Y.); (S.H.B.); (J.W.J.); (J.Y.C.); (S.K.Y.); (D.Y.L.)
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| |
Collapse
|
5
|
Wang M, Li QJ, Zhao HY, Zhang JL. Tetramerization of pyruvate kinase M2 attenuates graft-versus-host disease by inhibition of Th1 and Th17 differentiation. Hum Cell 2024; 37:633-647. [PMID: 38416276 DOI: 10.1007/s13577-024-01033-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Accepted: 01/17/2024] [Indexed: 02/29/2024]
Abstract
Lethal graft-versus-host disease (GVHD) is the major complication of allogeneic hematopoietic stem-cell transplantation (Allo-HSCT). Pyruvate kinase M2 (PKM2) is essential for CD4+ T-cell differentiation. Using the well-characterized mouse models of Allo-HSCT, we explored the effects of TEPP-46-induced PKM2 tetramerization on GVHD and graft-versus-leukemia (GVL) activity. TEPP-46 administration significantly improved the survival rate of GVHD. The severity of GVHD and histopathological damage of GVHD-targeted organs were obviously alleviated by PKM2 tetramerization. Additionally, tetramerized PKM2 inhibited the activation of NF-κB pathway and decreased the inflammation level of GVHD mice. PKM2 tetramerization blocked Th1 and Th17 cell differentiation and secretion of pro-inflammatory cytokine (IFN-γ, TNF-α, and IL-17). Meanwhile, differentiation of Treg cells and IL-10 secretion were promoted by tetramerized PKM2. These findings demonstrated that PKM2 enhanced the augment of Th1 and Th17 cells to accelerate the progression of GVHD, and allosteric activation of PKM2 targeted Th1 and Th17 cells attenuated GVHD. Furthermore, we also confirmed that TEPP-46 administration did not compromise GVL activity and resulted in slightly improvement of leukemia-free survive. Thus, targeting Th1 and Th17 cell response with PKM2 allosteric activator may be a promising therapeutic strategy for GVHD prevention while preserving the GVL activity in patients receiving Allo-HSCT.
Collapse
Affiliation(s)
- Meng Wang
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, 1#, East Jianshe Road, Erqi District, Zhengzhou, Henan, China.
| | - Qiu-Jie Li
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, 1#, East Jianshe Road, Erqi District, Zhengzhou, Henan, China
| | - Hua-Yan Zhao
- Surgical Intensive Care Unit, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Jing-Lan Zhang
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, 1#, East Jianshe Road, Erqi District, Zhengzhou, Henan, China
| |
Collapse
|
6
|
Chen T, Li J, Wei X, Yao H, Zhu L, Liu J, Liu Y, Wang P, Feng Y, Gao S, Liu H, Wang L, Zhao L, Gao L, Zhang C, Gao L, Zhang X, Kong P. Efficiency and Toxicity of Imatinib Mesylate Combined with Atorvastatin Calcium in the Treatment of Steroid-Refractory Chronic Graft-versus-Host Disease: A Single-Center, Prospective, Single-Arm, Open-Label Study. Acta Haematol 2024; 147:499-510. [PMID: 38232716 DOI: 10.1159/000536174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Accepted: 12/26/2023] [Indexed: 01/19/2024]
Abstract
INTRODUCTION Steroid-refractory cGVHD (SR-cGVHD) presents new great challenges for treatment. We have reported that imatinib monotherapy was effective to SR-cGVHD, but the CR rate was not satisfactory and the benefit was not showed specific to some target organs, previously. Imatinib and statin drugs have been recognized to regulate T-cell function, statins also have been demonstrated endothelia protection, but whether this combination therapy was able to improve the efficacy remains unknown. Therefore, we designed this prospective, single-arm, open-label trial to investigate the efficacy of imatinib-based combination therapy in the treatment of SR-cGVHD for the first time. METHODS Sixty SR-cGVHD patients were entered into this trial to investigate the combination of imatinib mesylate and atorvastatin calcium for the treatment of SR-cGVHD. The primary endpoint included the overall response rate (ORR) after 6 months of combined treatment. The secondary endpoints included an evaluation of survival, changes in T-cell subsets, and adverse events. RESULTS At baseline, 45% (27/60) of patients had moderate cGVHD, and 55.0% (33/60) of patients had severe cGVHD. At the 6-month follow-up, a clinical response was achieved in 70.0% of patients, and a complete response (CR) was achieved in 26.7%. A total of 11.7% (7/60) of patients stopped immunosuppressive therapy at this point. After 6 months of treatment, the ORR rates of the liver, skin, eyes, and oral cavity were 80.6%, 78.1%, 61.5%, and 60.9%, respectively, with the liver also having the highest CR of 58.1%. The patients with moderate cGVHD had a better CR rate than those with severe cGVHD (55.6% vs. 3.0%, p < 0.0001). The overall survival in patients with ORR was improved (p = 0.0106). Lung involvement is an independent risk factor to affected ORR achievement (p = 0.021, HR = 0.335, 95% CI: 0.133-0.847), and the dosage of steroids was reduced in ORR patients. In clinical response patients, the ratio of CD8+ T cells (p = 0.0117) and Th17 cells (p = 0.0171) decreased, while the number of Treg cells (p = 0.0147) increased after 3 months. The most common adverse events were edema, nausea, and neutropenia, which were 13.3%, 11.7%, and 11.7%, respectively. CONCLUSION Combination treatment with imatinib mesylate and atorvastatin calcium was effective in treating SR-cGVHD and significantly decreased target organ injury, especially liver damage, indicating that T-cell regulatory function may play an important role in this process.
Collapse
Affiliation(s)
- Ting Chen
- Medical Center of Hematology, Xinqiao Hospital of Army Medical University, State Key Laboratory of Trauma, Burns and Combined Injury, Chongqing, China
| | - JiaLi Li
- Medical Center of Hematology, Xinqiao Hospital of Army Medical University, State Key Laboratory of Trauma, Burns and Combined Injury, Chongqing, China
| | - Xiao Wei
- Department of Endocrinology, The General Hospital of Western Theater Command PLA, Sichuan, China
| | - Han Yao
- Medical Center of Hematology, Xinqiao Hospital of Army Medical University, State Key Laboratory of Trauma, Burns and Combined Injury, Chongqing, China
| | - LiDan Zhu
- Medical Center of Hematology, Xinqiao Hospital of Army Medical University, State Key Laboratory of Trauma, Burns and Combined Injury, Chongqing, China
| | - Jia Liu
- Medical Center of Hematology, Xinqiao Hospital of Army Medical University, State Key Laboratory of Trauma, Burns and Combined Injury, Chongqing, China
| | - YuQing Liu
- Medical Center of Hematology, Xinqiao Hospital of Army Medical University, State Key Laboratory of Trauma, Burns and Combined Injury, Chongqing, China
| | - Ping Wang
- Medical Center of Hematology, Xinqiao Hospital of Army Medical University, State Key Laboratory of Trauma, Burns and Combined Injury, Chongqing, China
| | - YiMei Feng
- Medical Center of Hematology, Xinqiao Hospital of Army Medical University, State Key Laboratory of Trauma, Burns and Combined Injury, Chongqing, China
| | - ShiChun Gao
- Medical Center of Hematology, Xinqiao Hospital of Army Medical University, State Key Laboratory of Trauma, Burns and Combined Injury, Chongqing, China
| | - HuanFeng Liu
- Medical Center of Hematology, Xinqiao Hospital of Army Medical University, State Key Laboratory of Trauma, Burns and Combined Injury, Chongqing, China
| | - Lu Wang
- Medical Center of Hematology, Xinqiao Hospital of Army Medical University, State Key Laboratory of Trauma, Burns and Combined Injury, Chongqing, China
| | - Lu Zhao
- Medical Center of Hematology, Xinqiao Hospital of Army Medical University, State Key Laboratory of Trauma, Burns and Combined Injury, Chongqing, China
| | - Li Gao
- Medical Center of Hematology, Xinqiao Hospital of Army Medical University, State Key Laboratory of Trauma, Burns and Combined Injury, Chongqing, China
| | - Cheng Zhang
- Medical Center of Hematology, Xinqiao Hospital of Army Medical University, State Key Laboratory of Trauma, Burns and Combined Injury, Chongqing, China
| | - Lei Gao
- Medical Center of Hematology, Xinqiao Hospital of Army Medical University, State Key Laboratory of Trauma, Burns and Combined Injury, Chongqing, China
| | - Xi Zhang
- Medical Center of Hematology, Xinqiao Hospital of Army Medical University, State Key Laboratory of Trauma, Burns and Combined Injury, Chongqing, China
| | - PeiYan Kong
- Medical Center of Hematology, Xinqiao Hospital of Army Medical University, State Key Laboratory of Trauma, Burns and Combined Injury, Chongqing, China
| |
Collapse
|
7
|
Wan Y, Mao M, Li M, Liu J, Tong X, Wang J, Li J, Yin S, Wu C. Serum CXCL16: A new predictor of liver inflammation in patients with chronic hepatitis B. J Viral Hepat 2024; 31:107-119. [PMID: 38146125 DOI: 10.1111/jvh.13905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 10/14/2023] [Accepted: 11/30/2023] [Indexed: 12/27/2023]
Abstract
The prompt initiation of antiviral therapy is essential in patients with chronic hepatitis B (CHB), especially when severe liver inflammation is detected. However, transcutaneous liver puncture, the gold standard for assessing liver inflammation, is invasive and its widespread application is limited. Therefore, there is an urgent need for more non-invasive markers to predict liver inflammation. In our retrospective cross-sectional study, which included 120 CHB patients and 31 healthy subjects, we observed a significant increase in serum chemokine C-X-C-motif ligand 16 (CXCL16) in CHB patients compared to healthy controls (p < .001). Notably, patients with severe inflammation (Scheuer's grade G ≥ 3, n = 26) exhibited a substantial increase in serum CXCL16 compared to those with non-severe inflammation (Scheuer's grade G < 3, n = 96) [(median, IQR), 0.42 (0.24-0.71) ng/mL vs. 1.01 (0.25-2.09) ng/mL, p < .001]. Furthermore, we developed a predictive model that combined CXCL16 with platelet count (PLT), alanine aminotransferase (ALT) and albumin (ALB) to accurately predict liver inflammation in CHB patients. This model was more effective than ALT alone in predicting liver inflammation (AUC, 0.92 vs. 0.81, p = .015). Additionally, using an HBV-transduced mouse model, we demonstrated that blocking CXCL16 led to a reduction in liver inflammation and impaired infiltration and function of natural killer T (NKT) and natural killer (NK) cells. These findings suggest that CXCL16 is a promising non-invasive biomarker of liver inflammation in CHB patients and may play a role in inducing liver inflammation via a NKT and NK cell pathway.
Collapse
Affiliation(s)
- Yawen Wan
- Department of Infectious Diseases, Nanjing Drum Tower Hospital Clinical College of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Minxin Mao
- Department of Infectious Diseases, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Ming Li
- Department of Infectious Diseases, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Jiacheng Liu
- Department of Infectious Diseases, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Xin Tong
- Department of Infectious Diseases, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Jian Wang
- Department of Infectious Diseases, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Jie Li
- Department of Infectious Diseases, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Shengxia Yin
- Department of Infectious Diseases, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
- Institute of Viruses and Infectious Diseases, Nanjing University, Nanjing, Jiangsu, China
| | - Chao Wu
- Department of Infectious Diseases, Nanjing Drum Tower Hospital Clinical College of Xuzhou Medical University, Xuzhou, Jiangsu, China
- Department of Infectious Diseases, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
- Department of Infectious Diseases, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
- Institute of Viruses and Infectious Diseases, Nanjing University, Nanjing, Jiangsu, China
| |
Collapse
|
8
|
Christensen PKF, Hansen AK, Skov S, Engkilde K, Larsen J, Høyer-Hansen MH, Koch J. Sustaining the T-cell activity in xenografted psoriasis skin. PLoS One 2023; 18:e0278390. [PMID: 36649237 PMCID: PMC9844869 DOI: 10.1371/journal.pone.0278390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Accepted: 11/15/2022] [Indexed: 01/18/2023] Open
Abstract
Xenografting of psoriasis skin onto immune deficient mice has been widely used to obtain proof-of-principle of new drug candidates. However, the lack of human T-cell activity in the grafts limits the use of the model. Here, we show that xenografting of lesional skin from psoriasis patients onto human IL-2 NOG mice results in increased numbers of human CD3+ cells in the grafts, axillary lymph nodes and blood from human IL-2 NOG mice compared to C.B-17 scid and NOG mice. In addition, disease relevant human cytokine levels were higher in graft lysates and serum from human IL-2 NOG mice. However, the epidermis was lacking and no efficacy of ustekinumab, a human anti-P40 antibody targeting both IL-12 and IL-23, was shown. Thus, despite the sustained T-cell activity, the model needs further investigations and validation to capture more aspects of psoriasis.
Collapse
Affiliation(s)
- Pernille Kristine Fisker Christensen
- LEO Pharma A/S, Ballerup, Denmark
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg C, Denmark
- * E-mail:
| | - Axel Kornerup Hansen
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg C, Denmark
| | - Søren Skov
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg C, Denmark
| | | | | | | | | |
Collapse
|
9
|
Qiu Y, Hu B, Peng RM, Huang JF, Hong J. Tear Cytokines as Biomarkers for Acute Ocular Graft-Versus-Host Disease. Cornea 2022; 41:1405-1411. [PMID: 35184125 DOI: 10.1097/ico.0000000000002959] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Accepted: 10/25/2021] [Indexed: 11/26/2022]
Abstract
PURPOSE The purpose of this study was to analyze tear cytokine and complement levels in patients diagnosed with acute ocular graft-versus-host disease (oGVHD) and examine the consistency of these levels with the severity of clinical manifestations. METHODS Ten patients with acute oGVHD (20 eyes) were enrolled for the assessment of tear cytokine levels and ocular surface parameters, and 18 healthy people (36 eyes) were selected as the control group. The tear cytokine and complement levels were measured using microsphere-based immunoassay analysis. RESULTS The main clinical manifestations of acute oGVHD include eye redness, a large amount of purulent exudate, eye pain, and even false membranes. The levels of intercellular cell adhesion molecule-1, interleukin 6 (IL-6), interleukin 1 beta (IL-1β), interleukin 8, epidermal growth factor (EGF), interleukin 7 (IL-7), B-cell activating factor, granulocyte-macrophage colony-stimulating factor (GM-CSF), and complement in patients with acute oGVHD showed significant differences compared with those in normal people. Furthermore, the levels of IL-6, IL-1β, EGF, GM-CSF, IL-7, and C3a showed a stronger correlation with ocular surface parameters. CONCLUSIONS Our study was the first to enroll patients with acute oGVHD to assess tear cytokine levels as a method contributing to the diagnosis of acute oGVHD. In addition, it has been demonstrated that certain tear cytokines, including intercellular cell adhesion molecule-1, IL-6, IL-1β, interleukin 8, B-cell activating factor, GM-CSF, IL-7, EGF, and complement, may be new diagnostic biomarkers for acute oGVHD.
Collapse
Affiliation(s)
- Yuan Qiu
- Department of Ophthalmology, Peking University Third Hospital, China; and
| | - Bohao Hu
- Department of Ophthalmology, Peking University Third Hospital, China; and
| | - Rong-Mei Peng
- Department of Ophthalmology, Peking University Third Hospital, China; and
| | | | - Jing Hong
- Department of Ophthalmology, Peking University Third Hospital, China; and
| |
Collapse
|
10
|
Belpaire A, van Geel N, Speeckaert R. From IL-17 to IFN-γ in inflammatory skin disorders: Is transdifferentiation a potential treatment target? Front Immunol 2022; 13:932265. [PMID: 35967358 PMCID: PMC9367984 DOI: 10.3389/fimmu.2022.932265] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 06/29/2022] [Indexed: 11/13/2022] Open
Abstract
The targeted inhibition of effector cytokines such as interleukin 17 (IL-17) in psoriasis and IL-13 in atopic dermatitis offers impressive efficacy with a favorable side effect profile. In contrast, the downregulation of interferon gamma (IFN-γ) in T helper (Th) 1-dominant skin disorders may lead to more adverse events, given the crucial role of IFN-γ in antiviral and antitumoral immunity. Modulating Th17 and Th2 cell differentiation is performed by blocking IL-23 and IL-4, respectively, whereas anti-IL-12 antibodies are only moderately effective in downregulating Th1 lymphocyte differentiation. Therefore, a targeted approach of IFN-γ-driven disorders remains challenging. Recent literature suggests that certain pathogenic Th17 cell subsets with Th1 characteristics, such as CD4+CD161+CCR6+CXCR3+IL-17+IFN-y+ (Th17.1) and CD4+CD161+CCR6+CXCR3+IL-17-IFN-y+ (exTh17), are important contributors in Th1-mediated autoimmunity. Differentiation to a Th17.1 or exTh17 profile results in the upregulation of IFN-y. Remarkably, these pathogenic Th17 cell subsets are resistant to glucocorticoid therapy and the dampening effect of regulatory T cells (Treg). The identification of Th17.1/exTh17 cells in auto-immune disorders may explain the frequent treatment failure of conventional immunosuppressants. In this review, we summarize the current evidence regarding the cellular plasticity of Th17 cells in inflammatory skin disorders. A deeper understanding of this phenomenon may lead to better insights into the pathogenesis of various skin diseases and the discovery of a potential new treatment target.
Collapse
|
11
|
Thangavelu G, Zaiken MC, Mohamed FA, Flynn R, Du J, Rhee SY, Riddle MJ, Aguilar EG, Panoskaltsis-Mortari A, Sanders ME, Blazar BR. Targeting the Retinoid X Receptor Pathway Prevents and Ameliorates Murine Chronic Graft-Versus-Host Disease. Front Immunol 2022; 13:765319. [PMID: 35359939 PMCID: PMC8963714 DOI: 10.3389/fimmu.2022.765319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Accepted: 01/26/2022] [Indexed: 02/03/2023] Open
Abstract
Most allogeneic hematopoietic stem cell transplant (allo-HSCT) recipients receive peripheral blood stem cell grafts resulting in a 30%-70% incidence of chronic graft-versus-host disease (cGVHD), a major cause of mortality and morbidity in long-term survivors. While systemic steroids remain the standard of care for first-line therapy, patients may require long-term administration, and those with steroid-resistant or refractory cGVHD have a worse prognosis. Although durable and deep responses with second-line therapies can be achieved in some patients, there remains an urgent need for new therapies. In this study, we evaluated the efficacy of IRX4204, a novel agonist that activates RXRs and is in clinical trials for cancer treatment to prevent and treat cGVHD in two complementary murine models. In a major histocompatibility complex mismatched, non-sclerodermatous multiorgan system model with bronchiolitis obliterans, IRX4204 prevented and reversed cGVHD including associated pulmonary dysfunction with restoration of germinal center T-follicular helper: T-follicular regulatory cell balance. In a minor histocompatibility antigen disparate sclerodermatous model, IRX4204 treatment significantly prevented and ameliorated skin cGVHD by reducing Th1 and Th17 differentiation due to anti-inflammatory properties. Together, these results indicate that IRX4204 is a promising therapeutic option to treat cGVHD with bronchiolitis obliterans or sclerodermatous manifestations.
Collapse
Affiliation(s)
- Govindarajan Thangavelu
- Department of Pediatrics, Division of Blood & Marrow Transplantation & Cellular Therapy, University of Minnesota, Minneapolis, MN, United States
| | - Michael C. Zaiken
- Department of Pediatrics, Division of Blood & Marrow Transplantation & Cellular Therapy, University of Minnesota, Minneapolis, MN, United States
| | - Fathima A. Mohamed
- Department of Pediatrics, Division of Blood & Marrow Transplantation & Cellular Therapy, University of Minnesota, Minneapolis, MN, United States
| | - Ryan Flynn
- Department of Pediatrics, Division of Blood & Marrow Transplantation & Cellular Therapy, University of Minnesota, Minneapolis, MN, United States
| | - Jing Du
- Department of Pediatrics, Division of Blood & Marrow Transplantation & Cellular Therapy, University of Minnesota, Minneapolis, MN, United States
| | - Stephanie Y. Rhee
- Department of Pediatrics, Division of Blood & Marrow Transplantation & Cellular Therapy, University of Minnesota, Minneapolis, MN, United States
| | - Megan J. Riddle
- Department of Pediatrics, Division of Blood & Marrow Transplantation & Cellular Therapy, University of Minnesota, Minneapolis, MN, United States
| | - Ethan G. Aguilar
- Department of Pediatrics, Division of Blood & Marrow Transplantation & Cellular Therapy, University of Minnesota, Minneapolis, MN, United States
| | - Angela Panoskaltsis-Mortari
- Department of Pediatrics, Division of Blood & Marrow Transplantation & Cellular Therapy, University of Minnesota, Minneapolis, MN, United States
| | | | - Bruce R. Blazar
- Department of Pediatrics, Division of Blood & Marrow Transplantation & Cellular Therapy, University of Minnesota, Minneapolis, MN, United States
| |
Collapse
|
12
|
Choi HJ, Tang CHA, Tian L, Wu Y, Sofi MH, Ticer T, Schutt SD, Hu CCA, Yu XZ. XBP-1s Promotes B Cell Pathogenicity in Chronic GVHD by Restraining the Activity of Regulated IRE-1α-Dependent Decay. Front Immunol 2021; 12:705484. [PMID: 34659198 PMCID: PMC8517405 DOI: 10.3389/fimmu.2021.705484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 09/15/2021] [Indexed: 11/20/2022] Open
Abstract
Allogeneic hematopoietic cell transplantation (allo-HCT) is an effective therapeutic procedure to treat hematological malignancies. However, the benefit of allo-HCT is limited by a major complication, chronic graft-versus-host disease (cGVHD). Since transmembrane and secretory proteins are generated and modified in the endoplasmic reticulum (ER), the ER stress response is of great importance to secretory cells including B cells. By using conditional knock-out (KO) of XBP-1, IRE-1α or both specifically on B cells, we demonstrated that the IRE-1α/XBP-1 pathway, one of the major ER stress response mediators, plays a critical role in B cell pathogenicity on the induction of cGVHD in murine models of allo-HCT. Endoribonuclease activity of IRE-1α activates XBP-1 signaling by converting unspliced XBP-1 (XBP-1u) mRNA into spliced XBP-1 (XBP-1s) mRNA but also cleaves other ER-associated mRNAs through regulated IRE-1α-dependent decay (RIDD). Further, ablation of XBP-1s production leads to unleashed activation of RIDD. Therefore, we hypothesized that RIDD plays an important role in B cells during cGVHD development. In this study, we found that the reduced pathogenicity of XBP-1 deficient B cells in cGVHD was reversed by RIDD restriction in IRE-1α kinase domain KO mice. Restraining RIDD activity per se in B cells resulted in an increased severity of cGVHD. Besides, inhibition of RIDD activity compromised B cell differentiation and led to dysregulated expression of MHC II and costimulatory molecules such as CD86, CD40, and ICOSL in B cells. Furthermore, restraining the RIDD activity without affecting XBP-1 splicing increased B cell ability to induce cGVHD after allo-HCT. These results suggest that RIDD is an important mediator for reducing cGVHD pathogenesis through targeting XBP-1s.
Collapse
Affiliation(s)
- Hee-Jin Choi
- Microbiology & Immunology, Medical University of South Carolina, Charleston, SC, United States
| | - Chih-Hang Anthony Tang
- Center for Translational Research in Hematologic Malignancies, Houston Methodist Cancer Center, Houston Methodist Research Institute, Houston, TX, United States
| | - Linlu Tian
- Microbiology & Immunology, Medical University of South Carolina, Charleston, SC, United States
| | - Yongxia Wu
- Microbiology & Immunology, Medical University of South Carolina, Charleston, SC, United States
| | - M Hanief Sofi
- Microbiology & Immunology, Medical University of South Carolina, Charleston, SC, United States
| | - Taylor Ticer
- Microbiology & Immunology, Medical University of South Carolina, Charleston, SC, United States
| | - Steven D Schutt
- Microbiology & Immunology, Medical University of South Carolina, Charleston, SC, United States
| | - Chih-Chi Andrew Hu
- Center for Translational Research in Hematologic Malignancies, Houston Methodist Cancer Center, Houston Methodist Research Institute, Houston, TX, United States
| | - Xue-Zhong Yu
- Microbiology & Immunology, Medical University of South Carolina, Charleston, SC, United States.,Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, United States
| |
Collapse
|
13
|
Molecular determinants of peaceful coexistence versus invasiveness of non-Typhoidal Salmonella: Implications in long-term side-effects. Mol Aspects Med 2021; 81:100997. [PMID: 34311996 DOI: 10.1016/j.mam.2021.100997] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Revised: 07/02/2021] [Accepted: 07/16/2021] [Indexed: 01/28/2023]
Abstract
The genus Salmonella represents a wide range of strains including Typhoidal and Non-Typhoidal Salmonella (NTS) isolates that exhibit illnesses of varied pathophysiologies. The more frequent NTS ensues a self-limiting enterocolitis with rare occasions of bacteremia or systemic infections. These self-limiting Salmonella strains are capable of subverting and dampening the host immune system to achieve a more prolonged survival inside the host system thus leading to chronic manifestations. Notably, emergence of new invasive NTS isolates known as invasive Non-Typhoidal Salmonella (iNTS) have worsened the disease burden significantly in some parts of the world. NTS strains adapt to attain persister phenotype intracellularly and cause relapsing infections. These chronic infections, in susceptible hosts, are also capable of causing diseases like IBS, IBD, reactive arthritis, gallbladder cancer and colorectal cancer. The present understanding of molecular mechanism of how these chronic infections are manifested is quite limited. The current work is an effort to review the prevailing knowledge emanating from a large volume of research focusing on various forms of NTS infections including those that cause localized, systemic and persistent disease. The review will further dwell into the understanding of how this pathogen contributes to the associated long term sequelae.
Collapse
|
14
|
Dai H, Rachakonda SP, Penack O, Blau IW, Blau O, Radujkovic A, Müller-Tidow C, Dreger P, Kumar R, Luft T. Polymorphisms in CXCR3 ligands predict early CXCL9 recovery and severe chronic GVHD. Blood Cancer J 2021; 11:42. [PMID: 33640906 PMCID: PMC7914250 DOI: 10.1038/s41408-021-00434-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 12/17/2020] [Accepted: 02/03/2021] [Indexed: 12/16/2022] Open
Abstract
Chronic graft-versus-host disease (cGVHD) is a major cause of mortality and morbidity after allogeneic stem cell transplantation (alloSCT). The individual risk of severe cGVHD remains difficult to predict and may involve CXCR3 ligands. This study investigated the role of single-nucleotide polymorphisms (SNPs) of CXCL4, CXCL9, CXCL10, and CXCL11, and their day +28 serum levels, in cGVHD pathogenesis. Eighteen CXCR3 and CXCL4, CXCL9-11 SNPs as well as peri-transplant CXCL9-11 serum levels were analyzed in 688 patients without (training cohort; n = 287) or with statin-based endothelial protection cohort (n = 401). Clinical outcomes were correlated to serum levels and SNP status. Significant polymorphisms were further analyzed by luciferase reporter assays. Findings were validated in an independent cohort (n = 202). A combined genetic risk comprising four CXCR3 ligand SNPs was significantly associated with increased risk of severe cGVHD in both training cohort (hazard ratio (HR) 2.48, 95% confidence interval (CI) 1.33-4.64, P = 0.004) and validation cohort (HR 2.95, 95% CI 1.56-5.58, P = 0.001). In reporter assays, significantly reduced suppressive effects of calcineurin inhibitors in constructs with variant alleles of rs884304 (P < 0.001) and rs884004 (P < 0.001) were observed. CXCL9 serum levels at day +28 after alloSCT correlated with both genetic risk and risk of severe cGVHD (HR 1.38, 95% CI 1.10-1.73, P = 0.006). This study identifies patients with high genetic risk to develop severe cGVHD.
Collapse
Affiliation(s)
- Hao Dai
- Department of Epidemiology, German Cancer Research Centre (DKFZ), Heidelberg, Germany
| | | | - Olaf Penack
- Division of Hematology, Oncology and Tumorimmunology, Charité University Medicine Berlin, Berlin, Germany
| | - Igor W Blau
- Division of Hematology, Oncology and Tumorimmunology, Charité University Medicine Berlin, Berlin, Germany
| | - Olga Blau
- Division of Hematology, Oncology and Tumorimmunology, Charité University Medicine Berlin, Berlin, Germany
| | | | | | - Peter Dreger
- Department of Medicine V, University Hospital Heidelberg, Heidelberg, Germany
| | - Rajiv Kumar
- Department of Epidemiology, German Cancer Research Centre (DKFZ), Heidelberg, Germany
| | - Thomas Luft
- Department of Medicine V, University Hospital Heidelberg, Heidelberg, Germany.
| |
Collapse
|
15
|
Wan L, Jin Z, Hu B, Lv K, Lei L, Liu Y, Song Y, Zhu Y, Gong H, Xu M, Du Y, Xu Y, Liu H, Wu D, Liu Y. IL-Y Aggravates Murine Chronic Graft- Versus-Host Disease by Enhancing T and B Cell Responses. Front Immunol 2020; 11:559740. [PMID: 33329519 PMCID: PMC7719702 DOI: 10.3389/fimmu.2020.559740] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 10/20/2020] [Indexed: 11/16/2022] Open
Abstract
IL-Y, a synthetic member of IL-12 cytokine family, was found to exert potent immunosuppressive effects by inhibiting the differentiation and activation of Th1 and Th17 cells. However, the role of IL-Y in the development of chronic graft-versus-host disease (cGVHD) remains unknown. Here, using murine models of scleroderma-like and lupus-like cGVHD, we examined the function of IL-Y in the pathogenesis of cGVHD by hydrodynamically injecting minicircle-IL-Y expressing plasmids (MC IL-Y). In contrast with the reported immune suppressive function of IL-Y, administration of MC IL-Y enhanced cGVHD severity reflected by deteriorated multi-organ pathologic damages. In lupus-like cGVHD model, urine protein and the serum anti-dsDNA antibody (IgG) were significantly upregulated by IL-Y treatment. Further study demonstrated that IL-Y impacts both donor T and B cell response. In T cells, IL-Y inhibited the generation of CD4+Foxp3+ regulator T (Treg) cells during the development of cGVHD. IL-Y may also increase the infiltration of pathogenic TNF-α producing CD4+ and CD8+ T cells through IL-27Rα in recipient spleens, as this effect was diminished in IL-27Rα deficient T cells. Moreover, IL-Y enhanced the differentiation of ICOS+ T follicular helper (Tfh) cells. In B cells, the percentage of germinal center (GC) B cells in recipient spleens was significantly upregulated by MC IL-Y plasmid administration. The levels of co-stimulatory molecules, MHC-II and CD86, on B cells were also enhanced by IL-Y expression. Taken together, our data indicated that IL-Y promoted the process of cGVHD by activating pathogenic T and B cells.
Collapse
Affiliation(s)
- Li Wan
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Institute of Blood and Marrow Transplantation, Suzhou, China.,Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Ziqi Jin
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Institute of Blood and Marrow Transplantation, Suzhou, China.,Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Bo Hu
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Institute of Blood and Marrow Transplantation, Suzhou, China.,Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Kangkang Lv
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Institute of Blood and Marrow Transplantation, Suzhou, China.,Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Lei Lei
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Institute of Blood and Marrow Transplantation, Suzhou, China.,Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Yonghao Liu
- Immunology Programme, Life Sciences Institute and Department of Microbiology and Immunology, Yoo Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Yuan Song
- Immunology Programme, Life Sciences Institute and Department of Microbiology and Immunology, Yoo Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Ying Zhu
- Immunology Programme, Life Sciences Institute and Department of Microbiology and Immunology, Yoo Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Huanle Gong
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Institute of Blood and Marrow Transplantation, Suzhou, China.,Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Mimi Xu
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Institute of Blood and Marrow Transplantation, Suzhou, China.,Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Yuanyuan Du
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Institute of Blood and Marrow Transplantation, Suzhou, China.,Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Yang Xu
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Institute of Blood and Marrow Transplantation, Suzhou, China.,Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Haiyan Liu
- Immunology Programme, Life Sciences Institute and Department of Microbiology and Immunology, Yoo Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Depei Wu
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Institute of Blood and Marrow Transplantation, Suzhou, China.,Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Yuejun Liu
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Institute of Blood and Marrow Transplantation, Suzhou, China.,Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| |
Collapse
|
16
|
Tang D, Liu S, Sun H, Qin X, Zhou N, Zheng W, Zhang M, Zhou H, Tuersunayi A, Duan C, Chen J. All-trans-retinoic acid shifts Th1 towards Th2 cell differentiation by targeting NFAT1 signalling to ameliorate immune-mediated aplastic anaemia. Br J Haematol 2020; 191:906-919. [PMID: 32729137 DOI: 10.1111/bjh.16871] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2020] [Accepted: 05/20/2020] [Indexed: 12/12/2022]
Abstract
Severe acquired aplastic anaemia (AA) is a serious disease characterised by autoreactive T cells attacking haematopoietic stem cells, leading to marrow hypoplasia and pancytopenia. Immunosuppressive therapy combined with antithymocyte globulin and ciclosporin can rescue most patients with AA. However, the relapse after ciclosporin withdrawal and the severe side effects of long-term ciclosporin administration remain unresolved. As such, new strategies should be developed to supplement current therapeutics and treat AA. In this study, the possibility of all-trans-retinoic acid (ATRA) as an alternative AA treatment was tested by using an immune-mediated mouse model of AA. Results revealed that ATRA inhibited T-cell proliferation, activation and effector function. It also restrained the Fas/Fasl pathway, shifted Th1 towards Th2 cell development, rebalanced T-cell subsets at a relatively high level and corrected the Th1/Th2 ratio by targeting NFAT1 signalling. In addition, ATRA inhibited Th17 cell differentiation and promoted regulatory T-cell development. Therefore, ATRA was an effective agent to improve AA treatment outcomes.
Collapse
Affiliation(s)
- Dabin Tang
- Shanghai Children's Medical Center, Key Laboratory of Pediatric Hematology and Oncology Ministry of Health and Pediatric Translational Medicine Institute, Department of Hematology and Oncology, Shanghai Collaborative Innovation Center for Translational Medicine and Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai, China
| | - Shengli Liu
- Shanghai Children's Medical Center, Key Laboratory of Pediatric Hematology and Oncology Ministry of Health and Pediatric Translational Medicine Institute, Department of Hematology and Oncology, Shanghai Collaborative Innovation Center for Translational Medicine and Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai, China
| | - Huiying Sun
- Shanghai Children's Medical Center, Key Laboratory of Pediatric Hematology and Oncology Ministry of Health and Pediatric Translational Medicine Institute, Department of Hematology and Oncology, Shanghai Collaborative Innovation Center for Translational Medicine and Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai, China
| | - Xia Qin
- Shanghai Children's Medical Center, Key Laboratory of Pediatric Hematology and Oncology Ministry of Health and Pediatric Translational Medicine Institute, Department of Hematology and Oncology, Shanghai Collaborative Innovation Center for Translational Medicine and Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai, China
| | - Neng Zhou
- Shanghai Children's Medical Center, Key Laboratory of Pediatric Hematology and Oncology Ministry of Health and Pediatric Translational Medicine Institute, Department of Hematology and Oncology, Shanghai Collaborative Innovation Center for Translational Medicine and Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai, China
| | - Weiwei Zheng
- Shanghai Children's Medical Center, Key Laboratory of Pediatric Hematology and Oncology Ministry of Health and Pediatric Translational Medicine Institute, Department of Hematology and Oncology, Shanghai Collaborative Innovation Center for Translational Medicine and Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai, China
| | - Mengyi Zhang
- Shanghai Children's Medical Center, Key Laboratory of Pediatric Hematology and Oncology Ministry of Health and Pediatric Translational Medicine Institute, Department of Hematology and Oncology, Shanghai Collaborative Innovation Center for Translational Medicine and Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai, China
| | - Hang Zhou
- Shanghai Children's Medical Center, Key Laboratory of Pediatric Hematology and Oncology Ministry of Health and Pediatric Translational Medicine Institute, Department of Hematology and Oncology, Shanghai Collaborative Innovation Center for Translational Medicine and Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai, China
| | - Abudureheman Tuersunayi
- Shanghai Children's Medical Center, Key Laboratory of Pediatric Hematology and Oncology Ministry of Health and Pediatric Translational Medicine Institute, Department of Hematology and Oncology, Shanghai Collaborative Innovation Center for Translational Medicine and Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai, China
| | - Caiwen Duan
- Shanghai Children's Medical Center, Key Laboratory of Pediatric Hematology and Oncology Ministry of Health and Pediatric Translational Medicine Institute, Department of Hematology and Oncology, Shanghai Collaborative Innovation Center for Translational Medicine and Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai, China
| | - Jing Chen
- Shanghai Children's Medical Center, Key Laboratory of Pediatric Hematology and Oncology Ministry of Health and Pediatric Translational Medicine Institute, Department of Hematology and Oncology, Shanghai Collaborative Innovation Center for Translational Medicine and Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai, China
| |
Collapse
|
17
|
Hu B, Qiu Y, Hong J. Tear cytokine levels in the diagnosis and severity assessment of ocular chronic graft-versus-host disease(GVHD). Ocul Surf 2020; 18:298-304. [DOI: 10.1016/j.jtos.2019.12.005] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 11/26/2019] [Accepted: 12/12/2019] [Indexed: 12/20/2022]
|
18
|
Berberine combined with cyclosporine A alleviates acute graft-versus-host disease in murine models. Int Immunopharmacol 2020; 81:106205. [DOI: 10.1016/j.intimp.2020.106205] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 12/25/2019] [Accepted: 01/07/2020] [Indexed: 12/22/2022]
|
19
|
Thangavelu G, Du J, Paz KG, Loschi M, Zaiken MC, Flynn R, Taylor PA, Kirchmeier AK, Panoskaltsis-Mortari A, Luznik L, MacDonald KP, Hill GR, Maillard I, Munn DH, Serody JS, Murphy WJ, Miklos D, Cutler CS, Koreth J, Antin JH, Soiffer RJ, Ritz J, Dahlberg C, Miller AT, Blazar BR. Inhibition of inositol kinase B controls acute and chronic graft-versus-host disease. Blood 2020; 135:28-40. [PMID: 31697815 PMCID: PMC6940197 DOI: 10.1182/blood.2019000032] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Accepted: 10/03/2019] [Indexed: 02/06/2023] Open
Abstract
T-cell activation releases inositol 1,4,5-trisphosphate (IP3), inducing cytoplasmic calcium (Ca2+) influx. In turn, inositol 1,4,5-trisphosphate 3-kinase B (Itpkb) phosphorylates IP3 to negatively regulate and thereby tightly control Ca2+ fluxes that are essential for mature T-cell activation and differentiation and protection from cell death. Itpkb pathway inhibition increases intracellular Ca2+, induces apoptosis of activated T cells, and can control T-cell-mediated autoimmunity. In this study, we employed genetic and pharmacological approaches to inhibit Itpkb signaling as a means of controlling graft-versus-host disease (GVHD). Murine-induced, Itpkb-deleted (Itpkb-/-) T cells attenuated acute GVHD in 2 models without eliminating A20-luciferase B-cell lymphoma graft-versus-leukemia (GVL). A highly potent, selective inhibitor, GNF362, ameliorated acute GVHD without impairing GVL against 2 acute myeloid leukemia lines (MLL-AF9-eGFP and C1498-luciferase). Compared with FK506, GNF362 more selectively deleted donor alloreactive vs nominal antigen-responsive T cells. Consistent with these data and as compared with FK506, GNF362 had favorable acute GVHD and GVL properties against MLL-AF9-eGFP cells. In chronic GVHD preclinical models that have a pathophysiology distinct from acute GVHD, Itpkb-/- donor T cells reduced active chronic GVHD in a multiorgan system model of bronchiolitis obliterans (BO), driven by germinal center reactions and resulting in target organ fibrosis. GNF362 treatment reduced active chronic GVHD in both BO and scleroderma models. Thus, intact Itpkb signaling is essential to drive acute GVHD pathogenesis and sustain active chronic GVHD, pointing toward a novel clinical application to prevent acute or treat chronic GVHD.
Collapse
Affiliation(s)
- Govindarajan Thangavelu
- Division of Blood and Marrow Transplantation, Department of Pediatrics, Masonic Cancer Center, University of Minnesota, Minneapolis, MN
| | - Jing Du
- Division of Blood and Marrow Transplantation, Department of Pediatrics, Masonic Cancer Center, University of Minnesota, Minneapolis, MN
| | - Katelyn G Paz
- Division of Blood and Marrow Transplantation, Department of Pediatrics, Masonic Cancer Center, University of Minnesota, Minneapolis, MN
| | - Michael Loschi
- Division of Blood and Marrow Transplantation, Department of Pediatrics, Masonic Cancer Center, University of Minnesota, Minneapolis, MN
| | - Michael C Zaiken
- Division of Blood and Marrow Transplantation, Department of Pediatrics, Masonic Cancer Center, University of Minnesota, Minneapolis, MN
| | - Ryan Flynn
- Division of Blood and Marrow Transplantation, Department of Pediatrics, Masonic Cancer Center, University of Minnesota, Minneapolis, MN
| | - Patricia A Taylor
- Division of Blood and Marrow Transplantation, Department of Pediatrics, Masonic Cancer Center, University of Minnesota, Minneapolis, MN
| | - Andrew Kemal Kirchmeier
- Division of Blood and Marrow Transplantation, Department of Pediatrics, Masonic Cancer Center, University of Minnesota, Minneapolis, MN
| | - Angela Panoskaltsis-Mortari
- Division of Blood and Marrow Transplantation, Department of Pediatrics, Masonic Cancer Center, University of Minnesota, Minneapolis, MN
| | - Leo Luznik
- Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD
| | - Kelli P MacDonald
- Department of Immunology, Queensland Institute of Medical Research (QIMR) Berghofer Medical Research Institute and School of Medicine, University of Queensland, Brisbane, QLD, Australia
| | - Geoffrey R Hill
- Department of Immunology, Queensland Institute of Medical Research (QIMR) Berghofer Medical Research Institute and School of Medicine, University of Queensland, Brisbane, QLD, Australia
| | - Ivan Maillard
- Division of Hematology-Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - David H Munn
- Georgia Cancer Center and
- Department of Pediatrics, Medical College of Georgia, Augusta University, Augusta, GA
| | - Jonathan S Serody
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC
| | - William J Murphy
- Department of Dermatology and
- Department of Internal Medicine, Laboratory of Cancer Immunology, University of California Davis Medical Center, Sacramento, CA
| | - David Miklos
- Division of Blood and Marrow Transplantation, Stanford University School of Medicine, Stanford, CA
| | - Corey S Cutler
- Stem Cell/Bone Marrow Transplantation Program, Division of Hematologic Malignancy, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA; and
| | - John Koreth
- Stem Cell/Bone Marrow Transplantation Program, Division of Hematologic Malignancy, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA; and
| | - Joseph H Antin
- Stem Cell/Bone Marrow Transplantation Program, Division of Hematologic Malignancy, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA; and
| | - Robert J Soiffer
- Stem Cell/Bone Marrow Transplantation Program, Division of Hematologic Malignancy, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA; and
| | - Jerome Ritz
- Stem Cell/Bone Marrow Transplantation Program, Division of Hematologic Malignancy, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA; and
| | - Carol Dahlberg
- The Genomics Institute, Novartis Research Foundation (GNF), San Diego, CA
| | - Andrew T Miller
- The Genomics Institute, Novartis Research Foundation (GNF), San Diego, CA
| | - Bruce R Blazar
- Division of Blood and Marrow Transplantation, Department of Pediatrics, Masonic Cancer Center, University of Minnesota, Minneapolis, MN
| |
Collapse
|
20
|
Chen T, Li XP, Zhang C, Kong PY, Gao QG, Tang L, Wang R, Yang SJ, Gao L, Liu Y, Gao L, Feng YM, Rao J, Peng XG, Zhang X. [The clinical observation of serum specific biomarkers in patients with chronic graft-versus-host disease]. ZHONGHUA XUE YE XUE ZA ZHI = ZHONGHUA XUEYEXUE ZAZHI 2019; 40:948-952. [PMID: 31856446 PMCID: PMC7342379 DOI: 10.3760/cma.j.issn.0253-2727.2019.11.012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
目的 研究异基因造血干细胞移植后患者血清生物标志物表达水平对慢性移植物抗宿主病(cGVHD)早期诊断的价值。 方法 采用液相悬浮芯片法检测接受异基因造血干细胞移植后发生和未发生cGVHD患者5种血清蛋白标志物(IL-1b、IL-16、CXCL9、CCL19、CCL17)表达水平。 结果 相较于未发生cGVHD的对照组,cGVHD患者血清中CXCL9、CCL17表达水平显著升高(P<0.05),其中CCL17与cGVHD的疾病严重程度相关(P<0.001);CXCL9在皮肤损害的cGVHD患者血清中显著升高(P<0.01),CCL17在肝脏为靶器官的cGVHD患者中表达水平显著升高(P<0.01)。 结论 CXCL9联合CCL17可作为cGVHD的血清生物标志物,对辅助cGVHD诊断和评估严重程度有一定参考价值。
Collapse
Affiliation(s)
- T Chen
- Medical Center of Hematology, Xinqiao Hospital of Army Medical University, State Key Laboratory of Trauma, Burns and Combined Injury, Chongqing 400037, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Debureaux P, Masson A, Battistella M, Fontbrune F, Socié G, Bouaziz J, Michonneau D. Chronic graft‐versus‐host disease and inhibition of interleukin‐17: proof of concept in humans. Br J Dermatol 2019; 182:1038-1041. [DOI: 10.1111/bjd.18214] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/04/2019] [Indexed: 01/09/2023]
Affiliation(s)
- P.‐E. Debureaux
- Hematology and Transplantation Unit Saint Louis Hospital, APHP Paris France
| | - A. Masson
- Dermatology Unit Saint Louis Hospital, APHP Paris France
- Université de Paris, INSERM U976 Paris France
| | - M. Battistella
- Pathology Department Saint Louis Hospital, APHP Paris France
| | - F. Fontbrune
- Hematology and Transplantation Unit Saint Louis Hospital, APHP Paris France
| | - G. Socié
- Hematology and Transplantation Unit Saint Louis Hospital, APHP Paris France
- Université de Paris, INSERM U976 Paris France
| | - J.D. Bouaziz
- Dermatology Unit Saint Louis Hospital, APHP Paris France
- Université de Paris, INSERM U976 Paris France
| | - D. Michonneau
- Hematology and Transplantation Unit Saint Louis Hospital, APHP Paris France
- Université de Paris, INSERM U976 Paris France
| |
Collapse
|
22
|
Müller AMS, Min D, Wernig G, Levy RB, Perez VL, Herretes S, Florek M, Burnett C, Weinberg K, Shizuru JA. Modeling Chronic Graft-versus-Host Disease in MHC-Matched Mouse Strains: Genetics, Graft Composition, and Tissue Targets. Biol Blood Marrow Transplant 2019; 25:2338-2349. [PMID: 31415899 DOI: 10.1016/j.bbmt.2019.08.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2018] [Revised: 06/22/2019] [Accepted: 08/06/2019] [Indexed: 02/06/2023]
Abstract
Graft-versus-host disease (GVHD) remains a major complication of allogeneic hematopoietic cell transplantation. Acute GVHD (aGVHD) results from direct damage by donor T cells, whereas the biology of chronic GVHD (cGVHD) with its autoimmune-like manifestations remains poorly understood, mainly because of the paucity of representative preclinical models. We examined over an extended time period 7 MHC-matched, minor antigen-mismatched mouse models for development of cGVHD. Development and manifestations of cGVHD were determined by a combination of MHC allele type and recipient strain, with BALB recipients being the most susceptible. The C57BL/6 into BALB.B combination most closely modeled the human syndrome. In this strain combination moderate aGVHD was observed and BALB.B survivors developed overt cGVHD at 6 to 12 months affecting eyes, skin, and liver. Naïve CD4+ cells caused this syndrome as no significant pathology was induced by grafts composed of purified hematopoietic stem cells (HSCs) or HSC plus effector memory CD4+ or CD8+ cells. Furthermore, co-transferred naïve and effector memory CD4+ T cells demonstrated differential homing patterns and locations of persistence. No clear association with donor Th17 cells and the phenotype of aGVHD or cGVHD was observed in this model. Donor CD4+ cells caused injury to medullary thymic epithelial cells, a key population responsible for negative T cell selection, suggesting that impaired thymic selection was an underlying cause of the cGVHD syndrome. In conclusion, we report for the first time that the C57BL/6 into BALB.B combination is a representative model of cGVHD that evolves from immunologic events during the early post-transplant period.
Collapse
Affiliation(s)
- Antonia M S Müller
- Division of Blood and Marrow Transplantation, Department of Medicine, Stanford University School of Medicine, Stanford, California; Department of Hematology, University Hospital and University Zurich, Zurich, Switzerland.
| | - Dullei Min
- Division of Pediatric Blood and Marrow Transplantation, Stanford University School of Medicine, Stanford, California
| | - Gerlinde Wernig
- Department of Pathology, Stanford University School of Medicine, Stanford, California
| | - Robert B Levy
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, Florida
| | - Victor L Perez
- Department of Ophthalmology, University of Miami Miller School of Medicine, Miami, Florida
| | - Samantha Herretes
- Department of Ophthalmology, University of Miami Miller School of Medicine, Miami, Florida
| | - Mareike Florek
- Division of Blood and Marrow Transplantation, Department of Medicine, Stanford University School of Medicine, Stanford, California
| | - Casey Burnett
- Division of Blood and Marrow Transplantation, Department of Medicine, Stanford University School of Medicine, Stanford, California
| | - Kenneth Weinberg
- Department of Hematology, University Hospital and University Zurich, Zurich, Switzerland
| | - Judith A Shizuru
- Division of Blood and Marrow Transplantation, Department of Medicine, Stanford University School of Medicine, Stanford, California; Division of Pediatric Blood and Marrow Transplantation, Stanford University School of Medicine, Stanford, California
| |
Collapse
|
23
|
Yi J, Chen Z, Xu F, Wang Z, Zhang A, Liu T, Zhao N, Xiong Y, Jiang G, Ma J, Luan X. IL-27 Promotes Human Placenta-Derived Mesenchymal Stromal Cell Ability To Induce the Generation of CD4 +IL-10 +IFN-γ + T Cells via the JAK/STAT Pathway in the Treatment of Experimental Graft-versus-Host Disease. THE JOURNAL OF IMMUNOLOGY 2019; 202:1124-1136. [PMID: 30651340 DOI: 10.4049/jimmunol.1800963] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Accepted: 12/10/2018] [Indexed: 12/12/2022]
Abstract
Human mesenchymal stromal cells (MSCs) harbor immunomodulatory properties to induce the generation of suppressive T cells. MSCs have been successfully used in treating graft-versus-host disease (GVHD) accompanied by abundant inflammatory cytokines such as IL-27. This study investigated the effects of IL-27 on the human placenta-derived MSCs (hPMSCs) to induce generation of CD4+IL-10+IFN-γ+ T cells in vitro and in the humanized xenogenic GVHD NOD/SCID model. The results showed that the percentages of CD4+IL-10+IFN-γ+ T cells were significantly increased in activated human PBMC from both healthy donors and GVHD patients with hPMSCs and in the liver and spleen of hPMSC-treated GVHD mice, and the level of CD4+IL-10+IFN-γ+ T cells in the liver was greater than that in the spleen in hPMSC-treated GVHD mice. The serum level of IL-27 decreased and the symptoms abated in hPMSC-treated GVHD. Further, in vitro results showed that IL-27 promoted the regulatory effects of hPMSCs by enhancing the generation of CD4+IL-10+IFN-γ+ T cells from activated PBMC. Activation occurred through increases in the expression of programmed death ligand 2 (PDL2) in hPMSCs via the JAK/STAT signaling pathway. These findings indicated that hPMSCs could alleviate GVHD mice symptoms by upregulating the production of CD4+IL-10+IFN-γ+ T cells in the spleen and liver and downregulating serum levels of IL-27. In turn, the ability of hPMSCs to induce the generation of CD4+IL-10+IFN-γ+ T cells could be promoted by IL-27 through increases in PDL2 expression in hPMSCs. The results of this study will be of benefit for the application of hPMSCs in clinical trials.
Collapse
Affiliation(s)
- Junzhu Yi
- Department of Immunology, Binzhou Medical University, Yantai, Shandong Province 264003, China
| | - Zhenghua Chen
- Yantai Affiliated Hospital of Binzhou Medical University, Yantai, Shandong Province 264100, China
| | - Fenghuang Xu
- The First Affiliated Hospital of Hainan Medical University, Haikou, Hainan Province 570102, China
| | - ZhuoYa Wang
- Department of Immunology, Binzhou Medical University, Yantai, Shandong Province 264003, China
| | - Aiping Zhang
- Department of Immunology, Binzhou Medical University, Yantai, Shandong Province 264003, China
| | - Tongshen Liu
- Department of Histology and Embryology, Binzhou Medical University, Yantai, Shandong Province 264003, China
| | - Nannan Zhao
- Department of Immunology, Binzhou Medical University, Yantai, Shandong Province 264003, China
| | - Yanlian Xiong
- Department of Anatomy, Binzhou Medical University, Yantai, Shandong Province 264003, China
| | - Guosheng Jiang
- Department of Immunology, Binzhou Medical University, Yantai, Shandong Province 264003, China
| | - Junjie Ma
- Yuhuangding Hospital Affiliated to Qingdao University, Yantai, Shandong Province 264000, China; and
| | - Xiying Luan
- Department of Immunology, Binzhou Medical University, Yantai, Shandong Province 264003, China; .,Taishan Scholar Immunology Program, Binzhou Medical University, Yantai, Shandong Province 264003, China
| |
Collapse
|
24
|
Tamibarotene for the Treatment of Bronchiolitis Obliterans Associated With Chronic Graft-vs-Host Disease. Chest 2019; 155:e1-e4. [DOI: 10.1016/j.chest.2018.08.1052] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Revised: 07/16/2018] [Accepted: 08/01/2018] [Indexed: 11/18/2022] Open
|
25
|
Chen X, Mayne CG. The Role of Micronutrients in Graft-VS.-Host Disease: Immunomodulatory Effects of Vitamins A and D. Front Immunol 2018; 9:2853. [PMID: 30574143 PMCID: PMC6291446 DOI: 10.3389/fimmu.2018.02853] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Accepted: 11/20/2018] [Indexed: 01/15/2023] Open
Abstract
Graft-vs.-host disease (GVHD) remains a major obstacle to the success of allogeneic hematopoietic stem cell transplantation (HSCT). GVHD occurs because donor T cells in the allograft recognize the genetically disparate host as foreign and attack the transplant recipient's tissues. While genetic incompatibility between donor and recipient is the primary determinant for the extent of alloimmune response, GVHD incidence and severity are also influenced by non-genetic factors. Recent advances in immunology establish that environmental factors, including dietary micronutrients, contribute significantly to modulating various immune responses and may influence the susceptibility to autoimmune and inflammatory diseases of experimental animals and humans. Emerging clinical and preclinical evidence indicates that certain micronutrients may participate in regulating GVHD risk after allogeneic HSCT. In this review, we summarize recent advances in our understanding with respect to the potential role of micronutrients in the pathogenesis of acute and chronic GVHD, focusing on vitamins A and D.
Collapse
Affiliation(s)
- Xiao Chen
- Division of Hematology & Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, United States
| | | |
Collapse
|
26
|
Lai P, Chen X, Guo L, Wang Y, Liu X, Liu Y, Zhou T, Huang T, Geng S, Luo C, Huang X, Wu S, Ling W, Du X, He C, Weng J. A potent immunomodulatory role of exosomes derived from mesenchymal stromal cells in preventing cGVHD. J Hematol Oncol 2018; 11:135. [PMID: 30526632 PMCID: PMC6286548 DOI: 10.1186/s13045-018-0680-7] [Citation(s) in RCA: 122] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Accepted: 11/21/2018] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Mesenchymal stromal cells (MSCs) are a promising therapy for preventing chronic Graft-Versus-Host Disease (cGVHD) due to their potent immunomodulatory properties. However, the safety concerns regarding the use of MSCs remain unsolved, and conflicting effects are observed due to the heterogeneity of MSCs. Recently, exosomes were shown to mediate the paracrine effects of MSCs, making it a potential candidate for cell-free therapies. The aim of this study is to investigate the efficacy and safety of MSCs-derived exosomes (MSCs-exo) in an established cGVHD mouse model. METHODS Bone marrow (BM)-derived MSCs were cultured, and the supernatants of these cultures were collected to prepare exosomes using ultracentrifugation. Exosomes from human dermal fibroblasts (Fib-exo) were used as a negative control. The cGVHD model was established, and tail vein injections of MSCs-exo or Fib-exo were administered once per week for 6 weeks. The symptoms and signs of cGVHD were monitored, and histopathological changes were detected by hematoxylin and eosin and Masson staining. The effects of MSCs-exo on Th17, Th1, and Treg were evaluated by flow cytometry, qPCR, and Luminex. In addition, human peripheral blood mononuclear cells (PBMCs) were stimulated and treated with MSCs-exo in vitro. IL-17-expressing Th17 and IL-10-expressing Treg were evaluated by flow cytometry, qPCR, and ELISA. RESULTS We found that MSCs-exo effectively prolonged the survival of cGVHD mice and diminished the clinical and pathological scores of cGVHD. Fibrosis in the skin, lung, and liver was significantly ameliorated by MSCs-exo application. In MSCs-exo treated mice, activation of CD4+ T cells and their infiltration into the lung were reduced. Of note, MSCs-exo exhibited potent immunomodulatory effects via the inhibition of IL-17-expressing pathogenic T cells and induction of IL-10-expressing regulatory cells during cGVHD. The expressions of Th17 cell-relevant transcription factors and pro-inflammatory cytokines was markedly reduced after MSCs-exo treatment. In vitro, MSCs-exo blocked Th17 differentiation and improved the Treg phenotype in PBMCs obtained from healthy donors and patients with active cGVHD, further indicating the regulatory effect of MSCs-exo on GVHD effector T cells. CONCLUSIONS Our data suggested that MSCs-exo could improve the survival and ameliorate the pathologic damage of cGVHD by suppressing Th17 cells and inducing Treg. This finding provides a novel alternative approach for the treatment of cGVHD.
Collapse
Affiliation(s)
- Peilong Lai
- Department of Hematology, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, 510080, People's Republic of China.,Guangdong Geriatrics Institute, Guangzhou, Guangdong, 510080, People's Republic of China
| | - Xiaomei Chen
- Department of Hematology, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, 510080, People's Republic of China
| | - Liyan Guo
- Department of Hematology, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, 510080, People's Republic of China
| | - Yulian Wang
- Department of Hematology, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, 510080, People's Republic of China
| | - Xialin Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, 510060, People's Republic of China
| | - Yan Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, 510060, People's Republic of China
| | - Tian Zhou
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, 510060, People's Republic of China
| | - Tian Huang
- Department of Hematology, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, 510080, People's Republic of China
| | - Suxia Geng
- Department of Hematology, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, 510080, People's Republic of China
| | - Chengwei Luo
- Department of Hematology, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, 510080, People's Republic of China
| | - Xin Huang
- Department of Hematology, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, 510080, People's Republic of China
| | - Suijing Wu
- Department of Hematology, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, 510080, People's Republic of China
| | - Wei Ling
- Department of Hematology, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, 510080, People's Republic of China
| | - Xin Du
- Department of Hematology, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, 510080, People's Republic of China. .,Guangdong Geriatrics Institute, Guangzhou, Guangdong, 510080, People's Republic of China.
| | - Chang He
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, 510060, People's Republic of China.
| | - Jianyu Weng
- Department of Hematology, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, 510080, People's Republic of China. .,Guangdong Geriatrics Institute, Guangzhou, Guangdong, 510080, People's Republic of China.
| |
Collapse
|
27
|
Cafferata EA, Jerez A, Vernal R, Monasterio G, Pandis N, Faggion CM. The therapeutic potential of regulatory T lymphocytes in periodontitis: A systematic review. J Periodontal Res 2018; 54:207-217. [DOI: 10.1111/jre.12629] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Revised: 10/23/2018] [Accepted: 10/28/2018] [Indexed: 12/11/2022]
Affiliation(s)
- Emilio Alfredo Cafferata
- Periodontal Biology LaboratoryFaculty of DentistryUniversidad de Chile Santiago Chile
- Faculty of DentistryUniversidad Peruana Cayetano Heredia Lima Perú
| | - Alfredo Jerez
- Department of Oral SurgerySection of PeriodontologySchool of DentistryUniversidad de Concepción Concepción Chile
| | - Rolando Vernal
- Periodontal Biology LaboratoryFaculty of DentistryUniversidad de Chile Santiago Chile
- Dentistry UnitFaculty of Health SciencesUniversidad Autónoma de Chile Santiago Chile
| | - Gustavo Monasterio
- Periodontal Biology LaboratoryFaculty of DentistryUniversidad de Chile Santiago Chile
| | - Nikolaos Pandis
- Department of Orthodontics and Dentofacial OrthopedicsDental School/Medical FacultyUniversity of Bern Bern Switzerland
| | - Clovis M. Faggion
- Department of Periodontology and Operative DentistryFaculty of DentistryUniversity of Münster Münster Germany
| |
Collapse
|
28
|
Wu J, Gu J, Zhou S, Lu H, Lu Y, Lu L, Wang X. Anti-IL-22 Antibody Attenuates Acute Graft-versus-Host Disease via Increasing Foxp3 + T Cell through Modulation of CD11b + Cell Function. J Immunol Res 2018; 2018:1605341. [PMID: 30159338 PMCID: PMC6109487 DOI: 10.1155/2018/1605341] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2018] [Revised: 06/20/2018] [Accepted: 06/28/2018] [Indexed: 12/14/2022] Open
Abstract
Transfer of splenocytes isolated from B6 mice into normal B6D2F1 mice induces acute graft-versus-host disease (aGVHD), resulting in the expansion of donor cytotoxic T lymphocytes that eliminate recipient B cells. The cytokine IL-22, secreted by Th1 cells, Th17 cells, and innate immune cells, is structurally related to IL-10. To investigate the association between IL-22 and aGVHD, an anti-mouse IL-22 antibody (IL-22Ab) was used to ablate IL-22 activity in a mouse aGVHD model. Administration of IL-22Ab significantly reduced the progression of aGVHD in B6D2F1 recipients of B6 grafts. IL-22Ab treatment also decreased the percentage of interferon-γ+ and tumor necrosis factor-α+ T cells but increased the number of forkhead box p3+ regulatory T cells (Tregs). In the presence of Tregs and donor CD11b+ cells, IL-22Ab protected against aGVHD. In vitro Treg induction was more efficient when CD4+CD25- T cells differentiated in the presence of CD11b+ cells obtained from IL-22Ab-treated GVHD mice, compared with cocultured untreated control cells. Finally, IL-22Ab modulated the expression of cytokines and costimulatory molecules in CD11b+ cells in aGVHD mice. We therefore conclude that IL-22Ab administration represents a viable approach for treating aGVHD.
Collapse
Affiliation(s)
- Jianbo Wu
- Department of Liver Surgery, First Affiliated Hospital of Nanjing Medical University, Key Laboratory of Living Donor Liver Transplantation, National Health and Family Planning Commission, Nanjing, Jiangsu Province, China
- Department of General Surgery, Changzhou No. 2 People's Hospital Affiliated to Nanjing Medical University, Changzhou City, Jiangsu Province, China
| | - Jian Gu
- Department of Liver Surgery, First Affiliated Hospital of Nanjing Medical University, Key Laboratory of Living Donor Liver Transplantation, National Health and Family Planning Commission, Nanjing, Jiangsu Province, China
| | - Shun Zhou
- Department of Liver Surgery, First Affiliated Hospital of Nanjing Medical University, Key Laboratory of Living Donor Liver Transplantation, National Health and Family Planning Commission, Nanjing, Jiangsu Province, China
| | - Hao Lu
- Department of Liver Surgery, First Affiliated Hospital of Nanjing Medical University, Key Laboratory of Living Donor Liver Transplantation, National Health and Family Planning Commission, Nanjing, Jiangsu Province, China
| | - Yunjie Lu
- Department of Liver Surgery, First Affiliated Hospital of Nanjing Medical University, Key Laboratory of Living Donor Liver Transplantation, National Health and Family Planning Commission, Nanjing, Jiangsu Province, China
| | - Ling Lu
- Department of Liver Surgery, First Affiliated Hospital of Nanjing Medical University, Key Laboratory of Living Donor Liver Transplantation, National Health and Family Planning Commission, Nanjing, Jiangsu Province, China
| | - Xuehao Wang
- Department of Liver Surgery, First Affiliated Hospital of Nanjing Medical University, Key Laboratory of Living Donor Liver Transplantation, National Health and Family Planning Commission, Nanjing, Jiangsu Province, China
| |
Collapse
|
29
|
Phase 1 clinical trial evaluating abatacept in patients with steroid-refractory chronic graft-versus-host disease. Blood 2018; 131:2836-2845. [DOI: 10.1182/blood-2017-05-780239] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Accepted: 03/07/2018] [Indexed: 12/11/2022] Open
Abstract
Key Points
Costimulatory blockade using abatacept represents a novel therapeutic approach for the treatment of cGVHD. Abatacept resulted in a clinical response in 44% of patients with both decreased prednisone use and T-cell PD-1 expression in responders.
Collapse
|
30
|
Zhang L, Yu J, Wei W. Advance in Targeted Immunotherapy for Graft-Versus-Host Disease. Front Immunol 2018; 9:1087. [PMID: 29868032 PMCID: PMC5964137 DOI: 10.3389/fimmu.2018.01087] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2018] [Accepted: 05/01/2018] [Indexed: 01/08/2023] Open
Abstract
Graft-versus-host disease (GVHD) is a serious and deadly complication of patients, who undergo hematopoietic stem cell transplantation (HSCT). Despite prophylactic treatment with immunosuppressive agents, 20–80% of recipients develop acute GVHD after HSCT. And the incidence rates of chronic GVHD range from 6 to 80%. Standard therapeutic strategies are still lacking, although considerable advances have been gained in knowing of the predisposing factors, pathology, and diagnosis of GVHD. Targeting immune cells, such as regulatory T cells, as well as tolerogenic dendritic cells or mesenchymal stromal cells (MSCs) display considerable benefit in the relief of GVHD through the deletion of alloactivated T cells. Monoclonal antibodies targeting cytokines or signaling molecules have been demonstrated to be beneficial for the prevention of GVHD. However, these remain to be verified in clinical therapy. It is also important and necessary to consider adopting individualized treatment based on GVHD subtypes, pathological mechanisms involved and stages. In the future, it is hoped that the identification of novel therapeutic targets and systematic research strategies may yield novel safe and effective approaches in clinic to improve outcomes of GVHD further. In this article, we reviewed the current advances in targeted immunotherapy for the prevention of GVHD.
Collapse
Affiliation(s)
- Lingling Zhang
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-Inflammatory and Immunopharmacology of Education, Ministry of China, Anti-Inflammatory Immune Drugs Collaborative Innovation Center, Hefei, Anhui, China
| | - Jianhua Yu
- Division of Hematology, Department of Internal Medicine, College of Medicine, The Ohio State University, Columbus, OH, United States
| | - Wei Wei
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-Inflammatory and Immunopharmacology of Education, Ministry of China, Anti-Inflammatory Immune Drugs Collaborative Innovation Center, Hefei, Anhui, China
| |
Collapse
|
31
|
Nair S, Vanathi M, Mahapatra M, Seth T, Kaur J, Velpandian T, Ravi A, Titiyal JS, Tandon R. Tear inflammatory mediators and protein in eyes of post allogenic hematopoeitic stem cell transplant patients. Ocul Surf 2018; 16:352-367. [PMID: 29723628 DOI: 10.1016/j.jtos.2018.04.007] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Revised: 04/21/2018] [Accepted: 04/24/2018] [Indexed: 10/17/2022]
Abstract
AIM To analyze tear cytokines levels and their correlation to ocular surface parameters in allogenic hematopoietic stem cell transplants (allo-HSCT) patients. METHODS Prospective longitudinal study of allo-HSCT patients and controls for ocular surface evaluation (OSDI, TBUT, Schirmer's test, staining scores), tear biochemical analysis for protein, cytokines [IL-10, IL-12, IL-2, IL-4, IL-6, IL-17, interferon (IFN)-gamma, tumor necrosis factor (TNF)-alpha, VEGF], MMPs [MMP 2, 9, 7, 13, 10 and chemokine (IL-8)], & VEGF on three consecutive follow up visits (at three monthly interval) was done. RESULTS Of 24 post allo-HSCT patients (19 males, 5 females) & 12 controls (mean age 34.3 + 5.8 years) enrolled, 20 patients [mean age 33.4 + 7.77 years; mean time of recruitment of 5.2 + 2.12 months following alloHSCT] who completed three consecutive follow up visits were included for analysis. Ocular GVHD (oGVHD) was seen in 8 patients (33.3%). Tears biochemical analysis showed elevated levels of interferon γ, IL 6, IL 8, IL 10, IL 12AP70, IL 17A, MMP 9 and VEGF in oGVHD eyes as compared to non-oGVHD & control eyes. Non-oGVHD eyes showed elevated tear MMP 7 and MMP 9 as compared to healthy controls. Tear protein levels were significantly decreased in oGVHD eyes and were equivocal in nonGVHD and control eyes. TBUT and ocular staining scores to correlate best with tear interleukins and MMPs. CONCLUSION Evaluation of levels of tear VEGF, total protein & MMP 9 can be of significance in identifying oGVHD in post alloHSCT patients.
Collapse
Affiliation(s)
- Sridevi Nair
- Cornea & Ocular Surface Services, Dr Rajendra Prasad Centre for Ophthalmic Sciences, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Murugesan Vanathi
- Cornea & Ocular Surface Services, Dr Rajendra Prasad Centre for Ophthalmic Sciences, All India Institute of Medical Sciences, New Delhi, 110029, India.
| | - Manoranjan Mahapatra
- Department of Hematology, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Tulika Seth
- Department of Hematology, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Jasbir Kaur
- Ocular Biochemistry Services, Dr Rajendra Prasad Centre for Ophthalmic Sciences, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - T Velpandian
- Ocular Pharmacology Services, Dr Rajendra Prasad Centre for Ophthalmic Sciences, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Alok Ravi
- Ocular Biochemistry Services, Dr Rajendra Prasad Centre for Ophthalmic Sciences, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Jeewan Singh Titiyal
- Cornea & Ocular Surface Services, Dr Rajendra Prasad Centre for Ophthalmic Sciences, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Radhika Tandon
- Cornea & Ocular Surface Services, Dr Rajendra Prasad Centre for Ophthalmic Sciences, All India Institute of Medical Sciences, New Delhi, 110029, India
| |
Collapse
|
32
|
Vitamin A–coupled liposomes containing siRNA against HSP47 ameliorate skin fibrosis in chronic graft-versus-host disease. Blood 2018; 131:1476-1485. [DOI: 10.1182/blood-2017-04-779934] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Accepted: 01/16/2018] [Indexed: 02/07/2023] Open
Abstract
Key Points
HSP47+ myofibroblasts are accumulated in the fibrotic lesions of chronic GVHD and promote fibrosis in a CSF-1R+ macrophage-dependent manner. Vitamin A–coupled liposomes containing HSP47 siRNA abrogate HSP47 expression in myofibroblasts and ameliorate fibrosis in chronic GVHD.
Collapse
|
33
|
MicroRNA-17-92 is required for T-cell and B-cell pathogenicity in chronic graft-versus-host disease in mice. Blood 2018. [PMID: 29530952 DOI: 10.1182/blood-2017-06-789321] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Chronic graft-versus-host disease (cGVHD) is characterized as autoimmune-like fibrosis and antibody production mediated by pathogenic T cells and B cells. MicroRNA-17-92 (miR-17-92) influences the survival, differentiation, and function of lymphocytes in cancer, infections, and autoimmunity. To determine whether miR-17-92 regulates T- and B-cell responses in cGVHD, we generated mice conditionally deficient for miR-17-92 in T cells, B cells, or both. Using murine models of allogeneic bone marrow transplantation, we demonstrate that expression of miR-17-92 in donor T and B cells is essential for the induction of both scleroderma and bronchiolitis obliterans in cGVHD. Mechanistically, miR-17-92 expressed in T cells not only enhances the differentiation of pathogenic T helper 1 (Th1) and Th17 cells, but also promotes the generation of follicular Th cells, germinal center (GC) B cells, and plasma cells. In B cells, miR-17-92 expression is required for autoantibody production and immunoglobulin G deposition in the skin. Furthermore, we evaluated a translational approach using antagomirs specific for either miR-17 or miR-19, key members in miR-17-92 cluster. In a lupus-like cGVHD model, systemic administration of anti-miR-17, but not anti-miR-19, alleviates clinical manifestations and proteinuria incidence in recipients through inhibiting donor lymphocyte expansion, B-cell activation, and GC responses. Blockade of miR-17 also ameliorates skin damage by reducing Th17 differentiation in a scleroderma-cGVHD model. Taken together, our work reveals that miR-17-92 is required for T-cell and B-cell differentiation and function, and thus for the development of cGVHD. Furthermore, pharmacological inhibition of miR-17 represents a potential therapeutic strategy for the prevention of cGVHD.
Collapse
|
34
|
Joean O, Hueber A, Feller F, Jirmo AC, Lochner M, Dittrich AM, Albrecht M. Suppression of Th17-polarized airway inflammation by rapamycin. Sci Rep 2017; 7:15336. [PMID: 29127369 PMCID: PMC5681547 DOI: 10.1038/s41598-017-15750-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Accepted: 10/31/2017] [Indexed: 12/18/2022] Open
Abstract
Because Th17-polarized airway inflammation correlates with poor control in bronchial asthma and is a feature of numerous other difficult-to-treat inflammatory lung diseases, new therapeutic approaches for this type of airway inflammation are necessary. We assessed different licensed anti-inflammatory agents with known or expected efficacy against Th17-polarization in mouse models of Th17-dependent airway inflammation. Upon intravenous transfer of in vitro derived Th17 cells and intranasal challenge with the corresponding antigen, we established acute and chronic murine models of Th17-polarised airway inflammation. Consecutively, we assessed the efficacy of methylprednisolone, roflumilast, azithromycin, AM80 and rapamycin against acute or chronic Th17-dependent airway inflammation. Quantifiers for Th17-associated inflammation comprised: bronchoalveolar lavage (BAL) differential cell counts, allergen-specific cytokine and immunoglobulin secretion, as well as flow cytometric phenotyping of pulmonary inflammatory cells. Only rapamycin proved effective against acute Th17-dependent airway inflammation, accompanied by increased plasmacytoid dendritic cells (pDCs) and reduced neutrophils as well as reduced CXCL-1 levels in BAL. Chronic Th17-dependent airway inflammation was unaltered by rapamycin treatment. None of the other agents showed efficacy in our models. Our results demonstrate that Th17-dependent airway inflammation is difficult to treat with known agents. However, we identify rapamycin as an agent with inhibitory potential against acute Th17-polarized airway inflammation.
Collapse
Affiliation(s)
- Oana Joean
- Department for Pediatric Pneumology, Allergology and Neonatology, Medical School Hannover, Carl-Neuberg-Str. 1, Hannover, Germany.,Department of Internal Medicine B, University Medicine Greifswald, Ferdinand-Sauerbruch-Str., Greifswald, Germany
| | - Anja Hueber
- Department for Pediatric Pneumology, Allergology and Neonatology, Medical School Hannover, Carl-Neuberg-Str. 1, Hannover, Germany
| | - Felix Feller
- Department for Pediatric Pneumology, Allergology and Neonatology, Medical School Hannover, Carl-Neuberg-Str. 1, Hannover, Germany
| | - Adan Chari Jirmo
- Department for Pediatric Pneumology, Allergology and Neonatology, Medical School Hannover, Carl-Neuberg-Str. 1, Hannover, Germany.,German Center for Lunge Research, BREATH Carl-Neuberg-Str. 1, Hannover, Germany
| | - Matthias Lochner
- Institute of Infection Immunology, TWINCORE, Centre for Experimental and Clinical Infection Research, Hannover, Germany
| | - Anna-Maria Dittrich
- Department for Pediatric Pneumology, Allergology and Neonatology, Medical School Hannover, Carl-Neuberg-Str. 1, Hannover, Germany.,German Center for Lunge Research, BREATH Carl-Neuberg-Str. 1, Hannover, Germany
| | - Melanie Albrecht
- Department for Pediatric Pneumology, Allergology and Neonatology, Medical School Hannover, Carl-Neuberg-Str. 1, Hannover, Germany. .,German Center for Lunge Research, BREATH Carl-Neuberg-Str. 1, Hannover, Germany.
| |
Collapse
|
35
|
An aberrant NOTCH2-BCR signaling axis in B cells from patients with chronic GVHD. Blood 2017; 130:2131-2145. [PMID: 28851699 DOI: 10.1182/blood-2017-05-782466] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Accepted: 08/24/2017] [Indexed: 12/16/2022] Open
Abstract
B-cell receptor (BCR)-activated B cells contribute to pathogenesis in chronic graft-versus-host disease (cGVHD), a condition manifested by both B-cell autoreactivity and immune deficiency. We hypothesized that constitutive BCR activation precluded functional B-cell maturation in cGVHD. To address this, we examined BCR-NOTCH2 synergy because NOTCH has been shown to increase BCR responsiveness in normal mouse B cells. We conducted ex vivo activation and signaling assays of 30 primary samples from hematopoietic stem cell transplantation patients with and without cGVHD. Consistent with a molecular link between pathways, we found that BCR-NOTCH activation significantly increased the proximal BCR adapter protein BLNK. BCR-NOTCH activation also enabled persistent NOTCH2 surface expression, suggesting a positive feedback loop. Specific NOTCH2 blockade eliminated NOTCH-BCR activation and significantly altered NOTCH downstream targets and B-cell maturation/effector molecules. Examination of the molecular underpinnings of this "NOTCH2-BCR axis" in cGVHD revealed imbalanced expression of the transcription factors IRF4 and IRF8, each critical to B-cell differentiation and fate. All-trans retinoic acid (ATRA) increased IRF4 expression, restored the IRF4-to-IRF8 ratio, abrogated BCR-NOTCH hyperactivation, and reduced NOTCH2 expression in cGVHD B cells without compromising viability. ATRA-treated cGVHD B cells had elevated TLR9 and PAX5, but not BLIMP1 (a gene-expression pattern associated with mature follicular B cells) and also attained increased cytosine guanine dinucleotide responsiveness. Together, we reveal a mechanistic link between NOTCH2 activation and robust BCR responses to otherwise suboptimal amounts of surrogate antigen. Our findings suggest that peripheral B cells in cGVHD patients can be pharmacologically directed from hyperactivation toward maturity.
Collapse
|
36
|
MacDonald KP, Blazar BR, Hill GR. Cytokine mediators of chronic graft-versus-host disease. J Clin Invest 2017; 127:2452-2463. [PMID: 28665299 DOI: 10.1172/jci90593] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Substantial preclinical and clinical research into chronic graft-versus-host disease (cGVHD) has come to fruition in the last five years, generating a clear understanding of a complex cytokine-driven cellular network. cGVHD is mediated by naive T cells differentiating within IL-17-secreting T cell and follicular Th cell paradigms to generate IL-21 and IL-17A, which drive pathogenic germinal center (GC) B cell reactions and monocyte-macrophage differentiation, respectively. cGVHD pathogenesis includes thymic damage, impaired antigen presentation, and a failure in IL-2-dependent Treg homeostasis. Pathogenic GC B cell and macrophage reactions culminate in antibody formation and TGF-β secretion, respectively, leading to fibrosis. This new understanding permits the design of rational cytokine and intracellular signaling pathway-targeted therapeutics, reviewed herein.
Collapse
Affiliation(s)
- Kelli Pa MacDonald
- Antigen Presentation and Immunoregulation Laboratory, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Bruce R Blazar
- Masonic Cancer Center; and Division of Blood and Marrow Transplantation, Department of Pediatrics; University of Minnesota, Minneapolis, USA
| | - Geoffrey R Hill
- Bone Marrow Transplantation Laboratory, QIMR Berghofer Medical Research Institute, Brisbane, Australia.,Royal Brisbane and Women's Hospital, Brisbane, Australia
| |
Collapse
|
37
|
Forcade E, Paz K, Flynn R, Griesenauer B, Amet T, Li W, Liu L, Bakoyannis G, Jiang D, Chu HW, Lobera M, Yang J, Wilkes DS, Du J, Gartlan K, Hill GR, MacDonald KP, Espada EL, Blanco P, Serody JS, Koreth J, Cutler CS, Antin JH, Soiffer RJ, Ritz J, Paczesny S, Blazar BR. An activated Th17-prone T cell subset involved in chronic graft-versus-host disease sensitive to pharmacological inhibition. JCI Insight 2017; 2:92111. [PMID: 28614794 DOI: 10.1172/jci.insight.92111] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2016] [Accepted: 05/16/2017] [Indexed: 01/13/2023] Open
Abstract
Chronic graft-versus-host disease (cGvHD) remains a major complication of allogeneic stem cell transplantation requiring novel therapies. CD146 and CCR5 are expressed by activated T cells and associated with increased T cell migration capacity and Th17 polarization. We performed a multiparametric flow cytometry analysis in a cohort of 40 HSCT patients together with a cGvHD murine model to understand the role of CD146-expressing subsets. We observed an increased frequency of CD146+ CD4 T cells in the 20 patients with active cGvHD with enhanced RORγt expression. This Th17-prone subset was enriched for cells coexpressing CD146 and CCR5 that harbor mixed Th1/Th17 features and were more frequent in cGvHD patients. Utilizing a murine cGvHD model with bronchiolitis obliterans (BO), we observed that donor T cells from CD146-deficient mice versus those from WT mice caused significantly reduced pulmonary cGvHD. Reduced cGvHD was not the result of failed germinal center B cell or T follicular helper cell generation. Instead, CD146-deficient T cells had significantly lower pulmonary macrophage infiltration and T cell CCR5, IL-17, and IFN-γ coexpression, suggesting defective pulmonary end-organ effector mechanisms. We, thus, evaluated the effect of TMP778, a small-molecule RORγt activity inhibitor. TMP778 markedly alleviated cGvHD in murine models similarly to agents targeting the Th17 pathway, such as STAT3 inhibitor or IL-17-blocking antibody. Our data suggest CD146-expressing T cells as a cGvHD biomarker and suggest that targeting the Th17 pathway may represent a promising therapy for cGvHD.
Collapse
Affiliation(s)
- Edouard Forcade
- Division of Hematologic Malignancies and Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA.,Harvard Medical School, Boston, Massachusetts, USA.,Immunoconcept, CNRS UMR 5164, Bordeaux University, Bordeaux, France.,Department of Hematology and Cell Therapy, University Hospital, Bordeaux, France
| | - Katelyn Paz
- Division of Blood and Marrow Transplantation, University of Minnesota, Minneapolis, Minnesota, USA
| | - Ryan Flynn
- Division of Blood and Marrow Transplantation, University of Minnesota, Minneapolis, Minnesota, USA
| | - Brad Griesenauer
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana, USA.,Herman B. Wells Center for Pediatric Research.,Department of Microbiology and Immunology, and.,Melvin and Bren Simon Cancer Center, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Tohti Amet
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana, USA.,Herman B. Wells Center for Pediatric Research.,Department of Microbiology and Immunology, and.,Melvin and Bren Simon Cancer Center, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Wei Li
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana, USA.,Herman B. Wells Center for Pediatric Research.,Department of Microbiology and Immunology, and.,Melvin and Bren Simon Cancer Center, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Liangyi Liu
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana, USA.,Herman B. Wells Center for Pediatric Research.,Department of Microbiology and Immunology, and.,Melvin and Bren Simon Cancer Center, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Giorgos Bakoyannis
- Department of Biostatistics, Indiana University Fairbanks School of Public Health and School of Medicine, Indiana, USA
| | - Di Jiang
- National Jewish Health, Denver, Colorado, USA
| | | | | | | | - David S Wilkes
- Dean, University of Virginia, School of Medicine, Charlottesville, Virginia, USA
| | - Jing Du
- Division of Blood and Marrow Transplantation, University of Minnesota, Minneapolis, Minnesota, USA
| | - Kate Gartlan
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Geoffrey R Hill
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Kelli Pa MacDonald
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Eduardo L Espada
- Division of Hematologic Malignancies and Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA.,Harvard Medical School, Boston, Massachusetts, USA
| | - Patrick Blanco
- Immunoconcept, CNRS UMR 5164, Bordeaux University, Bordeaux, France
| | - Jonathan S Serody
- Lineberger Comprehensive Cancer Center and Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - John Koreth
- Division of Hematologic Malignancies and Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA.,Harvard Medical School, Boston, Massachusetts, USA
| | - Corey S Cutler
- Division of Hematologic Malignancies and Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA.,Harvard Medical School, Boston, Massachusetts, USA
| | - Joseph H Antin
- Division of Hematologic Malignancies and Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA.,Harvard Medical School, Boston, Massachusetts, USA
| | - Robert J Soiffer
- Division of Hematologic Malignancies and Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA.,Harvard Medical School, Boston, Massachusetts, USA
| | - Jerome Ritz
- Division of Hematologic Malignancies and Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA.,Harvard Medical School, Boston, Massachusetts, USA
| | - Sophie Paczesny
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana, USA.,Herman B. Wells Center for Pediatric Research.,Department of Microbiology and Immunology, and.,Melvin and Bren Simon Cancer Center, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Bruce R Blazar
- Division of Blood and Marrow Transplantation, University of Minnesota, Minneapolis, Minnesota, USA
| |
Collapse
|
38
|
Sprouty-Related Ena/Vasodilator-Stimulated Phosphoprotein Homology 1-Domain-Containing Protein-2 Critically Regulates Influenza A Virus-Induced Pneumonia. Crit Care Med 2017; 44:e530-43. [PMID: 26757161 DOI: 10.1097/ccm.0000000000001562] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
OBJECTIVES Influenza A virus causes acute respiratory infections that induce annual epidemics and occasional pandemics. Although a number of studies indicated that the virus-induced intracellular signaling events are important in combating influenza virus infection, the mechanism how specific molecule plays a critical role among various intracellular signaling events remains unknown. Raf/MEK/extracellular signal-regulated kinase cascade is one of the key signaling pathways during influenza virus infection, and the Sprouty-related Ena/vasodilator-stimulated phosphoprotein homology 1-domain-containing protein has recently been identified as a negative regulator of Raf-dependent extracellular signal-regulated kinase activation. Here, we examined the role of Raf/MEK/extracellular signal-regulated kinase cascade through sprouty-related Ena/vasodilator-stimulated phosphoprotein homology 1-domain-containing protein in influenza A viral infection because the expression of sprouty-related Ena/vasodilator-stimulated phosphoprotein homology 1-domain-containing protein was significantly enhanced in human influenza viral-induced pneumonia autopsy samples. DESIGN Prospective animal trial. SETTING Research laboratory. SUBJECTS Wild-type and sprouty-related Ena/vasodilator-stimulated phosphoprotein homology 1-domain-containing protein-2 knockout mice inoculated with influenza A. INTERVENTIONS Wild-type or sprouty-related Ena/vasodilator-stimulated phosphoprotein homology 1-domain-containing protein-2 knockout mice were infected by intranasal inoculation of influenza A (A/PR/8). An equal volume of phosphate-buffered saline was inoculated intranasally into mock-infected mice. MEASUREMENTS AND MAIN RESULTS Influenza A infection of sprouty-related Ena/vasodilator-stimulated phosphoprotein homology 1-domain-containing protein-2 knockout mice led to higher mortality with greater viral load, excessive inflammation, and enhanced cytokine production than wild-type mice. Administration of MEK inhibitor, U0126, improved mortality and reduced both viral load and cytokine levels. Furthermore, bone marrow chimeras indicated that influenza A-induced lung pathology was most severe when sprouty-related Ena/vasodilator-stimulated phosphoprotein homology 1-domain-containing protein-2 expression was lacking in nonimmune cell populations. Furthermore, microarray analysis revealed knockdown of sprouty-related Ena/vasodilator-stimulated phosphoprotein homology 1-domain-containing protein-2 led to enhanced phosphatidylinositol 3-kinase signaling pathway, resulting that viral clearance was regulated by sprouty-related Ena/vasodilator-stimulated phosphoprotein homology 1-domain-containing protein-2 expression through the phosphatidylinositol 3-kinase signaling pathway in murine lung epithelial cells. CONCLUSIONS These data support an important function of sprouty-related Ena/vasodilator-stimulated phosphoprotein homology 1-domain-containing protein-2 in controlling influenza virus-induced pneumonia and viral replication. Sprouty-related Ena/vasodilator-stimulated phosphoprotein homology 1-domain-containing protein-2 may be a novel therapeutic target for controlling the immune response against influenza influenza A virus infection.
Collapse
|
39
|
Amarnath S, Laurence A, Zhu N, Cunha R, Eckhaus MA, Taylor S, Foley JE, Ghosh M, Felizardo TC, Fowler DH. Tbet is a critical modulator of FoxP3 expression in autoimmune graft- versus-host disease. Haematologica 2017; 102:1446-1456. [PMID: 28473623 PMCID: PMC5541878 DOI: 10.3324/haematol.2016.155879] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2016] [Accepted: 05/03/2017] [Indexed: 12/14/2022] Open
Abstract
CD4+ T-helper subsets drive autoimmune chronic graft-versus-host disease, a major complication after allogeneic bone marrow transplantation. However, it remains unclear how specific T-helper subsets contribute to chronic graft-versus-host disease. T-helper type 1 cells are one of the major disease-mediating T-cell subsets and require interferon-γ signaling and Tbet expression for their function. Regulatory T cells on the other hand can inhibit T-helper type 1 cell-mediated responses. Using an established murine model that isolates the autoimmune component of graft-versus-host disease, we hypothesized that T-helper type 1 cells would restrict FoxP3-driven regulatory T cells. Upon transfer into immune-deficient syngeneic hosts, alloreactive Tbx21−/−CD4+ T cells led to marked increases in FoxP3+ cells and reduced clinical evidence of autoimmunity. To evaluate whether peripheral induction contributed to regulatory T-cell predominance, we adoptively transferred Tbx21−/− T cells that consisted of fate mapping for FoxP3: recipients of flow-purified effector cells that were Foxp3− and Tbx21−/− had enhanced T-regulatory-cell predominance during autoimmune graft-versus-host disease. These data directly demonstrated that peripheral T-regulatory-cell induction was inhibited by Tbet. Finally, Tbx21−/− T-regulatory cells cross-regulated autoimmune wild-type T-effector-cell cytokine production in vivo. The Tbet pathway therefore directly impairs T-regulatory-cell reconstitution and is consequently a feasible target in efforts to prevent autoimmune graft-versus-host disease.
Collapse
Affiliation(s)
- Shoba Amarnath
- Institute of Cellular Medicine, Newcastle University, Newcastle Upon Tyne, UK, USA
| | - Arian Laurence
- Institute of Cellular Medicine, Newcastle University, Newcastle Upon Tyne, UK, USA
| | - Nathaniel Zhu
- Experimental Transplantation Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Renato Cunha
- Experimental Transplantation Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Michael A Eckhaus
- Division of Veterinary Resources, Office of Research Services, Bethesda, MD, USA
| | - Samuel Taylor
- Experimental Transplantation Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Jason E Foley
- Experimental Transplantation Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Monalisa Ghosh
- Experimental Transplantation Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Tania C Felizardo
- Experimental Transplantation Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Daniel H Fowler
- Experimental Transplantation Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
40
|
Ito R, Katano I, Kawai K, Yagoto M, Takahashi T, Ka Y, Ogura T, Takahashi R, Ito M. A Novel Xenogeneic Graft-Versus-Host Disease Model for Investigating the Pathological Role of Human CD4 + or CD8 + T Cells Using Immunodeficient NOG Mice. Am J Transplant 2017; 17:1216-1228. [PMID: 27862942 DOI: 10.1111/ajt.14116] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2016] [Revised: 10/24/2016] [Accepted: 11/08/2016] [Indexed: 02/06/2023]
Abstract
Graft-versus-host disease (GVHD) is a major complication of allogenic bone marrow transplantation and involves the infiltration of donor CD4+ and/or CD8+ T cells into various organs of the recipient. The pathological role of human CD4+ and CD8+ T cells in GVHD remains controversial. In this study, we established two novel xenogeneic (xeno)-GVHD models. Human CD4+ or CD8+ T cells were purified from peripheral blood and were transplanted into immunodeficient NOD/Shi-scid IL2rgnull (NOG) mice. Human CD8+ T cells did not induce major GVHD symptoms in conventional NOG mice. However, CD8+ T cells immediately proliferated and induced severe GVHD when transferred into NOG mice together with at least 0.5 × 106 CD4+ T cells or into NOG human interleukin (IL)-2 transgenic mice. Human CD4+ T cell-transplanted NOG mice developed skin inflammations including alopecia, epidermal hyperplasia, and neutrophilia. Pathogenic T helper (Th)17 cells accumulated in the skin of CD4+ T cell-transplanted NOG mice. Further, an anti-human IL-17 antibody (secukinumab) significantly suppressed these skin pathologies. These results indicate that pathogenic human Th17 cells induce cutaneous GVHD via IL-17-dependent pathways. This study provides fundamental insights into the pathogenesis of xeno-GVHD, and these humanized mouse models may be useful as preclinical tools for the prevention of GVHD.
Collapse
Affiliation(s)
- R Ito
- Central Institute for Experimental Animals, Kawasaki, Kanagawa, Japan
| | - I Katano
- Central Institute for Experimental Animals, Kawasaki, Kanagawa, Japan
| | - K Kawai
- Central Institute for Experimental Animals, Kawasaki, Kanagawa, Japan
| | - M Yagoto
- Central Institute for Experimental Animals, Kawasaki, Kanagawa, Japan
| | - T Takahashi
- Central Institute for Experimental Animals, Kawasaki, Kanagawa, Japan
| | - Y Ka
- Central Institute for Experimental Animals, Kawasaki, Kanagawa, Japan
| | - T Ogura
- Central Institute for Experimental Animals, Kawasaki, Kanagawa, Japan
| | - R Takahashi
- Central Institute for Experimental Animals, Kawasaki, Kanagawa, Japan
| | - M Ito
- Central Institute for Experimental Animals, Kawasaki, Kanagawa, Japan
| |
Collapse
|
41
|
Cooke KR, Luznik L, Sarantopoulos S, Hakim FT, Jagasia M, Fowler DH, van den Brink MRM, Hansen JA, Parkman R, Miklos DB, Martin PJ, Paczesny S, Vogelsang G, Pavletic S, Ritz J, Schultz KR, Blazar BR. The Biology of Chronic Graft-versus-Host Disease: A Task Force Report from the National Institutes of Health Consensus Development Project on Criteria for Clinical Trials in Chronic Graft-versus-Host Disease. Biol Blood Marrow Transplant 2017; 23:211-234. [PMID: 27713092 PMCID: PMC6020045 DOI: 10.1016/j.bbmt.2016.09.023] [Citation(s) in RCA: 280] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2016] [Accepted: 09/30/2016] [Indexed: 12/12/2022]
Abstract
Chronic graft-versus-host disease (GVHD) is the leading cause of late, nonrelapse mortality and disability in allogeneic hematopoietic cell transplantation recipients and a major obstacle to improving outcomes. The biology of chronic GVHD remains enigmatic, but understanding the underpinnings of the immunologic mechanisms responsible for the initiation and progression of disease is fundamental to developing effective prevention and treatment strategies. The goals of this task force review are as follows: This document is intended as a review of our understanding of chronic GVHD biology and therapies resulting from preclinical studies, and as a platform for developing innovative clinical strategies to prevent and treat chronic GVHD.
Collapse
Affiliation(s)
- Kenneth R Cooke
- Department of Oncology, Sidney Kimmel Cancer Center at Johns Hopkins Hospital, Baltimore, Maryland.
| | - Leo Luznik
- Department of Oncology, Sidney Kimmel Cancer Center at Johns Hopkins Hospital, Baltimore, Maryland
| | - Stefanie Sarantopoulos
- Division of Hematological Malignancies and Cellular Therapy, Department of Immunology and Duke Cancer Institute, Duke University, Durham, North Carolina
| | - Frances T Hakim
- Experimental Transplantation and Immunology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Madan Jagasia
- Division of Hematology-Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Daniel H Fowler
- Experimental Transplantation and Immunology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Marcel R M van den Brink
- Departments of Immunology and Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - John A Hansen
- Division of Clinical Research, Fred Hutchinson Cancer Research Center, Department of Medicine, University of Washington, Seattle, Washington
| | - Robertson Parkman
- Division of Pediatric Stem Cell Transplantation and Regenerative Medicine, Stanford University, Palo Alto, California
| | - David B Miklos
- Division of Blood and Marrow Transplantation, Stanford University, Palo Alto, California
| | - Paul J Martin
- Division of Clinical Research, Fred Hutchinson Cancer Research Center, Department of Medicine, University of Washington, Seattle, Washington
| | - Sophie Paczesny
- Departments of Pediatrics and Immunology, Wells Center for Pediatric Research, Melvin and Bren Simon Cancer Center, Indiana University School of Medicine, Indianapolis, Indiana
| | - Georgia Vogelsang
- Department of Oncology, Sidney Kimmel Cancer Center at Johns Hopkins Hospital, Baltimore, Maryland
| | - Steven Pavletic
- Experimental Transplantation and Immunology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Jerome Ritz
- Division of Hematologic Malignancies, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Kirk R Schultz
- Michael Cuccione Childhood Cancer Research Program, Department of Pediatrics, BC Children's Hospital, University of British Columbia, Vancouver, British Columbia, Canada.
| | - Bruce R Blazar
- Masonic Cancer Center and Department of Pediatrics, Division of Blood and Marrow Transplantation, University of Minnesota, Minneapolis, Minnesota.
| |
Collapse
|
42
|
Fujii U, Miyahara N, Taniguchi A, Oda N, Morichika D, Murakami E, Nakayama H, Waseda K, Kataoka M, Kakuta H, Tanimoto M, Kanehiro A. Effect of a retinoid X receptor partial agonist on airway inflammation and hyperresponsiveness in a murine model of asthma. Respir Res 2017; 18:23. [PMID: 28114934 PMCID: PMC5260083 DOI: 10.1186/s12931-017-0507-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Accepted: 01/13/2017] [Indexed: 12/21/2022] Open
Abstract
Background Retinoid X receptors (RXRs) are members of the nuclear receptor (NR) superfamily that mediate signaling by 9-cis retinoic acid, a vitamin A (retinol) derivative. RXRs play key roles not only as homodimers but also as heterodimeric partners—e.g., retinoic acid receptors (RARs), vitamin D receptors (VDRs), liver X receptors (LXRs), and peroxisome proliferator-activated receptors (PPARs). The NR family was recently associated with allergic diseases, but the role of RXRs in allergen-induced airway responses is not well defined. The goal of this study is to elucidate the role of RXRs in asthma pathogenesis and the potency of RXR partial agonist in the treatment of allergic airway inflammation and airway hyperresponsiveness using a murine model of asthma. Methods We investigated the effect of a novel RXR partial agonist (NEt-4IB) on the development of allergic airway inflammation and airway hyperresponsiveness (AHR) in a murine model of asthma. Balb/c mice were sensitized (days 0 and 14) and challenged (days 28–30) with ovalbumin (OVA), and airway inflammation and airway responses were monitored 48 h after the last OVA challenge. NEt-4IB was administered orally on days 25 to 32. Results Oral administration of NEt-4IB significantly suppressed AHR and inflammatory cell accumulation in the airways and attenuated the levels of TNF-α in the lung and IL-5, IL-13 and NO levels in bronchoalveolar lavage (BAL) fluid and the number of periodic acid Schiff (PAS)-positive goblet cells in lung tissue. Treatment with NEt-4IB also significantly suppressed NF-κB expression. Conclusion These data suggest that RXRs may be of crucial importance in the mechanism of allergic asthma and that the novel RXR partial agonist NEt-4IB may be a promising candidate for the treatment of allergic airway inflammation and airway hyperresponsiveness in a model of allergic asthma.
Collapse
Affiliation(s)
- Utako Fujii
- Department of Hematology, Oncology, Allergy and Respiratory Medicine, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama, 700-8558, Japan
| | - Nobuaki Miyahara
- Department of Hematology, Oncology, Allergy and Respiratory Medicine, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama, 700-8558, Japan
| | - Akihiko Taniguchi
- Department of Hematology, Oncology, Allergy and Respiratory Medicine, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama, 700-8558, Japan
| | - Naohiro Oda
- Department of Hematology, Oncology, Allergy and Respiratory Medicine, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama, 700-8558, Japan
| | - Daisuke Morichika
- Department of Hematology, Oncology, Allergy and Respiratory Medicine, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama, 700-8558, Japan
| | - Etsuko Murakami
- Department of Hematology, Oncology, Allergy and Respiratory Medicine, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama, 700-8558, Japan
| | - Hikari Nakayama
- Department of Hematology, Oncology, Allergy and Respiratory Medicine, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama, 700-8558, Japan
| | - Koichi Waseda
- Department of Hematology, Oncology, Allergy and Respiratory Medicine, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama, 700-8558, Japan
| | - Mikio Kataoka
- Department of Hematology, Oncology, Allergy and Respiratory Medicine, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama, 700-8558, Japan
| | - Hiroki Kakuta
- Division of Pharmaceutical Sciences, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, 1-1-1 Tsushima-naka, Kita-ku, Okayama, 700-8530, Japan
| | - Mitsune Tanimoto
- Department of Hematology, Oncology, Allergy and Respiratory Medicine, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama, 700-8558, Japan
| | - Arihiko Kanehiro
- Department of Hematology, Oncology, Allergy and Respiratory Medicine, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama, 700-8558, Japan.
| |
Collapse
|
43
|
Presland RB. Biology of chronic graft- vs-host disease: Immune mechanisms and progress in biomarker discovery. World J Transplant 2016; 6:608-619. [PMID: 28058210 PMCID: PMC5175218 DOI: 10.5500/wjt.v6.i4.608] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2016] [Revised: 07/02/2016] [Accepted: 09/18/2016] [Indexed: 02/05/2023] Open
Abstract
Chronic graft-vs-host disease (cGVHD) is the leading cause of long-term morbidity and mortality following allogeneic hematopoietic stem cell transplantation. It presents as a chronic inflammatory and sclerotic autoimmune-like condition that most frequently affects the skin, oral mucosa, liver, eyes and gastrointestinal tract. Both clinical and animal studies have shown that multiple T cell subsets including Th1, Th2, Th17, T follicular helper cells and regulatory T-cells play some role in cGVHD development and progression; B cells also play an important role in the disease including the production of antibodies to HY and nuclear antigens that can cause serious tissue damage. An array of cytokines and chemokines produced by different types of immune cells also mediate tissue inflammation and damage of cGVHD target tissues such as the skin and oral cavity. Many of these same immune regulators have been studied as candidate cGVHD biomarkers. Recent studies suggest that some of these biomarkers may be useful for determining disease prognosis and planning long-term clinical follow-up of cGVHD patients.
Collapse
|
44
|
Hakim FT, Memon S, Jin P, Imanguli MM, Wang H, Rehman N, Yan XY, Rose J, Mays JW, Dhamala S, Kapoor V, Telford W, Dickinson J, Davis S, Halverson D, Naik HB, Baird K, Fowler D, Stroncek D, Cowen EW, Pavletic SZ, Gress RE. Upregulation of IFN-Inducible and Damage-Response Pathways in Chronic Graft-versus-Host Disease. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2016; 197:3490-3503. [PMID: 27694491 PMCID: PMC5101132 DOI: 10.4049/jimmunol.1601054] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Accepted: 09/06/2016] [Indexed: 12/29/2022]
Abstract
Although chronic graft-versus-host disease (CGVHD) is the primary nonrelapse complication of allogeneic transplantation, understanding of its pathogenesis is limited. To identify the main operant pathways across the spectrum of CGVHD, we analyzed gene expression in circulating monocytes, chosen as in situ systemic reporter cells. Microarrays identified two interrelated pathways: 1) IFN-inducible genes, and 2) innate receptors for cellular damage. Corroborating these with multiplex RNA quantitation, we found that multiple IFN-inducible genes (affecting lymphocyte trafficking, differentiation, and Ag presentation) were concurrently upregulated in CGVHD monocytes compared with normal subjects and non-CGVHD control patients. IFN-inducible chemokines were elevated in both lichenoid and sclerotic CGHVD plasma and were linked to CXCR3+ lymphocyte trafficking. Furthermore, the levels of the IFN-inducible genes CXCL10 and TNFSF13B (BAFF) were correlated at both the gene and the plasma levels, implicating IFN induction as a factor in elevated BAFF levels in CGVHD. In the second pathway, damage-/pathogen-associated molecular pattern receptor genes capable of inducing type I IFN were upregulated. Type I IFN-inducible MxA was expressed in proportion to CGVHD activity in skin, mucosa, and glands, and expression of TLR7 and DDX58 receptor genes correlated with upregulation of type I IFN-inducible genes in monocytes. Finally, in serial analyses after transplant, IFN-inducible and damage-response genes were upregulated in monocytes at CGVHD onset and declined upon therapy and resolution in both lichenoid and sclerotic CGVHD patients. This interlocking analysis of IFN-inducible genes, plasma analytes, and tissue immunohistochemistry strongly supports a unifying hypothesis of induction of IFN by innate response to cellular damage as a mechanism for initiation and persistence of CGVHD.
Collapse
Affiliation(s)
- Frances T Hakim
- Experimental Transplantation and Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892;
| | - Sarfraz Memon
- Experimental Transplantation and Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - Ping Jin
- Department of Transfusion Medicine, Clinical Center, National Institutes of Health, Bethesda, MD 20892
| | - Matin M Imanguli
- Experimental Transplantation and Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - Huan Wang
- Department of Transfusion Medicine, Clinical Center, National Institutes of Health, Bethesda, MD 20892
| | - Najibah Rehman
- Experimental Transplantation and Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - Xiao-Yi Yan
- Experimental Transplantation and Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - Jeremy Rose
- Experimental Transplantation and Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - Jacqueline W Mays
- Clinical Research Core, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892
| | - Susan Dhamala
- Experimental Transplantation and Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - Veena Kapoor
- Experimental Transplantation and Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - William Telford
- Experimental Transplantation and Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - John Dickinson
- Experimental Transplantation and Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - Sean Davis
- Cancer Genetics Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - David Halverson
- Experimental Transplantation and Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - Haley B Naik
- Dermatology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892; and
| | - Kristin Baird
- Pediatric Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - Daniel Fowler
- Experimental Transplantation and Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - David Stroncek
- Department of Transfusion Medicine, Clinical Center, National Institutes of Health, Bethesda, MD 20892
| | - Edward W Cowen
- Dermatology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892; and
| | - Steven Z Pavletic
- Experimental Transplantation and Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - Ronald E Gress
- Experimental Transplantation and Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| |
Collapse
|
45
|
Fujii U, Miyahara N, Taniguchi A, Waseda K, Morichika D, Kurimoto E, Koga H, Kataoka M, Gelfand EW, Cua DJ, Yoshimura A, Tanimoto M, Kanehiro A. IL-23 Is Essential for the Development of Elastase-Induced Pulmonary Inflammation and Emphysema. Am J Respir Cell Mol Biol 2016; 55:697-707. [DOI: 10.1165/rcmb.2016-0015oc] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
|
46
|
Magenau J, Runaas L, Reddy P. Advances in understanding the pathogenesis of graft-versus-host disease. Br J Haematol 2016; 173:190-205. [PMID: 27019012 DOI: 10.1111/bjh.13959] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Accepted: 12/15/2015] [Indexed: 01/24/2023]
Abstract
Allogeneic haematopoietic stem cell transplantation (HCT) is a potent immunotherapy with curative potential for several haematological disorders. Overcoming the immunological barrier of acute graft-versus-host disease (GVHD) remains a fundamental impediment to expanding the efficacy of HCT. GVHD reflects a complex pathological interaction between the innate and adaptive immune systems of the host and donor. Over the past decade there has been a tremendous advancement in our understanding of the cellular and molecular underpinnings of this devastating disease. In this review, we cover several recently appreciated facets of GVHD pathogenesis including novel extracellular mediators of inflammation, immune subsets, intracellular signal transduction, post-translation modifications and epigenetic regulation. We begin to develop general themes regarding the immunological pathways in GVHD pathogenesis, discuss critical outstanding questions, and explore new avenues for GVHD treatment and prevention.
Collapse
Affiliation(s)
- John Magenau
- Blood and Marrow Transplant Program, Division of Hematology/Oncology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Lyndsey Runaas
- Blood and Marrow Transplant Program, Division of Hematology/Oncology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Pavan Reddy
- Blood and Marrow Transplant Program, Division of Hematology/Oncology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
47
|
Villa NY, Rahman MM, McFadden G, Cogle CR. Therapeutics for Graft-versus-Host Disease: From Conventional Therapies to Novel Virotherapeutic Strategies. Viruses 2016; 8:85. [PMID: 27011200 PMCID: PMC4810275 DOI: 10.3390/v8030085] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2015] [Revised: 03/09/2016] [Accepted: 03/09/2016] [Indexed: 02/06/2023] Open
Abstract
Allogeneic hematopoietic stem cell transplantation (allo-HSCT) has a curative potential for many hematologic malignancies and blood diseases. However, the success of allo-HSCT is limited by graft-versus-host disease (GVHD), an immunological syndrome that involves inflammation and tissue damage mediated by donor lymphocytes. Despite immune suppression, GVHD is highly incident even after allo-HSCT using human leukocyte antigen (HLA)-matched donors. Therefore, alternative and more effective therapies are needed to prevent or control GVHD while preserving the beneficial graft-versus-cancer (GVC) effects against residual disease. Among novel therapeutics for GVHD, oncolytic viruses such as myxoma virus (MYXV) are receiving increased attention due to their dual role in controlling GVHD while preserving or augmenting GVC. This review focuses on the molecular basis of GVHD, as well as state-of-the-art advances in developing novel therapies to prevent or control GVHD while minimizing impact on GVC. Recent literature regarding conventional and the emerging therapies are summarized, with special emphasis on virotherapy to prevent GVHD. Recent advances using preclinical models with oncolytic viruses such as MYXV to ameliorate the deleterious consequences of GVHD, while maintaining or improving the anti-cancer benefits of GVC will be reviewed.
Collapse
Affiliation(s)
- Nancy Y Villa
- Division of Hematology and Oncology, Department of Medicine, University of Florida, Gainesville, FL 32610, USA.
| | - Masmudur M Rahman
- Department of Molecular Genetics and Microbiology, University of Florida, Gainesville, FL 32610, USA.
| | - Grant McFadden
- Department of Molecular Genetics and Microbiology, University of Florida, Gainesville, FL 32610, USA.
| | - Christopher R Cogle
- Division of Hematology and Oncology, Department of Medicine, University of Florida, Gainesville, FL 32610, USA.
| |
Collapse
|
48
|
Preclinical models of acute and chronic graft-versus-host disease: how predictive are they for a successful clinical translation? Blood 2016; 127:3117-26. [PMID: 26994149 DOI: 10.1182/blood-2016-02-699082] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Accepted: 03/15/2016] [Indexed: 02/07/2023] Open
Abstract
Despite major advances in recent years, graft-versus-host disease (GVHD) remains a major life-threatening complication of allogeneic hematopoietic cell transplantation (allo-HCT). To improve our therapeutic armory against GVHD, preclinical evidence is most frequently generated in mouse and large animal models of GVHD. However, because every model has shortcomings, it is important to understand how predictive the different models are and why certain findings in these models could not be translated into the clinic. Weaknesses of the animal GVHD models include the irradiation only-based conditioning regimen, the homogenous donor/recipient genetics in mice, canine or non-human primates (NHP), anatomic site of T cells used for transfer in mice, the homogenous microbial environment in mice housed under specific pathogen-free conditions, and the lack of pharmacologic GVHD prevention in control groups. Despite these major differences toward clinical allo-HCT, findings generated in animal models of GVHD have led to the current gold standards for GVHD prophylaxis and therapy. The homogenous nature of the preclinical models allows for reproducibility, which is key for the characterization of the role of a new cytokine, chemokine, transcription factor, microRNA, kinase, or immune cell population in the context of GVHD. Therefore, when carefully balancing reasons to apply small and large animal models, it becomes evident that they are valuable tools to generate preclinical hypotheses, which then have to be rigorously evaluated in the clinical setting. In this study, we discuss several clinical approaches that were motivated by preclinical evidence, novel NHP models and their advantages, and highlight the recent advances in understanding the pathophysiology of GVHD.
Collapse
|
49
|
Malard F, Gaugler B, Lamarthee B, Mohty M. Translational opportunities for targeting the Th17 axis in acute graft-vs.-host disease. Mucosal Immunol 2016; 9:299-308. [PMID: 26813345 DOI: 10.1038/mi.2015.143] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Accepted: 12/02/2015] [Indexed: 02/04/2023]
Abstract
Allogeneic stem cell transplantation (allo-SCT) is a curative therapy for different life-threatening malignant and non-malignant hematologic disorders. Acute graft-vs.-host disease (aGVHD) and particularly gastrointestinal aGVHD remains a major source of morbidity and mortality following allo-SCT, which limits the use of this treatment in a broader spectrum of patients. Better understanding of aGVHD pathophysiology is indispensable to identify new therapeutic targets for aGVHD prevention and therapy. Growing amount of data suggest a role for T helper (Th)17 cells in aGVHD pathophysiology. In this review, we will discuss the current knowledge in this area in animal models and in humans. We will then describe new potential treatments for aGVHD along the Th17 axis.
Collapse
Affiliation(s)
- F Malard
- Université Pierre et Marie Curie, Paris, France.,Centre de recherche Saint-Antoine, INSERM, UMRs 938, Paris, France.,Service d'Hématologie Clinique et de Thérapie Cellulaire, Hôpital Saint Antoine, APHP, Paris, France.,INSERM, UMR 1064-Center for Research in Transplantation and Immunology, Nantes, F44093 France
| | - B Gaugler
- Université Pierre et Marie Curie, Paris, France.,Centre de recherche Saint-Antoine, INSERM, UMRs 938, Paris, France
| | - B Lamarthee
- Université Pierre et Marie Curie, Paris, France.,Centre de recherche Saint-Antoine, INSERM, UMRs 938, Paris, France
| | - M Mohty
- Université Pierre et Marie Curie, Paris, France.,Centre de recherche Saint-Antoine, INSERM, UMRs 938, Paris, France.,Service d'Hématologie Clinique et de Thérapie Cellulaire, Hôpital Saint Antoine, APHP, Paris, France
| |
Collapse
|
50
|
Li W, Li H, Zhang M, Wang M, Zhong Y, Wu H, Yang Y, Morel L, Wei Q. Quercitrin ameliorates the development of systemic lupus erythematosus-like disease in a chronic graft-versus-host murine model. Am J Physiol Renal Physiol 2016; 311:F217-26. [PMID: 26911849 DOI: 10.1152/ajprenal.00249.2015] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Accepted: 02/17/2016] [Indexed: 12/22/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is a serious disorder of immune regulation characterized by overproduction of autoantibodies, lupus nephritis, CD4+ T cell aberrant activation, and immune complex-mediated inflammation. The chronic graft vs. host disease (cGVHD) mouse model is a well-established model of SLE. Quercitrin is a natural compound found in Tartary buckwheat with a potential anti-inflammatory effect that is used to treat heart and vascular conditions. In our previous study, we determined that quercitrin is an immunosuppressant with beneficial effects in mouse models of immune diseases. We hypothesized that quercitrin could prevent lupus nephritis in the cGVHD mouse model by decreasing the production of autoantibodies and inflammatory cytokines, and reducing immune cell activation. cGVHD was induced by injecting DBA/2 spleen cells into the tail vein of BDF1 mice. The cGVHD mice exhibited significant proteinuria, which is a marker of nephritis. Quercitrin decreased the number of serum antibodies, CD4+ T cell activation, as well as the expression levels of T-bet, GATA-3, and selected cytokines. Moreover, quercitrin treatment decreased the expression of inflammatory genes and cytokines in the kidney, as well as in peritoneal macrophages. In addition, quercitrin inhibited LPS-induced cytokines as well as the phosphorylation of ERK, p38 MAPK, and JNK in Raw264.7 cells. Overall, quercitrin ameliorated the symptoms of lupus nephritis in the cGVHD mouse model, which may be due to the inhibition of CD4 T cell activation and anti-inflammatory effects on macrophages.
Collapse
Affiliation(s)
- Wei Li
- Beijing Key Laboratory of Gene Engineering and Biotechnology, Department of Biochemistry and Molecular Biology, Beijing Normal University, Beijing, China
| | - Hu Li
- Beijing Key Laboratory of Gene Engineering and Biotechnology, Department of Biochemistry and Molecular Biology, Beijing Normal University, Beijing, China
| | - Mu Zhang
- Beijing Key Laboratory of Gene Engineering and Biotechnology, Department of Biochemistry and Molecular Biology, Beijing Normal University, Beijing, China
| | - Mengqi Wang
- Beijing Key Laboratory of Gene Engineering and Biotechnology, Department of Biochemistry and Molecular Biology, Beijing Normal University, Beijing, China
| | - Youxiu Zhong
- Beijing Key Laboratory of Gene Engineering and Biotechnology, Department of Biochemistry and Molecular Biology, Beijing Normal University, Beijing, China
| | - Hezhen Wu
- College of Pharmacy, Hubei University of Chinese Medicine, Wuhan, China; and
| | - Yanfang Yang
- College of Pharmacy, Hubei University of Chinese Medicine, Wuhan, China; and
| | - Laurence Morel
- Department of Pathology, University of Florida, Gainesville, Florida
| | - Qun Wei
- Beijing Key Laboratory of Gene Engineering and Biotechnology, Department of Biochemistry and Molecular Biology, Beijing Normal University, Beijing, China;
| |
Collapse
|