1
|
Bernard-Bloch R, Lebrault E, Li X, Sutra Del Galy A, Garcia A, Doliger C, Parietti V, Legembre P, Socié G, Karray S. Ambivalent role of FasL in murine acute graft-versus-host-disease. J Leukoc Biol 2023; 114:205-211. [PMID: 37013690 DOI: 10.1093/jleuko/qiad040] [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: 08/25/2022] [Revised: 02/28/2023] [Accepted: 03/29/2023] [Indexed: 04/05/2023] Open
Abstract
Fas ligand is increased in several immune-mediated diseases, including acute graft-versus-host disease, a donor cell-mediated disorder post-hematopoietic stem cell transplantation. In this disease, Fas ligand is involved in T-cell-mediated damage to host tissues. However, the role of its expression on donor non-T cells has, so far, never been addressed. Using a well-established CD4- and CD8-mediated graft-versus-host disease murine model, we found that precocious gut damage and mice mortality are increased with a graft of donor T- and B-depleted bone marrow cells devoid of Fas ligand as compared with their wild-type counterparts. Interestingly, serum levels of both soluble Fas ligand and IL-18 are drastically reduced in the recipients of Fas ligand-deficient grafts, indicating that soluble Fas ligand stems from donor bone marrow-derived cells. In addition, the correlation between the concentrations of these 2 cytokines suggests that IL-18 production arises through a soluble Fas ligand-driven mechanism. These data highlight the importance of Fas ligand-dependent production in IL-18 production and in mitigating acute graft-versus-host disease. Overall, our data reveal the functional duality of Fas ligand according to its source.
Collapse
Affiliation(s)
- Robin Bernard-Bloch
- Département d'Immunologie, Université de Paris Cité, INSERM U976, Institut de Recherche Saint Louis, 1 avenue Claude Vellefaux, 75010 Paris, France
| | - Eden Lebrault
- Département of CRIBL, UMR, CNRS 7276, INSERM 1262, Université de Limoges, 2 rue du Docteur Marcland, 87025 Limoges, France
| | - Xiaofan Li
- Département d'Immunologie, Université de Paris Cité, INSERM U976, Institut de Recherche Saint Louis, 1 avenue Claude Vellefaux, 75010 Paris, France
| | - Aurélien Sutra Del Galy
- AP-HP, Département d'hématologie-greffe, Hôpital Saint Louis, 1 avenue Claude Villefaux, 75010 Paris, France
| | - Arlette Garcia
- Département d'Immunologie, Université de Paris Cité, INSERM U976, Institut de Recherche Saint Louis, 1 avenue Claude Vellefaux, 75010 Paris, France
| | - Christelle Doliger
- Université de Paris Cité, Institut de Recherche Saint Louis, Département Technologique, 1 avenue Claude Villefaux, 75010 Paris, France
| | - Véronique Parietti
- Département Expérimentation Animale, Université de Paris Cité, Institut de Recherche Saint Louis, 1 avenue Claude Villefaux, 75010 Paris, France
| | - Patrick Legembre
- Département of CRIBL, UMR, CNRS 7276, INSERM 1262, Université de Limoges, 2 rue du Docteur Marcland, 87025 Limoges, France
| | - Gérard Socié
- Département d'Immunologie, Université de Paris Cité, INSERM U976, Institut de Recherche Saint Louis, 1 avenue Claude Vellefaux, 75010 Paris, France
- AP-HP, Département d'hématologie-greffe, Hôpital Saint Louis, 1 avenue Claude Villefaux, 75010 Paris, France
| | - Saoussen Karray
- Département d'Immunologie, Université de Paris Cité, INSERM U976, Institut de Recherche Saint Louis, 1 avenue Claude Vellefaux, 75010 Paris, France
- CNRS, Délégation Régionale 01, 7 rue Guy Môquet, 94800 Villejuif, France
| |
Collapse
|
2
|
Tollemar V, Garming Legert K, Sugars RV. Perspectives on oral chronic graft-versus-host disease from immunobiology to morbid diagnoses. Front Immunol 2023; 14:1151493. [PMID: 37449200 PMCID: PMC10338056 DOI: 10.3389/fimmu.2023.1151493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Accepted: 06/12/2023] [Indexed: 07/18/2023] Open
Abstract
Chronic Graft-versus-Host Disease (cGVHD) is a major long-term complication, associated with morbidity and mortality in patients following allogenic hematopoietic cell transplantation (HCT) for immune hematopoietic disorders. The mouth is one of the most frequently affected organs after HCT (45-83%) and oral cGVHD, which may appear as the first visible sign. Manifestations present with mucosal lichenoid lesions, salivary gland dysfunction and limited oral aperture. Diagnosis of oral cGVHD severity is based on mucosal lesions with symptoms of sensitivity and pain and reduced oral intake. However, diagnostic difficulties arise due to subjective definitions and low specificity to cover the spectrum of oral cGVHD. In recent years there have been significant improvements in our understanding of the underlying oral cGVHD disease mechanisms. Drawing upon the current knowledge on the pathophysiology and biological phases of oral cGVHD, we address oral mucosa lichenoid and Sjogren's Syndrome-like sicca syndromes. We consider the response of alloreactive T-cells and macrophages to recipient tissues to drive the pathophysiological reactions and biological phases of acute inflammation (phase 1), chronic inflammation and dysregulated immunity (phase 2), and subsequent aberrant fibrotic healing (phase 3), which in time may be associated with an increased malignant transformation rate. When formulating treatment strategies, the pathophysiological spectrum of cGVHD is patient dependent and not every patient may progress chronologically through the biological stages. As such there remains a need to address and clarify personalized diagnostics and management to improve treatment descriptions. Within this review, we highlight the current state of the art knowledge on oral cGVHD pathophysiology and biological phases. We address knowledge gaps of oral cGVHD, with a view to facilitate clinical management and improve research quality on lichenoid biology and morbid forms of oral cGVHD.
Collapse
Affiliation(s)
| | | | - Rachael V. Sugars
- Division of Oral Diagnostics and Rehabilitation, Department of Dental Medicine, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
3
|
Liu H, Yu Z, Tang B, Miao S, Qin C, Li Y, Liang Z, Shi Y, Zhang Y, Wang Q, Yan M, Song Z, Ren H, Dong Y. LYG1 Deficiency Attenuates the Severity of Acute Graft-Versus-Host Disease via Skewing Allogeneic T Cells Polarization Towards Treg Cells. Front Immunol 2021; 12:647894. [PMID: 34262560 PMCID: PMC8273552 DOI: 10.3389/fimmu.2021.647894] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Accepted: 06/11/2021] [Indexed: 11/13/2022] Open
Abstract
Acute graft-versus-host disease (aGVHD) is a lethal complication after allogeneic hematopoietic stem cell transplantation. The mechanism involves the recognition of host antigens by donor-derived T cells which induces augmented response of alloreactive T cells. In this study, we characterized the role of a previously identified novel classical secretory protein with antitumor function-LYG1 (Lysozyme G-like 1), in aGVHD. LYG1 deficiency reduced the activation of CD4+ T cells and Th1 ratio, but increased Treg ratio in vitro by MLR assay. By using major MHC mismatched aGVHD model, LYG1 deficiency in donor T cells or CD4+ T cells attenuated aGVHD severity, inhibited CD4+ T cells activation and IFN-γ expression, promoted FoxP3 expression, suppressed CXCL9 and CXCL10 expression, restrained allogeneic CD4+ T cells infiltrating in target organs. The function of LYG1 in aGVHD was also confirmed using haploidentical transplant model. Furthermore, administration of recombinant human LYG1 protein intraperitoneally aggravated aGVHD by promoting IFN-γ production and inhibiting FoxP3 expression. The effect of rhLYG1 could partially be abrogated with the absence of IFN-γ. Furthermore, LYG1 deficiency in donor T cells preserved graft-versus-tumor response. In summary, our results indicate LYG1 regulates aGVHD by the alloreactivity of CD4+ T cells and the balance of Th1 and Treg differentiation of allogeneic CD4+ T cells, targeting LYG1 maybe a novel therapeutic strategy for preventing aGVHD.
Collapse
Affiliation(s)
- Huihui Liu
- Department of Hematology, Peking University First Hospital, Beijing, China
| | - Zhengyu Yu
- Department of Hematology, Peking University First Hospital, Beijing, China
| | - Bo Tang
- Department of Hematology, Peking University First Hospital, Beijing, China
| | - Shengchao Miao
- Department of Hematology, Peking University First Hospital, Beijing, China
| | - Chenchen Qin
- Department of Hematology, Peking University First Hospital, Beijing, China
| | - Yuan Li
- Department of Hematology, Peking University First Hospital, Beijing, China
| | - Zeyin Liang
- Department of Hematology, Peking University First Hospital, Beijing, China
| | - Yongjin Shi
- Department of Hematology, Peking University First Hospital, Beijing, China
| | - Yang Zhang
- Department of Hematology, Peking University First Hospital, Beijing, China
| | - Qingya Wang
- Department of Hematology, Peking University First Hospital, Beijing, China
| | - Miao Yan
- Department of Hematology, Peking University First Hospital, Beijing, China
| | - Zhengyang Song
- Department of Hematology, Peking University First Hospital, Beijing, China
| | - Hanyun Ren
- Department of Hematology, Peking University First Hospital, Beijing, China
| | - Yujun Dong
- Department of Hematology, Peking University First Hospital, Beijing, China
| |
Collapse
|
4
|
Wang YM, Khederzadeh S, Li SR, Otecko NO, Irwin DM, Thakur M, Ren XD, Wang MS, Wu DD, Zhang YP. Integrating Genomic and Transcriptomic Data to Reveal Genetic Mechanisms Underlying Piao Chicken Rumpless Trait. GENOMICS PROTEOMICS & BIOINFORMATICS 2021; 19:787-799. [PMID: 33631431 PMCID: PMC9170765 DOI: 10.1016/j.gpb.2020.06.019] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Revised: 04/14/2020] [Accepted: 06/10/2020] [Indexed: 11/19/2022]
Abstract
Piao chicken, a rare Chinese native poultry breed, lacks primary tail structures, such as pygostyle, caudal vertebra, uropygial gland, and tail feathers. So far, the molecular mechanisms underlying tail absence in this breed remain unclear. In this study, we comprehensively employed comparative transcriptomic and genomic analyses to unravel potential genetic underpinnings of rumplessness in Piao chicken. Our results reveal many biological factors involved in tail development and several genomic regions under strong positive selection in this breed. These regions contain candidate genes associated with rumplessness, including Irx4, Il18, Hspb2, and Cryab. Retrieval of quantitative trait loci (QTL) and gene functions implies that rumplessness might be consciously or unconsciously selected along with the high-yield traits in Piao chicken. We hypothesize that strong selection pressures on regulatory elements might lead to changes in gene activity in mesenchymal stem cells of the tail bud. The ectopic activity could eventually result in tail truncation by impeding differentiation and proliferation of the stem cells. Our study provides fundamental insights into early initiation and genetic basis of the rumpless phenotype in Piao chicken.
Collapse
Affiliation(s)
- Yun-Mei Wang
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China; Kunming College of Life Science, University of the Chinese Academy of Sciences, Kunming 650223, China; Center for Neurobiology and Brain Restoration, Skolkovo Institute of Science and Technology, Moscow 143026, Russia
| | - Saber Khederzadeh
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China; Kunming College of Life Science, University of the Chinese Academy of Sciences, Kunming 650223, China
| | - Shi-Rong Li
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China; Kunming College of Life Science, University of the Chinese Academy of Sciences, Kunming 650223, China
| | - Newton Otieno Otecko
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China; Kunming College of Life Science, University of the Chinese Academy of Sciences, Kunming 650223, China
| | - David M Irwin
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto M5S 1A8, Canada
| | - Mukesh Thakur
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China; Zoological Survey of India, Kolkata 700053, India
| | - Xiao-Die Ren
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China; Kunming College of Life Science, University of the Chinese Academy of Sciences, Kunming 650223, China
| | - Ming-Shan Wang
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China; Kunming College of Life Science, University of the Chinese Academy of Sciences, Kunming 650223, China.
| | - Dong-Dong Wu
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China; Kunming College of Life Science, University of the Chinese Academy of Sciences, Kunming 650223, China.
| | - Ya-Ping Zhang
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China; Kunming College of Life Science, University of the Chinese Academy of Sciences, Kunming 650223, China.
| |
Collapse
|
5
|
Hill GR, Koyama M. Cytokines and costimulation in acute graft-versus-host disease. Blood 2020; 136:418-428. [PMID: 32526028 PMCID: PMC7378458 DOI: 10.1182/blood.2019000952] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Accepted: 01/18/2020] [Indexed: 12/11/2022] Open
Abstract
Allogeneic hematopoietic stem cell transplantation (alloSCT) is an important curative therapy for high-risk hematological malignancies, but the development of severe and/or steroid-refractory acute graft-versus-host disease (aGVHD) remains a significant limitation to optimal outcomes. New approaches to prevent and treat aGVHD remain an unmet need that can be best addressed by understanding the complex disease pathophysiology. It is now clear that chemoradiotherapy used prior to alloSCT induces the release of endogenous alarmins (eg, HMGB-1, ATP, IL-1α, IL-33) from recipient tissue. Exogenous pathogen-derived molecules (eg, lipopolysaccharide, nucleic acids) also translocate from the gastrointestinal tract lumen. Together, these danger signals activate antigen-presenting cells (APCs) to efficiently present alloantigen to donor T cells while releasing cytokines (eg, interleukin-12 [IL-12], IL-23, IL-6, IL-27, IL-10, transforming growth factor-β) that expand and differentiate both pathogenic and regulatory donor T cells. Concurrent costimulatory signals at the APC-T-cell interface (eg, CD80/CD86-CD28, CD40-CD40L, OX40L-OX40, CD155/CD112-DNAM-1) and subsequent coinhibitory signals (eg, CD80/CD86-CTLA4, PDL1/2-PD1, CD155/CD112-TIGIT) are critical to the acquisition of effector T-cell function and ensuing secretion of pathogenic cytokines (eg, IL-17, interferon-γ, tissue necrosis factor, granulocyte-macrophage colony-stimulating factor) and cytolytic degranulation pathway effectors (eg, perforin/granzyme). This review focuses on the combination of cytokine and costimulatory networks at the T-cell surface that culminates in effector function and subsequent aGVHD in target tissue. Together, these pathways now represent robust and clinically tractable targets for preventing the initiation of deleterious immunity after alloSCT.
Collapse
Affiliation(s)
- Geoffrey R Hill
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA; and
- Division of Medical Oncology, Department of Medicine, University of Washington, Seattle, WA
| | - Motoko Koyama
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA; and
| |
Collapse
|
6
|
Danger-associated extracellular ATP counters MDSC therapeutic efficacy in acute GVHD. Blood 2020; 134:1670-1682. [PMID: 31533918 DOI: 10.1182/blood.2019001950] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Accepted: 08/24/2019] [Indexed: 02/07/2023] Open
Abstract
Myeloid-derived suppressor cells (MDSCs) can subdue inflammation. In mice with acute graft-versus-host disease (GVHD), donor MDSC infusion enhances survival that is only partial and transient because of MDSC inflammasome activation early posttransfer, resulting in differentiation and loss of suppressor function. Here we demonstrate that conditioning regimen-induced adenosine triphosphate (ATP) release is a primary driver of MDSC dysfunction through ATP receptor (P2x7R) engagement and NLR pyrin family domain 3 (NLRP3) inflammasome activation. P2x7R or NLRP3 knockout (KO) donor MDSCs provided significantly higher survival than wild-type (WT) MDSCs. Although in vivo pharmacologic targeting of NLRP3 or P2x7R promoted recipient survival, indicating in vivo biologic effects, no synergistic survival advantage was seen when combined with MDSCs. Because activated inflammasomes release mature interleukin-1β (IL-1β), we expected that IL-1β KO donor MDSCs would be superior in subverting GVHD, but such MDSCs proved inferior relative to WT. IL-1β release and IL-1 receptor expression was required for optimal MDSC function, and exogenous IL-1β added to suppression assays that included MDSCs increased suppressor potency. These data indicate that prolonged systemic NLRP3 inflammasome inhibition and decreased IL-1β could diminish survival in GVHD. However, loss of inflammasome activation and IL-1β release restricted to MDSCs rather than systemic inhibition allowed non-MDSC IL-1β signaling, improving survival. Extracellular ATP catalysis with peritransplant apyrase administered into the peritoneum, the ATP release site, synergized with WT MDSCs, as did regulatory T-cell infusion, which we showed reduced but did not eliminate MDSC inflammasome activation, as assessed with a novel inflammasome reporter strain. These findings will inform future clinical using MDSCs to decrease alloresponses in inflammatory environments.
Collapse
|
7
|
|
8
|
Muraki M. Sensitization to cell death induced by soluble Fas ligand and agonistic antibodies with exogenous agents: A review. AIMS MEDICAL SCIENCE 2020. [DOI: 10.3934/medsci.2020011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
|
9
|
Ahmed Z, Vierling JM. Graft-Versus-Host Disease. LIVER IMMUNOLOGY 2020:551-582. [DOI: 10.1007/978-3-030-51709-0_34] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
10
|
Saha A, Taylor PA, Lees CJ, Panoskaltsis-Mortari A, Osborn MJ, Feser CJ, Thangavelu G, Melchinger W, Refaeli Y, Hill GR, Munn DH, Murphy WJ, Serody JS, Maillard I, Kreymborg K, van den Brink M, Dong C, Huang S, Zang X, Allison JP, Zeiser R, Blazar BR. Donor and host B7-H4 expression negatively regulates acute graft-versus-host disease lethality. JCI Insight 2019; 4:127716. [PMID: 31578305 DOI: 10.1172/jci.insight.127716] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2019] [Accepted: 08/23/2019] [Indexed: 12/30/2022] Open
Abstract
B7-H4 is a negative regulatory B7 family member. We investigated the role of host and donor B7-H4 in regulating acute graft-versus-host disease (GVHD). Allogeneic donor T cells infused into B7-H4-/- versus WT recipients markedly accelerated GVHD-induced lethality. Chimera studies pointed toward B7-H4 expression on host hematopoietic cells as more critical than parenchymal cells in controlling GVHD. Rapid mortality in B7-H4-/- recipients was associated with increased donor T cell expansion, gut T cell homing and loss of intestinal epithelial integrity, increased T effector function (proliferation, proinflammatory cytokines, cytolytic molecules), and reduced apoptosis. Higher metabolic demands of rapidly proliferating donor T cells in B7-H4-/- versus WT recipients required multiple metabolic pathways, increased extracellular acidification rates (ECARs) and oxygen consumption rates (OCRs), and increased expression of fuel substrate transporters. During GVHD, B7-H4 expression was upregulated on allogeneic WT donor T cells. B7-H4-/- donor T cells given to WT recipients increased GVHD mortality and had function and biological properties similar to WT T cells from allogeneic B7-H4-/- recipients. Graft-versus-leukemia responses were intact regardless as to whether B7-H4-/- mice were used as hosts or donors. Taken together, these data provide new insights into the negative regulatory processes that control GVHD and provide support for developing therapeutic strategies directed toward the B7-H4 pathway.
Collapse
Affiliation(s)
- Asim Saha
- Masonic Cancer Center and Department of Pediatrics, Division of Blood and Marrow Transplantation, University of Minnesota, Minneapolis, Minnesota, USA
| | - Patricia A Taylor
- Masonic Cancer Center and Department of Pediatrics, Division of Blood and Marrow Transplantation, University of Minnesota, Minneapolis, Minnesota, USA
| | - Christopher J Lees
- Masonic Cancer Center and Department of Pediatrics, Division of Blood and Marrow Transplantation, University of Minnesota, Minneapolis, Minnesota, USA
| | - Angela Panoskaltsis-Mortari
- Masonic Cancer Center and Department of Pediatrics, Division of Blood and Marrow Transplantation, University of Minnesota, Minneapolis, Minnesota, USA
| | - Mark J Osborn
- Masonic Cancer Center and Department of Pediatrics, Division of Blood and Marrow Transplantation, University of Minnesota, Minneapolis, Minnesota, USA
| | - Colby J Feser
- Masonic Cancer Center and Department of Pediatrics, Division of Blood and Marrow Transplantation, University of Minnesota, Minneapolis, Minnesota, USA
| | - Govindarajan Thangavelu
- Masonic Cancer Center and Department of Pediatrics, Division of Blood and Marrow Transplantation, University of Minnesota, Minneapolis, Minnesota, USA
| | - Wolfgang Melchinger
- Department of Hematology, Oncology, and Stem-Cell Transplantation, Freiburg University Medical Center, Albert Ludwigs University Freiburg, Freiburg, Germany
| | - Yosef Refaeli
- Department of Dermatology, University of Colorado, Aurora, Colorado, USA
| | - Geoffrey R Hill
- Fred Hutchinson Cancer Research Center, University of Washington, Seattle, Washington, USA
| | - David H Munn
- Department of Pediatrics, Georgia Health Sciences University, Augusta, Georgia, USA
| | - William J Murphy
- Department of Dermatology, UC Davis School of Medicine, Sacramento, California, USA
| | - Jonathan S Serody
- Department of Medicine, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Ivan Maillard
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Katharina Kreymborg
- Department of Immunology and Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Marcel van den Brink
- Department of Immunology and Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Chen Dong
- Institute for Immunology and School of Medicine, Tsinghua University, Beijing, China
| | - Shuyu Huang
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Xingxing Zang
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York, USA
| | - James P Allison
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Robert Zeiser
- Department of Hematology, Oncology, and Stem-Cell Transplantation, Freiburg University Medical Center, Albert Ludwigs University Freiburg, Freiburg, Germany
| | - Bruce R Blazar
- Masonic Cancer Center and Department of Pediatrics, Division of Blood and Marrow Transplantation, University of Minnesota, Minneapolis, Minnesota, USA
| |
Collapse
|
11
|
Matsuoka S, Hashimoto D, Kadowaki M, Ohigashi H, Hayase E, Yokoyama E, Hasegawa Y, Tateno T, Chen X, Aoyama K, Oka H, Onozawa M, Takeda K, Akashi K, Teshima T. Myeloid differentiation factor 88 signaling in donor T cells accelerates graft- versus-host disease. Haematologica 2019; 105:226-234. [PMID: 31048358 PMCID: PMC6939524 DOI: 10.3324/haematol.2018.203380] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Accepted: 04/30/2019] [Indexed: 01/30/2023] Open
Abstract
Myeloid differentiation factor 88 (MyD88) signaling has a crucial role in activation of both innate and adoptive immunity. MyD88 transduces signals via Toll-like receptor and interleukin-1 receptor superfamily to the NFκB pathway and inflammasome by forming a molecular complex with interleukin-1 receptor-associated kinase 4. The MyD88/interleukin-1 receptor-associated kinase 4 pathway plays an important role, not only in innate immunity, but also T-cell immunity; however, its role in donor T cells on the pathophysiology of graft-versus-host disease (GvHD) remains to be elucidated. We addressed this issue by using MyD88-deficient T cells in a mouse model of allogeneic hematopoietic stem cell transplantation (allo-SCT). While MyD88-deficient and wild-type T cells proliferated equivalently after transplantation, MyD88-deficient T cells demonstrated impaired survival and differentiation toward Th1, Tc1, and Th17, and induced less severe GvHD compared to wild-type T cells. Administration of interleukin-1 receptor-associated kinase 4 inhibitor PF-06650833 significantly ameliorated GvHD after allo-SCT. These results thus demonstrate that donor T-cell MyD88/interleukin-1 receptor-associated kinase 4 pathway is a novel therapeutic target against GvHD after allo-SCT.
Collapse
Affiliation(s)
- Satomi Matsuoka
- Department of Hematology, Faculty of Medicine, Hokkaido University, Sapporo
| | - Daigo Hashimoto
- Department of Hematology, Faculty of Medicine, Hokkaido University, Sapporo
| | - Masanori Kadowaki
- Department of Medicine and Biosystemic Science, Kyushu University Graduate School of Medical Sciences, Fukuoka
| | - Hiroyuki Ohigashi
- Department of Hematology, Faculty of Medicine, Hokkaido University, Sapporo
| | - Eiko Hayase
- Department of Hematology, Faculty of Medicine, Hokkaido University, Sapporo
| | - Emi Yokoyama
- Department of Hematology, Faculty of Medicine, Hokkaido University, Sapporo
| | - Yuta Hasegawa
- Department of Hematology, Faculty of Medicine, Hokkaido University, Sapporo
| | - Takahiro Tateno
- Department of Hematology, Faculty of Medicine, Hokkaido University, Sapporo
| | - Xuanzhong Chen
- Department of Hematology, Faculty of Medicine, Hokkaido University, Sapporo
| | - Kazutoshi Aoyama
- Department of Medicine and Biosystemic Science, Kyushu University Graduate School of Medical Sciences, Fukuoka
| | - Hideyo Oka
- Department of Medicine and Biosystemic Science, Kyushu University Graduate School of Medical Sciences, Fukuoka
| | - Masahiro Onozawa
- Department of Hematology, Faculty of Medicine, Hokkaido University, Sapporo
| | - Kiyoshi Takeda
- Department of Microbiology and Immunology, Graduate School of Medicine, WPI Immunology Frontier Research Center, Osaka University, Suita, Japan
| | - Koichi Akashi
- Department of Medicine and Biosystemic Science, Kyushu University Graduate School of Medical Sciences, Fukuoka
| | - Takanori Teshima
- Department of Hematology, Faculty of Medicine, Hokkaido University, Sapporo
| |
Collapse
|
12
|
Du W, Cao X. Cytotoxic Pathways in Allogeneic Hematopoietic Cell Transplantation. Front Immunol 2018; 9:2979. [PMID: 30631325 PMCID: PMC6315278 DOI: 10.3389/fimmu.2018.02979] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Accepted: 12/04/2018] [Indexed: 12/11/2022] Open
Abstract
Allogeneic hematopoietic cell transplantation (allo-HCT) is a potentially curative treatment for hematologic malignancies, and other hematologic and immunologic diseases. Donor-derived immune cells identify and attack cancer cells in the patient producing a unique graft-vs.-tumor (GVT) effect. This beneficial response renders allo-HCT one of the most effective forms of tumor immunotherapy. However, alloreactive donor T cells can damage normal host cells thereby causing graft-vs.-host disease (GVHD), which results in substantial morbidity and mortality. To date, GVHD remains as the major obstacle for more successful application of allo-HCT. Of special significance in this context are a number of cytotoxic pathways that are involved in GVHD and GVT response as well as donor cell engraftment. In this review, we summarize progress in the investigation of these cytotoxic pathways, including Fas/Fas ligand (FasL), perforin/granzyme, and cytokine pathways. Many studies have delineated their distinct operating mechanisms and how they are involved in the complex cellular interactions amongst donor, host, tumor, and infectious pathogens. Driven by progressing elucidation of their contributions in immune reconstitution and regulation, various interventional strategies targeting these pathways have entered translational stages with aims to improve the effectiveness of allo-HCT.
Collapse
Affiliation(s)
- Wei Du
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States
| | - Xuefang Cao
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States.,Department of Microbiology and Immunology, Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland, Baltimore, MD, United States
| |
Collapse
|
13
|
Kamel AM, Gameel A, Ebid GTA, Radwan ER, Mohammed Saleh MF, Abdelfattah R. The impact of cytokine gene polymorphisms on the outcome of HLA matched sibling hematopoietic stem cell transplantation. Cytokine 2018; 110:404-411. [PMID: 29801972 DOI: 10.1016/j.cyto.2018.05.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Revised: 04/10/2018] [Accepted: 05/07/2018] [Indexed: 01/01/2023]
Abstract
Graft-versus-host disease (GVHD) is the major complication of allogeneic hematopoietic stem cell transplantation (HSCT); cytokines are recognized as important mediators in its pathogenesis. In this study we investigated the role of cytokine gene polymorphisms on HSCT outcome. A total of 106 patient and 98 donors were genotyped by polymerase chain reaction sequence specific primers (PCR-SSP) based assay for tumor necrosis factor-α-308 (TNFα -308), interleukin (IL)-6-174, IL-10-1082, -819, -592, Interferon-γ+874 (IFN-γ+874), and transforming growth factor-β1 (TGF-β1) codon10 and 25 polymorphisms. Except one in each category, all patients and donors were TNFα -308 high producers and the majority were IL-6-174 high producers (93.3% and 90.8% respectively); a pattern that would alleviate any potential biological impact. Patient's IFN-γ+874 showed significant association with the development of chronic GVHD. Patients with IFN-γ +874 high producer showed an 8 folds likelihood to develop chronic GVHD as compared to those with IFN-γ+874 low producer predicted phenotype (95% CI: 1.59-40.2, p = 0.01). Patient's TGFβ1-codon 10 and 25 high/intermediate producers showed a lower incidence of acute GVHD though it did not achieve statistical significance (p = 0.065) on account of the low frequency of this genotype in our patients and donors (11.4 and 8.2% respectively). Other factors contributing to risk of GVHD included older age for both acute and chronic (p = 0.01 and 0.02 respectively) with age 24 as the best discriminating cutoff; CD34+ cell dose for chronic GVHD (p = 0.045) with a dose of 8 × 106/kg as the best discriminating cutoff; and conditioning regimen with Flu/Bu associated with the lowest incidence of acute GVHD (p = 0.003) and no impact on chronic GVHD. In conclusion the current study further indicates a potential role of some cytokine gene polymorphisms in the development of GVHD. The relative distribution of high and low producer genotypes in different ethnic groups contributes to their biological impact in different populations.
Collapse
Affiliation(s)
- Azza M Kamel
- Clinical Pathology Department, National Cancer Institute, Cairo University, Cairo, Egypt.
| | - Abdallah Gameel
- Clinical Pathology Department, National Cancer Institute, Cairo University, Cairo, Egypt
| | - Gamal T A Ebid
- Clinical Pathology Department, National Cancer Institute, Cairo University, Cairo, Egypt
| | - Eman R Radwan
- Clinical and Chemical Pathology Department, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Mostafa F Mohammed Saleh
- Clinical Hematology Unit, Internal Medicine Department, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Raafat Abdelfattah
- Medical Oncology Department, National Cancer Institute, Cairo University, Cairo, Egypt; Bone Marrow Transplantation Unit, Nasser Institute Hospital for Research and Treatment, Cairo, Egypt
| |
Collapse
|
14
|
The healthy donor profile of immunoregulatory soluble mediators is altered by stem cell mobilization and apheresis. Cytotherapy 2018; 20:740-754. [PMID: 29576502 DOI: 10.1016/j.jcyt.2018.02.366] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Revised: 02/08/2018] [Accepted: 02/11/2018] [Indexed: 11/23/2022]
Abstract
BACKGROUND Peripheral blood stem cells from healthy donors mobilized by granulocyte colony-stimulating factor (G-CSF) and thereafter harvested by leukapheresis are commonly used for allogeneic stem cell transplantation. METHODS Plasma levels of 38 soluble mediators (cytokines, soluble adhesion molecules, proteases, protease inhibitors) were analyzed in samples derived from healthy stem cell donors before G-CSF treatment and after 4 days, both immediately before and after leukapheresis. RESULTS Donors could be classified into two main subsets based on their plasma mediator profile before G-CSF treatment. Seventeen of 36 detectable mediators were significantly altered by G-CSF; generally an increase in mediator levels was seen, including pro-inflammatory cytokines, soluble adhesion molecules and proteases. Several leukocyte- and platelet-released mediators were increased during apheresis. Both plasma and graft mediator profiles were thus altered and showed correlations to graft concentrations of leukocytes and platelets; these concentrations were influenced by the apheresis device used. Finally, the mediator profile of the allotransplant recipients was altered by graft infusion, and based on their day +1 post-transplantation plasma profile our recipients could be divided into two major subsets that differed in overall survival. DISCUSSION G-CSF alters the short-term plasma mediator profile of healthy stem cell donors. These effects together with the leukocyte and platelet levels in the graft determine the mediator profile of the stem cell grafts. Graft infusion also alters the systemic mediator profile of the recipients, but further studies are required to clarify whether such graft-induced alterations have a prognostic impact.
Collapse
|
15
|
Histone deacetylase inhibitors suppress immature dendritic cell's migration by regulating CC chemokine receptor 1 expression. Cell Immunol 2017; 316:11-20. [PMID: 28341057 DOI: 10.1016/j.cellimm.2017.02.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Revised: 02/25/2017] [Accepted: 02/27/2017] [Indexed: 11/23/2022]
Abstract
The modulation of immature dendritic cells (iDCs), which involves processes such as phagocytosis, migration, and maturation, is considered a beneficial research theme. Once activated by an antigen, iDCs turn to mature DCs (mDCs) and migrate towards secondary lymphoid organs, and initiate the progress of cellular immunity. Histone deacetylase inhibitors (HDACis) are also thought to be a major modulator of cellular immunity. Herein, we demonstrate that HDACis (trichostatin-A (TSA), sodium butylate (SB), scriptaid (ST)) play a central regulatory role in the migratory activity of iDCs. In our results, TSA, SB and ST showed the potent inhibitory effect on the migration of iDCs stimulated by MIP-1α. The inhibitory activities of HDACis were found to be caused by reduction of CCR1 expression on the cell surface, and by the inhibition of phosphorylation of p38 mitogen-activated protein kinase (MAPK), extracellular signal-regulated kinases 1 and 2 (ERK 1/2), and c-Jun N-terminal kinase (JNK).
Collapse
|
16
|
Boieri M, Shah P, Jalapothu D, Zaitseva O, Walter L, Rolstad B, Naper C, Dressel R, Inngjerdingen M. Rat acute GvHD is Th1 driven and characterized by predominant donor CD4 + T-cell infiltration of skin and gut. Exp Hematol 2017; 50:33-45.e3. [PMID: 28238806 DOI: 10.1016/j.exphem.2017.02.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Revised: 01/05/2017] [Accepted: 02/13/2017] [Indexed: 01/07/2023]
Abstract
Acute graft-versus-host disease (aGvHD) remains a significant hurdle to successful treatment of many hematological disorders. The disease is caused by infiltration of alloactivated donor T cells primarily into the gastrointestinal tract and skin. Although cytotoxic T cells mediate direct cellular damage, T helper (Th) cells differentially secrete immunoregulatory cytokines. aGvHD is thought to be initiated primarily by Th1 cells but a consensus is still lacking regarding the role of Th2 and Th17 cells. The aim of this study was to determine the contribution of distinct T-cell subsets to aGvHD in the rat. aGvHD was induced by transplanting irradiated rats with T-cell-depleted major histocompatibility complex-mismatched bone marrow, followed 2 weeks later by donor lymphocyte infusion. Near complete donor T-cell chimerism was achieved in the blood and lymphatic tissues, in contrast to mixed chimerism in the skin and gut. Skin and gut donor T cells were predominantly CD4+, in contrast to T cells in the blood and lymphatic tissues. Genes associated with Th1 cells were upregulated in gut, liver, lung, and skin tissues affected by aGvHD. Increased serum levels of CXCL10 and IL-18 preceded symptoms of aGvHD, accompanied by increased responsiveness to CXCL10 by blood CD4+ T cells. No changes in the expression of Th2- or Th17-associated genes were observed, indicating that aGvHD in this rat model is mainly Th1 driven. The rat model of aGvHD could be instrumental for further investigations of donor T-cell subsets in the skin and gut and for exploring therapeutic options to ameliorate symptoms of aGvHD.
Collapse
Affiliation(s)
- Margherita Boieri
- Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway; Department of Immunology, Oslo University Hospital - Rikshospitalet, Oslo, Norway
| | - Pranali Shah
- Institute of Cellular and Molecular Immunology, University Medical Center Göttingen, Göttingen, Germany
| | - Dasaradha Jalapothu
- Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Olena Zaitseva
- Primate Genetics Laboratory, German Primate Center, Göttingen, Germany
| | - Lutz Walter
- Primate Genetics Laboratory, German Primate Center, Göttingen, Germany
| | - Bent Rolstad
- Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Christian Naper
- Department of Immunology, Oslo University Hospital - Rikshospitalet, Oslo, Norway
| | - Ralf Dressel
- Institute of Cellular and Molecular Immunology, University Medical Center Göttingen, Göttingen, Germany
| | - Marit Inngjerdingen
- Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway; Department of Immunology, Oslo University Hospital - Rikshospitalet, Oslo, Norway.
| |
Collapse
|
17
|
Meng XQ, Chen XH, Sahebally Z, Xu YN, Yin SY, Wu LM, Zheng SS. Cytokines are early diagnostic biomarkers of graft-versus-host disease in liver recipients. Hepatobiliary Pancreat Dis Int 2017; 16:45-51. [PMID: 28119258 DOI: 10.1016/s1499-3872(16)60157-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
BACKGROUND Graft-versus-host disease (GVHD) is associated with high mortality. Early diagnosis is essential to start treatment and to improve outcomes. Because of the inflammatory nature, we hypothesis that cytokine profile of patients with GVHD may serve as diagnostic markers. The present study was to evaluate the role of cytokine profile in the diagnosis of GVHD. METHODS An immunoassay was used to detect 29 cytokines simultaneously in the serum; the measuring sensitivity of all cytokines was pg/mL. Healthy subjects undergoing annual routine physical examinations served as negative controls; 23 patients with hepatocellular carcinoma (HCC) who had undergone liver transplantation (the LT group) comprised the test subjects. A total of 22 kidney recipients with biopsy-confirmed GVHD (the RT group) were included for comparison. HCC patients with radical surgery (the HCC group, n=22) served as positive control. The liver contents of the three cytokines, IL-2, IL-18, and IFN-gamma, were detected with immunohistochemistry. Serum granzyme B and perforin were measured by flow cytometry. RESULTS Of the 29 cytokines, the levels of IL-2 and IL-18 were increased significantly in liver recipients with GVHD compared with healthy controls (P<0.05). The serum levels of these three cytokines in the healthy, HCC, LT, and RT groups were IL-2: 0.90+/-0.02, 4.14+/-0.61, 5.10+/-0.89, and 1.48+/-0.09 pg/mL; IL-18: 80.61+/-9.35, 109.51+/-10.93, 230.11+/-12.92, and 61.98+/-7.88 pg/mL; IFN-gamma: 24.06+/-3.88, 24.84+/-3.21, 40.37+/-5.88, and 15.33+/-4.72 pg/mL, respectively. Immunohistochemistry showed that these 3 cytokines expressions in the liver were parallel to the serum cytokine. After standard anti-GVHD treatment, the expressions of IL-2, IL-18, and IFN-gamma were decreased in the liver (P<0.05). Serum granzyme B and perforin were significantly increased in GVHD patients (P<0.05). CONCLUSIONS IL-2, IL-18 and IFN-gamma were from liver and might serve as biomarkers for monitoring GVHD development and the effects of anti-GVHD treatment. Granzyme B and perforin may play a role in increasing IL-2, IL-18, and IFN-gamma levels in GVHD patients.
Collapse
Affiliation(s)
- Xue-Qin Meng
- Key Laboratory of Combined Multi-organ Transplantation, Ministry of Public Health, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, the First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou 310003, China.
| | | | | | | | | | | | | |
Collapse
|
18
|
Herzyk DJ, Bugelski PJ, Hart TK, Wier PJ. Preclinical Safety of Recombinant Human Interleukin-18. Toxicol Pathol 2016; 31:554-61. [PMID: 14692624 DOI: 10.1080/01926230390226681] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Recombinant human interleukin-18 (rHuIL-18) is currently in clinical trials for treatment of cancer. This report presents results of preclinical toxicity studies with rHuIL-18 in cynomolgus monkeys and recombinant murine IL-18 (rMuIL-18) in mice. The rHuIL-18 was administered intravenously in 1 or 2 different 5-day cycles at doses 0.3 to 75 mg/kg/day in monkeys. Decreases in red cell mass, neutrophil, and platelet counts, increases in monocyte and large unstained cell counts, and lymphoid hyperplasia in spleen and lymph nodes were mild, reversible, and likely related to the pharmacologic activity of IL-18. The only toxic effect was protein cast nephropathy, secondary to coprecipitation of administered IL-18 and Tamm-Horsfall protein in the distal nephron, that only occurred at 75 mg/kg/day. Other adverse effects of rHuIL-18 were related to strong immunogenicity in monkeys and were manifest only during a second dosing cycle. The rMuIL-18, at similar dosing levels and cycles in mice, resulted in reduced red cell mass, increased white blood cell counts, spleen and lymph node hyperplasia, and mild, reversible changes in intestine, liver, and lungs. Protein cast nephropathy occurred in mice at doses ≥30 mg/kg/day. In conclusion, preclinical safety studies showed that rIL-18 was well tolerated at pharmacologically active doses in both monkeys and mice.
Collapse
Affiliation(s)
- Danuta J Herzyk
- Department of Safety Assessment, GlaxoSmithKline Pharmaceuticals, King of Prussia, Pennsylvania 19406, USA.
| | | | | | | |
Collapse
|
19
|
Itamura H, Shindo T, Tawara I, Kubota Y, Kariya R, Okada S, Komanduri KV, Kimura S. The MEK inhibitor trametinib separates murine graft-versus-host disease from graft-versus-tumor effects. JCI Insight 2016; 1:e86331. [PMID: 27699218 PMCID: PMC5033881 DOI: 10.1172/jci.insight.86331] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Accepted: 05/31/2016] [Indexed: 01/21/2023] Open
Abstract
The efficacy of allogeneic hematopoietic stem cell transplantation for hematologic malignancies is limited by the difficulty in suppressing graft-versus-host disease (GVHD) without compromising graft-versus-tumor (GVT) effects. We previously showed that RAS/MEK/ERK signaling depends on memory differentiation in human T cells, which confers susceptibility to selective inhibition of naive T cells. Actually, antineoplastic MEK inhibitors selectively suppress alloreactive T cells, sparing virus-specific T cells in vitro. Here, we show that trametinib, a MEK inhibitor clinically approved for melanoma, suppresses GVHD safely without affecting GVT effects in vivo. Trametinib prolonged survival of GVHD mice and attenuated GVHD symptoms and pathology in the gut and skin. It inhibited ERK1/2 phosphorylation and expansion of donor T cells, sparing Tregs and B cells. Although high-dose trametinib inhibited myeloid cell engraftment, low-dose trametinib suppressed GVHD without severe adverse events. Notably, trametinib facilitated the survival of mice transplanted with allogeneic T cells and P815 tumor cells with no residual P815 cells observed in the livers and spleens, whereas tacrolimus resulted in P815 expansion. These results confirm that trametinib selectively suppresses GVHD-inducing T cells while sparing antitumor T cells in vivo, which makes it a promising candidate for translational studies aimed at preventing or treating GVHD.
Collapse
Affiliation(s)
- Hidekazu Itamura
- Department of Hematology, Respiratory Medicine and Oncology, Saga University School of Medicine, Saga, Japan
| | - Takero Shindo
- Department of Hematology, Respiratory Medicine and Oncology, Saga University School of Medicine, Saga, Japan
| | - Isao Tawara
- Department of Hematology/Oncology, Mie University School of Medicine, Tsu, Japan
| | - Yasushi Kubota
- Department of Hematology, Respiratory Medicine and Oncology, Saga University School of Medicine, Saga, Japan
| | - Ryusho Kariya
- Division of Hematopoiesis, Center for AIDS Research, Kumamoto University, Kumamoto, Japan
| | - Seiji Okada
- Division of Hematopoiesis, Center for AIDS Research, Kumamoto University, Kumamoto, Japan
| | - Krishna V. Komanduri
- Adult Stem Cell Transplant Program and Department of Medicine, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Shinya Kimura
- Department of Hematology, Respiratory Medicine and Oncology, Saga University School of Medicine, Saga, Japan
| |
Collapse
|
20
|
IL-18 and Cutaneous Inflammatory Diseases. Int J Mol Sci 2015; 16:29357-69. [PMID: 26690141 PMCID: PMC4691115 DOI: 10.3390/ijms161226172] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2015] [Revised: 11/25/2015] [Accepted: 12/03/2015] [Indexed: 12/15/2022] Open
Abstract
Interleukin (IL)-18, an IL-1 family cytokine, is a pleiotropic immune regulator. IL-18 plays a strong proinflammatory role by inducing interferon (IFN)-γ. Previous studies have implicated IL-18 in the pathogenesis of various diseases. However, it is not well understood biologic activities of IL-18 in the diverse skin diseases. Here, we have reviewed the expression and function of IL-18 in skin diseases including inflammatory diseases. This article provides an evidence-based understanding of the role of IL-18 in skin diseases and its relationship with disease activities.
Collapse
|
21
|
Ahmed SS, Wang XN, Norden J, Pearce K, El-Gezawy E, Atarod S, Hromadnikova I, Collin M, Holler E, Dickinson AM. Identification and validation of biomarkers associated with acute and chronic graft versus host disease. Bone Marrow Transplant 2015; 50:1563-71. [DOI: 10.1038/bmt.2015.191] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2015] [Revised: 07/14/2015] [Accepted: 07/15/2015] [Indexed: 12/28/2022]
|
22
|
Li X, Zhang C, Chen W, Pan B, Kong F, Zheng K, Tang R, Zeng L. Protective effect of neutralizing anti-IL-18α monoclonal antibody on a mouse model of acute graft-versus-host disease. Oncol Rep 2015; 34:2031-9. [PMID: 26252430 DOI: 10.3892/or.2015.4176] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2015] [Accepted: 06/24/2015] [Indexed: 11/06/2022] Open
Abstract
Graft-versus-host disease (GVHD) is a devastating complication of hematopoietic stem cell transplantation (HSCT), and is characterized by systemic inflammation and tissue damage in multiple organs, such as the liver and small intestine. Interleukin-18 (IL-18), an important pro-inflammatory cytokine, is elevated during the course of acute GVHD (aGVHD), and is associated with the severe clinical manifestations of the disease. The biological activity of IL-18 is based on its interaction with the IL-18 receptor (IL-18R) expressed in a variety of cells. The aim of this study was to assess whether blocking the interaction of IL-18 with IL-18R by the anti-IL‑18Rα antibody could attenuate the severity of aGVHD. We used a well-established mouse bone marrow transplantation (BMT) model (B6→BALB/c) to block the IL-18/IL-18R interaction by a neutralizing monoclonal antibody (mAb) against murine IL-18Rα. Administration of anti-IL-18Rα mAb had a significant protective effect on the clinical and pathologic manifestations of aGVHD, resulting in a markedly improved survival rate, modified inflammatory response and decreased tissue damage. Interfering with IL-18/IL-18R interaction affected levels of Th1, Th2 and Th17 subsets in the peripheral blood of the aGVHD animals. Additionally, it led to decreased tissue expression of IL-18 and apoptosis-associated molecules (Fas and FasL), and lower phosphorylation levels of p38MAPK in the liver and small intestine. These changes coincided with the decrease in cell apoptosis in aGVHD target organs. Thus, anti‑IL-18Rα therapy may, therefore, represent a new therapeutic interference approach for treating aGVHD.
Collapse
Affiliation(s)
- Xiaocui Li
- Department of Pathogenic Biology and Immunology, Laboratory of Infection and Immunity, Xuzhou Medical College, Xuzhou, Jiangsu 221004, P.R. China
| | - Cuiping Zhang
- Department of Pathogenic Biology and Immunology, Laboratory of Infection and Immunity, Xuzhou Medical College, Xuzhou, Jiangsu 221004, P.R. China
| | - Wei Chen
- Blood Diseases Institute, Xuzhou Medical College; Key Laboratory of Bone Marrow Stem Cells; Department of Hematology, The Affiliated Hospital of Xuzhou Medical College, Xuzhou, Jiangsu 221002, P.R. China
| | - Bin Pan
- Blood Diseases Institute, Xuzhou Medical College; Key Laboratory of Bone Marrow Stem Cells; Department of Hematology, The Affiliated Hospital of Xuzhou Medical College, Xuzhou, Jiangsu 221002, P.R. China
| | - Fanyun Kong
- Department of Pathogenic Biology and Immunology, Laboratory of Infection and Immunity, Xuzhou Medical College, Xuzhou, Jiangsu 221004, P.R. China
| | - Kuiyang Zheng
- Department of Pathogenic Biology and Immunology, Laboratory of Infection and Immunity, Xuzhou Medical College, Xuzhou, Jiangsu 221004, P.R. China
| | - Renxian Tang
- Department of Pathogenic Biology and Immunology, Laboratory of Infection and Immunity, Xuzhou Medical College, Xuzhou, Jiangsu 221004, P.R. China
| | - Lingyu Zeng
- Blood Diseases Institute, Xuzhou Medical College; Key Laboratory of Bone Marrow Stem Cells; Department of Hematology, The Affiliated Hospital of Xuzhou Medical College, Xuzhou, Jiangsu 221002, P.R. China
| |
Collapse
|
23
|
Koyama M, Cheong M, Markey KA, Gartlan KH, Kuns RD, Locke KR, Lineburg KE, Teal BE, Leveque-El Mouttie L, Bunting MD, Vuckovic S, Zhang P, Teng MWL, Varelias A, Tey SK, Wockner LF, Engwerda CR, Smyth MJ, Belz GT, McColl SR, MacDonald KPA, Hill GR. Donor colonic CD103+ dendritic cells determine the severity of acute graft-versus-host disease. THE JOURNAL OF EXPERIMENTAL MEDICINE 2015. [PMID: 26169940 DOI: 10.1084/jem.20150329.] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
The primacy of the gastrointestinal (GI) tract in dictating the outcome of graft-versus-host disease (GVHD) is broadly accepted; however, the mechanisms controlling this effect are poorly understood. Here, we demonstrate that GVHD markedly enhances alloantigen presentation within the mesenteric lymph nodes (mLNs), mediated by donor CD103(+)CD11b(-) dendritic cells (DCs) that migrate from the colon under the influence of CCR7. Expansion and differentiation of donor T cells specifically within the mLNs is driven by profound levels of alloantigen, IL-12, and IL-6 promoted by Toll-like receptor (TLR) and receptor for advanced glycation end products (RAGE) signals. Critically, alloantigen presentation in the mLNs imprints gut-homing integrin signatures on donor T cells, leading to their emigration into the GI tract where they mediate fulminant disease. These data identify a critical, anatomically distinct, donor DC subset that amplifies GVHD. We thus highlight multiple therapeutic targets and the ability of GVHD, once initiated by recipient antigen-presenting cells, to generate a profound, localized, and lethal feed-forward cascade of donor DC-mediated indirect alloantigen presentation and cytokine secretion within the GI tract.
Collapse
Affiliation(s)
- Motoko Koyama
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland 4006, Australia
| | - Melody Cheong
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland 4006, Australia
| | - Kate A Markey
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland 4006, Australia
| | - Kate H Gartlan
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland 4006, Australia
| | - Rachel D Kuns
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland 4006, Australia
| | - Kelly R Locke
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland 4006, Australia
| | - Katie E Lineburg
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland 4006, Australia
| | - Bianca E Teal
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland 4006, Australia
| | | | - Mark D Bunting
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland 4006, Australia
| | - Slavica Vuckovic
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland 4006, Australia
| | - Ping Zhang
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland 4006, Australia
| | - Michele W L Teng
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland 4006, Australia
| | - Antiopi Varelias
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland 4006, Australia
| | - Siok-Keen Tey
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland 4006, Australia The Royal Brisbane and Women's Hospital, Brisbane, Queensland 4029, Australia
| | - Leesa F Wockner
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland 4006, Australia
| | | | - Mark J Smyth
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland 4006, Australia
| | - Gabrielle T Belz
- Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria 3052, Australia
| | - Shaun R McColl
- The University of Adelaide, Adelaide, South Australia 5005, Australia
| | - Kelli P A MacDonald
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland 4006, Australia
| | - Geoffrey R Hill
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland 4006, Australia The Royal Brisbane and Women's Hospital, Brisbane, Queensland 4029, Australia
| |
Collapse
|
24
|
Koyama M, Cheong M, Markey KA, Gartlan KH, Kuns RD, Locke KR, Lineburg KE, Teal BE, Leveque-El Mouttie L, Bunting MD, Vuckovic S, Zhang P, Teng MWL, Varelias A, Tey SK, Wockner LF, Engwerda CR, Smyth MJ, Belz GT, McColl SR, MacDonald KPA, Hill GR. Donor colonic CD103+ dendritic cells determine the severity of acute graft-versus-host disease. ACTA ACUST UNITED AC 2015; 212:1303-21. [PMID: 26169940 PMCID: PMC4516799 DOI: 10.1084/jem.20150329] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2015] [Accepted: 06/26/2015] [Indexed: 12/23/2022]
Abstract
Koyama et al. show that GVHD markedly enhances alloantigen presentation within the mesenteric lymph nodes, mediated by donor CD103+CD11b− DCs that migrate from the colon under the influence of CCR7. This antigen presentation imprints gut-homing integrin signatures on donor T cells, leading to their migration to the GI tract where they mediate fulminant disease. The primacy of the gastrointestinal (GI) tract in dictating the outcome of graft-versus-host disease (GVHD) is broadly accepted; however, the mechanisms controlling this effect are poorly understood. Here, we demonstrate that GVHD markedly enhances alloantigen presentation within the mesenteric lymph nodes (mLNs), mediated by donor CD103+CD11b− dendritic cells (DCs) that migrate from the colon under the influence of CCR7. Expansion and differentiation of donor T cells specifically within the mLNs is driven by profound levels of alloantigen, IL-12, and IL-6 promoted by Toll-like receptor (TLR) and receptor for advanced glycation end products (RAGE) signals. Critically, alloantigen presentation in the mLNs imprints gut-homing integrin signatures on donor T cells, leading to their emigration into the GI tract where they mediate fulminant disease. These data identify a critical, anatomically distinct, donor DC subset that amplifies GVHD. We thus highlight multiple therapeutic targets and the ability of GVHD, once initiated by recipient antigen-presenting cells, to generate a profound, localized, and lethal feed-forward cascade of donor DC–mediated indirect alloantigen presentation and cytokine secretion within the GI tract.
Collapse
Affiliation(s)
- Motoko Koyama
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland 4006, Australia
| | - Melody Cheong
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland 4006, Australia
| | - Kate A Markey
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland 4006, Australia
| | - Kate H Gartlan
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland 4006, Australia
| | - Rachel D Kuns
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland 4006, Australia
| | - Kelly R Locke
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland 4006, Australia
| | - Katie E Lineburg
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland 4006, Australia
| | - Bianca E Teal
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland 4006, Australia
| | | | - Mark D Bunting
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland 4006, Australia
| | - Slavica Vuckovic
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland 4006, Australia
| | - Ping Zhang
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland 4006, Australia
| | - Michele W L Teng
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland 4006, Australia
| | - Antiopi Varelias
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland 4006, Australia
| | - Siok-Keen Tey
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland 4006, Australia The Royal Brisbane and Women's Hospital, Brisbane, Queensland 4029, Australia
| | - Leesa F Wockner
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland 4006, Australia
| | | | - Mark J Smyth
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland 4006, Australia
| | - Gabrielle T Belz
- Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria 3052, Australia
| | - Shaun R McColl
- The University of Adelaide, Adelaide, South Australia 5005, Australia
| | - Kelli P A MacDonald
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland 4006, Australia
| | - Geoffrey R Hill
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland 4006, Australia The Royal Brisbane and Women's Hospital, Brisbane, Queensland 4029, Australia
| |
Collapse
|
25
|
State-of-the-art acute and chronic GVHD treatment. Int J Hematol 2015; 101:452-66. [DOI: 10.1007/s12185-015-1785-1] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2015] [Accepted: 03/23/2015] [Indexed: 01/09/2023]
|
26
|
Wang H, Yang YG. The complex and central role of interferon-γ in graft-versus-host disease and graft-versus-tumor activity. Immunol Rev 2015; 258:30-44. [PMID: 24517424 DOI: 10.1111/imr.12151] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2013] [Revised: 11/01/2013] [Accepted: 11/11/2013] [Indexed: 12/22/2022]
Abstract
Allogeneic hematopoietic cell transplantation (allo-HCT) is increasingly being performed to treat patients with hematologic malignancies. However, separating the beneficial graft-versus-tumor (GVT) or graft-versus-leukemia effects from graft-versus-host disease (GVHD) has been difficult and remains a significant challenge toward improving therapeutic efficacy and reducing toxicity of allo-HCT. GVHD is induced by donor T cells that also mediate potent anti-tumor responses. However, despite the largely shared effector mechanisms, extensive animal studies have demonstrated the potential of dissociating the GVT effect from GVHD. Also in many clinical cases, long-term remission was achieved following allo-HCT, without significant GVHD. A better mechanistic understanding of the immunopathophysiology of GVHD and GVT effects may potentially help to improve allo-HCT as well as maximize the benefit of GVT effects while minimizing GVHD. In this article, we review the role of IFN-γ in regulation of alloresponses following allo-HCT, with a focus on the mechanisms of how this cytokine may separate GVHD from GVT effects.
Collapse
Affiliation(s)
- Hui Wang
- Columbia Center for Translational Immunology, Columbia University College of Physicians and Surgeons, New York, NY, USA
| | | |
Collapse
|
27
|
Raza A, Vierling JM. Graft-Versus-Host Disease. LIVER IMMUNOLOGY 2014:425-441. [DOI: 10.1007/978-3-319-02096-9_29] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
28
|
Saha A, Aoyama K, Taylor PA, Koehn BH, Veenstra RG, Panoskaltsis-Mortari A, Munn DH, Murphy WJ, Azuma M, Yagita H, Fife BT, Sayegh MH, Najafian N, Socie G, Ahmed R, Freeman GJ, Sharpe AH, Blazar BR. Host programmed death ligand 1 is dominant over programmed death ligand 2 expression in regulating graft-versus-host disease lethality. Blood 2013; 122:3062-73. [PMID: 24030385 PMCID: PMC3811178 DOI: 10.1182/blood-2013-05-500801] [Citation(s) in RCA: 137] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2013] [Accepted: 09/03/2013] [Indexed: 12/15/2022] Open
Abstract
Programmed death 1 (PD-1) and its ligands, PD-L1 and PD-L2, play an important role in the maintenance of peripheral tolerance. We explored the role of PD-1 ligands in regulating graft-versus-host disease (GVHD). Both PD-L1 and PD-L2 expression were upregulated in the spleen, liver, colon, and ileum of GVHD mice. Whereas PD-L2 expression was limited to hematopoietic cells, hematopoietic and endothelial cells expressed PD-L1. PD-1/PD-L1, but not PD-1/PD-L2, blockade markedly accelerated GVHD-induced lethality. Chimera studies suggest that PD-L1 expression on host parenchymal cells is more critical than hematopoietic cells in regulating acute GVHD. Rapid mortality onset in PD-L1-deficient hosts was associated with increased gut T-cell homing and loss of intestinal epithelial integrity, along with increased donor T-cell proliferation, activation, Th1 cytokine production, and reduced apoptosis. Bioenergetics profile analysis of proliferating alloreactive donor T-cells demonstrated increased aerobic glycolysis and oxidative phosphorylation in PD-L1-deficient hosts. Donor T-cells exhibited a hyperpolarized mitochondrial membrane potential, increased superoxide production, and increased expression of a glucose transporter in PD-L1-deficient hosts. Taken together, these data provide new insight into the differential roles of host PD-L1 and PD-L2 and their associated cellular and metabolic mechanisms controlling acute GVHD.
Collapse
Affiliation(s)
- Asim Saha
- Masonic Cancer Center and Department of Pediatrics, Division of Blood and Marrow Transplantation, University of Minnesota, Minneapolis, MN
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Kim YH, Han SB, Lee JK. Histone deacetylase inhibitors suppress CXCR4-mediated dendritic cell migration by regulation of maturation process. Cell Immunol 2013; 284:139-45. [DOI: 10.1016/j.cellimm.2013.07.014] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2013] [Revised: 07/05/2013] [Accepted: 07/28/2013] [Indexed: 10/26/2022]
|
30
|
The Protection and Therapy Effects of Bortezomib in Murine Acute Graft-Versus-Host Disease. Transplant Proc 2013; 45:2527-35. [DOI: 10.1016/j.transproceed.2013.03.042] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2012] [Revised: 12/11/2012] [Accepted: 03/06/2013] [Indexed: 12/12/2022]
|
31
|
Shikari H, Antin JH, Dana R. Ocular Graft-versus-Host Disease: A Review. Surv Ophthalmol 2013; 58:233-51. [DOI: 10.1016/j.survophthal.2012.08.004] [Citation(s) in RCA: 122] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2012] [Revised: 07/30/2012] [Accepted: 08/07/2012] [Indexed: 12/13/2022]
|
32
|
Tawara I, Liu C, Tamaki H, Toubai T, Sun Y, Evers R, Nieves E, Mathewson N, Nunez G, Reddy P. Influence of donor microbiota on the severity of experimental graft-versus-host-disease. Biol Blood Marrow Transplant 2012; 19:164-8. [PMID: 22982686 DOI: 10.1016/j.bbmt.2012.09.001] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2012] [Accepted: 09/06/2012] [Indexed: 01/10/2023]
Abstract
The link between microbial flora and the shaping of immune responses is being increasingly appreciated, and recent data have uncovered a role for recipient microbiota in the severity of graft-versus-host disease (GVHD). The impact of donor microbiota on T cell-mediated alloresponses and GVHD is not known, however. Using multiple clinically relevant murine models, we analyzed the effect of donor microbiota on the severity of GVHD induced by T cells from specific pathogen-free and germ-free donors, and found that donor microbiota does not alter the expansion or differentiation of alloreactive T cells or the severity of GVHD.
Collapse
Affiliation(s)
- Isao Tawara
- Department of Internal Medicine, University of Michigan Comprehensive Cancer Center, Ann Arbor, Michigan, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Koenecke C, Lee CW, Thamm K, Föhse L, Schafferus M, Mittrücker HW, Floess S, Huehn J, Ganser A, Förster R, Prinz I. IFN-γ production by allogeneic Foxp3+ regulatory T cells is essential for preventing experimental graft-versus-host disease. THE JOURNAL OF IMMUNOLOGY 2012; 189:2890-6. [PMID: 22869903 DOI: 10.4049/jimmunol.1200413] [Citation(s) in RCA: 99] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
It is emerging that CD4+Foxp3+ regulatory T (Treg) cells can produce the proinflammatory cytokine IFN-γ when stimulated in a Th1 cytokine environment. In this study, we report that Foxp3+ Treg cells readily produced IFN-γ in vivo in a highly inflammatory model of graft-versus-host disease (GVHD) and during a Th1-dominated immune response to intracellular bacteria. Moreover, stimulation in vitro via TCR in the presence of IL-12 alone was sufficient to induce IFN-γ production by Treg cells in a dose-dependent manner. Transfer of donor Treg cells can prevent lethal GVHD; therefore, we used this model as a robust readout for in vivo Treg function. Interestingly, >50% of allogeneic donor, but not residual recipient Foxp3+ Treg cells produced IFN-γ after transplantation, suggesting that this cytokine production was alloantigen specific. These IFN-γ producers were stable Foxp3+ Treg cells because methylation analysis of the Foxp3 gene locus of transferred and reisolated Treg cells during GVHD showed a fully demethylated Treg-specific-demethylated region. Next, we addressed whether IFN-γ production was supporting or rather impairing the immunosuppressive function of Treg cells during GVHD. Blocking of IFN-γ with specific mAb completely abolished the beneficial effect of donor Treg cells. We could further show that only wild-type Treg cells, but not Treg cells from IFN-γ-deficient donor mice, prevented GVHD. This indicated that Treg cell-intrinsic IFN-γ production was required for their protective function. In conclusion, our data show that IFN-γ produced by Foxp3+ Treg cells has essential immune-regulatory functions that are required for prevention of experimental GVHD.
Collapse
Affiliation(s)
- Christian Koenecke
- Institute of Immunology, Hannover Medical School, 30625 Hannover, Germany.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Resende RG, Correia-Silva JDF, Silva TA, Xavier SG, Bittencourt H, Gomez RS, Abreu MHNG. Saliva and blood interferon gamma levels andIFNGgenotypes in acute graft-versus-host disease. Oral Dis 2012; 18:816-22. [DOI: 10.1111/j.1601-0825.2012.01955.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
35
|
Suppression of T cell functions by hydroxamic acid-based histone deacetylase inhibitors. Arch Pharm Res 2012; 35:929-36. [DOI: 10.1007/s12272-012-0519-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2011] [Revised: 11/21/2011] [Accepted: 01/19/2012] [Indexed: 01/15/2023]
|
36
|
The interferon-dependent orchestration of innate and adaptive immunity after transplantation. Blood 2012; 119:5351-8. [PMID: 22517908 DOI: 10.1182/blood-2012-02-368076] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
The therapeutic GVL effect after allogeneic stem cell transplantation is limited by the development of GVHD. The ultimate aim of current research is to separate the 2 processes in a meaningful fashion. The IFNs are a pleiotropic group of cytokines that were originally recognized because of their ability to interfere with viral replication. However, it is now established that these cytokines play an important role in orchestrating both innate and adaptive immunity. Multiple studies have investigated the effects of both types I and II IFN on GVHD and GVL in preclinical transplant models. The results indicate variable effects that are dependent on the period of activity within the developing immune response, the presence and type of pretransplant conditioning and the differential mechanisms, and IFN sensitivity of immune pathology within individual target organs during GVHD. This Perspective discusses the current literature on the IFNs and their potential modulation within clinical transplantation, focusing particularly on enhancing the therapeutic GVL effects.
Collapse
|
37
|
Kim AR, Lim JY, Jeong DC, Park G, Lee BC, Min CK. Blockade of Vascular Endothelial Growth Factor (VEGF) Aggravates the Severity of Acute Graft-versus-host Disease (GVHD) after Experimental Allogeneic Hematopoietic Stem Cell Transplantation (allo-HSCT). Immune Netw 2011; 11:368-75. [PMID: 22346777 PMCID: PMC3275706 DOI: 10.4110/in.2011.11.6.368] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2011] [Revised: 10/28/2011] [Accepted: 11/03/2011] [Indexed: 12/01/2022] Open
Abstract
BACKGROUND Recent clinical observation reported that there was a significant correlation between change in circulating vascular endothelial growth factor (VEGF) levels and the occurrence of severe acute graft-versus-host disease (GVHD) following allogeneic hematopoietic stem cell transplantation (allo-HSCT), but the action mechanisms of VEGF in GVHD have not been demonstrated. METHODS This study investigated whether or not blockade of VEGF has an effect on acute GVHD in a lethally irradiated murine allo-HSCT model of B6 (H-2(b))→B6D2F1 (H-2(b/d)). Syngeneic or allogeneic recipient mice were injected subcutaneously with anti-VEGF peptides, dRK6 (50 µg/dose) or control diluent every other day for 2 weeks (total 7 doses). RESULTS Administration of the dRK6 peptide after allo-HSCT significantly reduced survival with greaterclinical GVHD scores and body weight loss. Allogeneic recipients injected with the dRK6 peptide exhibited significantly increased circulating levels of VEGF and expansion of donor CD3(+) T cells on day +7 compared to control treated animals. The donor CD4(+) and CD8(+) T-cell subsets have differential expansion caused by the dRK6 injection. The circulating VEGF levels were reduced on day +14 regardless of blockade of VEGF. CONCLUSION Together these findings demonstrate that the allo-reactive responses after allo-HSCT are exaggerated by the blockade of VEGF. VEGF seems to be consumed during the progression of acute GVHD in this murine allo-HSCT model.
Collapse
Affiliation(s)
- Ai-Ran Kim
- Department of Pediatrics, The Catholic University of Korea, Seoul 137-701, Korea
| | | | | | | | | | | |
Collapse
|
38
|
Alpha-1-antitrypsin monotherapy reduces graft-versus-host disease after experimental allogeneic bone marrow transplantation. Proc Natl Acad Sci U S A 2011; 109:564-9. [PMID: 22203983 DOI: 10.1073/pnas.1117665109] [Citation(s) in RCA: 105] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Acute graft-versus-host disease (GvHD) is a major complication that prevents successful outcomes after allogeneic bone marrow transplantation (BMT), an effective therapy for hematological malignancies. Several studies demonstrate that donor T cells and host antigen-presenting cells along with several proinflammatory cytokines are required for the induction of GvHD and contribute to its severity. Increasing evidence demonstrates that human serum-derived αalpha-1- anti-trypsin (AAT) reduces production of proinflammatory cytokines, induces anti-inflammatory cytokines, and interferes with maturation of dendritic cells. Using well-characterized mouse models of BMT, we have studied the effects of AAT on GvHD severity. Administration of AAT early after BMT decreased mortality in three models of GvHD and reduced serum levels of proinflammatory cytokines in the allogeneic recipients compared with vehicle (albumin) treated animals. AAT treatment reduced the expansion of alloreactive T effector cells but enhanced the recovery of T regulatory T cells, (Tregs) thus altering the ratio of donor T effector to T regulatory cells in favor of reducing the pathological process. However, despite altering the ratio in vivo, AAT had no direct effects on either the donor T effector cells or T regulatory cells Tregs in vitro. In contrast, AAT suppressed LPS-induced in vitro secretion of proinflammatory cytokines such as TNF-α and IL-1β, enhanced the production of the anti-inflammatory cytokine IL-10, and impaired NF-κB translocation in the host dendritic cells. In light of its long history of safety in humans, these findings suggest that administration of AAT represents a novel unique and viable strategy to mitigate clinical GvHD.
Collapse
|
39
|
Recipient nonhematopoietic antigen-presenting cells are sufficient to induce lethal acute graft-versus-host disease. Nat Med 2011; 18:135-42. [PMID: 22127134 DOI: 10.1038/nm.2597] [Citation(s) in RCA: 176] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2011] [Accepted: 11/08/2011] [Indexed: 11/08/2022]
Abstract
The presentation pathways by which allogeneic peptides induce graft-versus-host disease (GVHD) are unclear. We developed a bone marrow transplant (BMT) system in mice whereby presentation of a processed recipient peptide within major histocompatibility complex (MHC) class II molecules could be spatially and temporally quantified. Whereas donor antigen presenting cells (APCs) could induce lethal acute GVHD via MHC class II, recipient APCs were 100-1,000 times more potent in this regard. After myeloablative irradiation, T cell activation and memory differentiation occurred in lymphoid organs independently of alloantigen. Unexpectedly, professional hematopoietic-derived recipient APCs within lymphoid organs had only a limited capacity to induce GVHD, and dendritic cells were not required. In contrast, nonhematopoietic recipient APCs within target organs induced universal GVHD mortality and promoted marked alloreactive donor T cell expansion within the gastrointestinal tract and inflammatory cytokine generation. These data challenge current paradigms, suggesting that experimental lethal acute GVHD can be induced by nonhematopoietic recipient APCs.
Collapse
|
40
|
Lim JY, Choi MS, Youn H, Choi EY, Min CK. The influence of pretransplantation conditioning on graft-vs.-leukemia effect in mice. Exp Hematol 2011; 39:1018-29. [PMID: 21767512 DOI: 10.1016/j.exphem.2011.07.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2010] [Revised: 07/04/2011] [Accepted: 07/10/2011] [Indexed: 11/24/2022]
Abstract
OBJECTIVE The therapeutic potential of allogeneic hematopoietic stem cell transplantation relies on the graft-vs.-leukemia (GVL) effect to eradicate residual tumor cells immunologically. The relationship of conditioning intensity to GVL effect was investigated. MATERIALS AND METHODS Lethally irradiated (either 900 or 1300 cGy) B6D2F1 (H-2(b/d)) recipients were transplanted from B6 (H-2(b)) donors. P815 or L1214 (H-2(d)) tumor cells were injected intravenously or subcutaneously on day 1 post-transplantation to generate a GVL model. RESULTS Tumors in allogeneic mice treated with 1300 cGy exhibited markedly delayed subcutaneous tumor growth in vivo as compared with mice treated with 900 cGy, while intravenous tumor growths were comparable between the two radiation doses. Serum levels of tumor necrosis factor-α or interferon-γ were similar and the percentages of donor T-cell proliferation or apoptosis early after hematopoietic stem cell transplantation were comparable. The numbers of CD8(+) T cells from recipients that received 1300 cGy were significantly elevated in skin and tumor tissues. CD62L(low) and CD44(high) CD8(+) T-cell fractions were also elevated in 1300 cGy. After the higher radiation dose, P815-specific interferon-γ responses of splenic CD8(+) T cells were markedly enhanced and the fractions of T cells of interferon-γ-producing T cells in tumor tissues but not in spleen were increased. The protein concentrations of chemokines in skin and tumor tissues were substantially elevated in 1300 cGy compared to 900 cGy. CONCLUSIONS The higher radiation dose (1300 vs. 900 cGy) resulted in significantly enhanced GVL effect against an extramedullary tumor and the alterations in effector T-cell trafficking into tumor tissue are the most likely mechanism.
Collapse
Affiliation(s)
- Ji-Young Lim
- Department of Internal Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea
| | | | | | | | | |
Collapse
|
41
|
Teshima T, Maeda Y, Ozaki K. Regulatory T cells and IL-17-producing cells in graft-versus-host disease. Immunotherapy 2011; 3:833-52. [DOI: 10.2217/imt.11.51] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Graft-versus-host disease (GvHD), a major complication following allogeneic hematopoietic stem cell transplantation, is mediated by donor-derived T cells. On activation with alloantigens expressed on host antigen-presenting cells, naive CD4+ T cells differentiate into T-helper cell subsets of effector T cells expressing distinct sets of transcriptional factors and cytokines. Classically, acute GvHD was suggested to be predominantly related to Th1 responses. However, we now face a completely different and complex scenario involving possible roles of newly identified Th17 cells as well as Tregs in GvHD. Accumulating data from experimental and clinical studies suggest that the fine balance between Th1, Th2, Th17 and Tregs after transplantation may be an important determinant of the severity, manifestation and tissue distribution of GvHD. Understanding the dynamic process of reciprocal differentiation of regulatory and T-helper cell subsets as well as their interactions will be important in establishing novel strategies for preventing and treating GvHD.
Collapse
Affiliation(s)
- Takanori Teshima
- Center for Cellular & Molecular Medicine, Kyushu University Hospital, Fukuoka, Japan
| | - Yoshinobu Maeda
- Biopathological Science, Okayama University Graduate School of Medicine & Dentistry, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8558, Japan
| | - Katsutoshi Ozaki
- Division of Hematology, Department of Medicine, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke-shi, Tochigi 329–0498, Japan
| |
Collapse
|
42
|
Choi S, Reddy P. HDAC inhibition and graft versus host disease. Mol Med 2011; 17:404-16. [PMID: 21298214 DOI: 10.2119/molmed.2011.00007] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2011] [Accepted: 01/07/2011] [Indexed: 12/12/2022] Open
Abstract
Histone deacetylase (HDAC) inhibitors are currently used clinically as anticancer drugs. Recent data have demonstrated that some of these drugs have potent antiinflammatory or immunomodulatory effects at noncytotoxic doses. The immunomodulatory effects have shown potential for therapeutic benefit after allogeneic bone marrow transplantation in several experimental models of graft versus host disease (GVHD). These effects, at least in part, result from the ability of HDAC inhibitors (HDACi) to suppress the function of host antigen presenting cells such as dendritic cells (DC). HDACi reduce the dendritic cell (DC) responses, in part, by enhancing the expression of indoleamine 2,3-dioxygenase (IDO) in a signal transducer and activator of transcription-3 (STAT-3) dependent manner. They also alter the function of other immune cells such as T regulatory cells and natural killer (NK) cells, which also play important roles in the biology of GVHD. Based on these observations, a clinical trial has been launched to evaluate the impact of HDAC inhibitors on clinical GVHD. The experimental, mechanistic studies along with the brief preliminary observations from the ongoing clinical trial are discussed in this review.
Collapse
Affiliation(s)
- Sung Choi
- Department of Pediatrics, University of Michigan Comprehensive Cancer Center, Ann Arbor, Michigan 48105, USA
| | | |
Collapse
|
43
|
Lee S, Kim S, Bae S, Choi J, Hong J, Ryoo S, Jhun H, Hong K, Kim E, Jo S, Her E, Kim S. Development of isoform-specific monoclonal antibodies against human IL-18 binding protein. Hybridoma (Larchmt) 2010; 29:517-24. [PMID: 21087098 DOI: 10.1089/hyb.2010.0058] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Interleukin-18 binding protein (IL-18BP) is a soluble antagonist of IL-18 originally discovered while attempting to isolate a soluble receptor by using IL-18-ligand affinity column. IL-18BP has four isoforms (a, b, c, and d) in humans and two isoforms (c and d) in mice. The human isoforms IL-18BPa and IL-18BPc neutralize IL-18 activity sufficiently at an equimolar ratio; however IL-18BPb and IL-18BPd isoforms lack a complete Ig domain at C-terminus and lose the ability to neutralize IL-18 activity. Mouse IL-18BPc and IL-18BPd isoforms, possessing a similar complete Ig domain, also neutralize the biological activity of mouse IL-18 at an equimolar ratio. Here we expressed recombinant proteins of the active human IL-18BP isoforms and developed monoclonal antibodies (MAbs) against human IL-18BP a and c isoforms. We obtained two MAbs (78-4 and 38-3) of human IL-18BPa and two MAbs (18-7 and 29-6) of human IL-18BPc. The MAb clones 18-7 and 29-6 specifically recognized recombinant IL-18BPc in Western blot analyses and ELISA, whereas the MAb clone 78-4 recognized both isoforms in Western blot analyses, but only human IL-18BPa isoform in ELISA. We developed a sandwich ELISA by using the monoclonal antibody specific to human IL-18BPa isoform. The isoform-specific anti-human IL-18BP MAb may be a useful tool in categorizing a distinct group of patients from various autoimmune diseases related to IL-18BP.
Collapse
Affiliation(s)
- Siyoung Lee
- Department of Immunology, College of Medicine, Konkuk University, Chungju City, Chungbuk, Korea
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Tawara I, Koyama M, Liu C, Toubai T, Thomas D, Evers R, Chockley P, Nieves E, Sun Y, Lowler KP, Malter C, Nishimoto N, Hill GR, Reddy P. Interleukin-6 modulates graft-versus-host responses after experimental allogeneic bone marrow transplantation. Clin Cancer Res 2010; 17:77-88. [PMID: 21047980 DOI: 10.1158/1078-0432.ccr-10-1198] [Citation(s) in RCA: 149] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
PURPOSE The graft-versus-tumor (GVT) effect is a potent form of immunotherapy against many hematologic malignancies and some solid tumors. The beneficial GVT effect after allogeneic bone marrow transplantation (BMT) is tightly linked to its most significant complication, graft-versus-host disease (GVHD). The role of interleukin-6 (IL-6) after allogeneic BMT is not well understood. This study used a series of complementary knockout and antibody blockade strategies to analyze the impact of IL-6 in multiple clinically relevant murine models of GVHD and GVT. EXPERIMENTAL DESIGN We examined the effect of the source of IL-6 by analyzing the role IL-6 deficiency in donor T cells, donor bone marrow or in host tissues. We confirmed and extended the relevance of IL-6 deficiency on GVHD and GVT by treating BMT recipients with anti-mouse IL-6 receptor (IL-6R), MR16-1. RESULTS Deficiency of IL-6 in donor T cells led to prolongation of survival. Total inhibition of IL-6 with MR16-1 caused an even greater reduction in GVHD-induced mortality. The reduction in GVHD was independent of the direct effects on T effector cell expansion or donor regulatory T cells. GVT responses were preserved after treatment with MR16-1. CONCLUSION MR16-1 treatment reduced GVHD and preserved sufficient GVT. Tocilizumab, a humanized anti-IL-6R monoclonal antibody (mAb), is approved in several countries including the United States and European Union for the treatment of rheumatoid arthritis and other inflammatory diseases. Blockade of IL-6 with anti-IL-6R mAb therapy may be testable in clinical trials as an adjunct to prevent GVHD in BMT patients without a significant loss of GVT.
Collapse
Affiliation(s)
- Isao Tawara
- Department of Internal Medicine and University of Michigan Comprehensive Cancer Center, Ann Arbor, Michigan 48109-0942, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Feng D, Sangster-Guity N, Stone R, Korczeniewska J, Mancl ME, Fitzgerald-Bocarsly P, Barnes BJ. Differential requirement of histone acetylase and deacetylase activities for IRF5-mediated proinflammatory cytokine expression. THE JOURNAL OF IMMUNOLOGY 2010; 185:6003-12. [PMID: 20935208 DOI: 10.4049/jimmunol.1000482] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Recent evidence indicates a new role for histone deacetylases (HDACs) in the activation of genes governing the host immune response. Virus, along with other pathogenic stimuli, triggers an antiviral defense mechanism through the induction of IFN, IFN-stimulated genes, and other proinflammatory cytokines. Many of these genes have been shown to be regulated by transcription factors of the IFN regulatory factor (IRF) family. Recent studies from IRF5 knockout mice have confirmed a critical role for IRF5 in virus-induced type I IFN expression and proinflammatory cytokines IL-6, IL-12, and TNF-α; yet, little is known of the molecular mechanism of IRF5-mediated proinflammatory cytokine expression. In this study, we show that both HDACs and histone acetyltransferases (HATs) associate with IRF5, leading to alterations in its transactivation ability. Using the HDAC inhibitor trichostatin A, we demonstrate that ISRE, IFNA, and IL6 promoters require HDAC activity for transactivation and transcription, whereas TNFα does not. Mapping the interaction of corepressor proteins (HDAC1, silencing mediator of retinoid and thyroid receptor/nuclear corepressor of retinoid receptor, and Sin3a) and HATs to IRF5 revealed distinct differences, including the dependence of IRF5 phosphorylation on HAT association resulting in IRF5 acetylation. Data presented in this study support a mechanism whereby virus triggers the dynamic conversion of an IRF5-mediated silencing complex to that of an activating complex on promoters of target genes. These data provide the first evidence, to our knowledge, of a tightly controlled transcriptional mechanism whereby IRF5 regulates proinflammatory cytokine expression in conjunction with HATs and HDACs.
Collapse
Affiliation(s)
- Di Feng
- Department of Biochemistry and Molecular Biology, New Jersey Medical School, University of Medicine and Dentistry of New Jersey, Newark, NJ 07103, USA
| | | | | | | | | | | | | |
Collapse
|
46
|
Kim J, Park S, Kim HA, Jung D, Kim HJ, Choi HJ, Cho HR, Kwon B. Roles of Host Nonhematopoietic Cells in Autoimmunity and Donor Cell Engraftment in Graft-versus-host Disease. Immune Netw 2010; 10:46-54. [PMID: 20532124 PMCID: PMC2881427 DOI: 10.4110/in.2010.10.2.46] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2010] [Revised: 03/25/2010] [Accepted: 03/31/2010] [Indexed: 02/04/2023] Open
Abstract
Background Graft-versus-host disease (GVHD) is initiated when alloreactive donor T cells are primed by host APCs to undergo clonal expansion and maturation. Since there is a controversy regarding the role of nonhematopoietic cells in GVHD, we wanted to investigate the influence of MHC disparity on nonhematopoietic cells on the pathogenesis of GVHD in the MHC-haplomismatched C57BL/6 (H-2b) or DBA/2 (H-2d)→unirradiated (C57BL/6×DBA/2) F1(BDF1; H-2b/d) murine model of acute GVHD (aGVHD) or chronic GVHD (cGVHD). Methods We generated (BDF1→C57BL/6), (BDF1→DBA/2), and (BDF1→BDF1) chimeras and examined GVHD-related parameters and donor cell engraftment in those chimeras. Results Using this experimental system, we found that 1) severe aGVHD across MHC Ag barrier depends on the expression of nonhematopoietically rather than hematopoietically derived alloAgs for maximal GVHD manifestations; 2) host APCs were sufficient to break B cell tolerance to self molecules in cGVHD, whereas host APCs were insufficient to induce autoimmunity in aGVHD; 3) donor cell engraftment was greatly enhanced in the host with MHC-matched nonhematopoietic cells. Conclusion Taken together, our results provide an insight into how MHC disparity on GVHD target organs contribute to the pathogenesis of GVHD.
Collapse
Affiliation(s)
- Juyang Kim
- Biomedical Research Center, Ulsan University Hospital, School of Medicine, Ulsan, Korea
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Rosenblatt J, Bissonnette A, Ahmad R, Wu Z, Vasir B, Stevenson K, Zarwan C, Keefe W, Glotzbecker B, Mills H, Joyce R, Levine JD, Tzachanis D, Boussiotis V, Kufe D, Avigan D. Immunomodulatory effects of vitamin D: implications for GVHD. Bone Marrow Transplant 2010; 45:1463-8. [PMID: 20081878 DOI: 10.1038/bmt.2009.366] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
GVHD remains a major source of morbidity and mortality after allogeneic BMT. GVHD is mediated by alloreactive T cells derived from the hematopoietic graft that target host tissues. Pre-clinical models have shown that presentation of alloantigens by host DCs results in the activation of donor-derived T cells that mediate GVHD. Strategies that interfere with the Ag-presenting capacity of DCs after allogeneic transplantation may decrease the risk of developing GVHD. Vitamin D is a hormone essential for calcium metabolism that shows immunomodulatory properties. We showed that correction of vitamin D deficiency appeared to mitigate manifestations of GVHD. In pre-clinical studies, we have shown that vitamin D inhibits DC maturation, polarizes T-cell populations toward the expression of Th2 as compared with Th1 cytokines, and blunts allogeneic T-cell proliferation in response to DC stimulation. Exposure to vitamin D resulted in increased expression of IDO, an enzyme responsible for tryptophan metabolism that is upregulated in tolerizing DCs. These data suggest that exposure to vitamin D results in immature DC populations that bias toward tolerizing rather than stimulatory T-cell populations. Vitamin D may therefore have a role in the prevention of GVHD.
Collapse
Affiliation(s)
- J Rosenblatt
- Division of Hematology/Oncology, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Kim ES, Lee JK. Histone deacetylase inhibitors decrease the antigen presenting activity of murine bone marrow derived dendritic cells. Cell Immunol 2009; 262:52-7. [PMID: 20096829 DOI: 10.1016/j.cellimm.2009.12.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2009] [Accepted: 12/21/2009] [Indexed: 11/29/2022]
Abstract
Once activated by infected pathogens, dendritic cells (DCs) undergo activation and release inflammatory mediators responsible for the signs of inflammation. Our aim was to elucidate whether histone deacetylase inhibitors (HDACIs), trichostatine-A (TSA), scriptaid (ST) and sodium butylate (SB) regulate the inflammatory response of DCs. Pre-treatment with TSA and ST reduced the syngeneic and allogeneic-antigen presenting activity of LPS-stimulated DCs in a dose dependent manner to statistical significance. SB also reduced the antigen presenting activity of DCs, but not significantly. HDACIs mediate their effects through the modulation of DC maturation and pre-treatment of the DCs with TSA or ST prior to treatment with LPS reduced the expressions of DC mature markers to the level of immature dendritic cells (iDCs). Moreover, TSA and ST reduced the IL-2 production from LPS-stimulated mature DCs. Our results suggest that HDACIs may actively modulate the DC-induced inflammatory response through inhibition of phenotypical or functional maturation.
Collapse
Affiliation(s)
- Eun Sun Kim
- Department of Biology Education, College of Education, Chungbuk National University, Cheongju, Chungbuk 361-763, Republic of Korea
| | | |
Collapse
|
49
|
Lu Y, Waller EK. Dichotomous role of interferon-gamma in allogeneic bone marrow transplant. Biol Blood Marrow Transplant 2009; 15:1347-53. [PMID: 19822293 DOI: 10.1016/j.bbmt.2009.07.015] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2009] [Accepted: 07/16/2009] [Indexed: 01/14/2023]
Abstract
Interferon (IFN)-gamma is a pleiotropic cytokine with a central role in innate and adaptive immunity. As a potent pro-inflammatory and antitumor cytokine, IFN-gamma is conventionally thought to be responsible for driving cellular immune response. On the other hand, accumulating evidence suggests that IFN-gamma also has immunosuppressive activity. An important role for IFN-gamma in inhibiting graft-versus-host disease (GVHD) has been demonstrated in murine models, despite IFN-gamma being one of the key factors amplifying T cell activation during the process of acute GVHD (aGVHD), the major complication and cause of post-transplant mortality in allogeneic bone marrow transplantation (BMT). At the same time, IFN-gamma facilitates graft-versus-leukemia (GVL) activity. Dissociation of GVL effects from GVHD has been the ultimate goal of allogeneic BMT in the treatment of hematologic malignancies. This paradoxic role of IFN-gamma makes modulating its activity a promising strategy to maximize GVL while minimizing GVHD and improve clinical outcomes in BMT. In this review, the effects of IFN-gamma on GVHD and GVL are discussed with consideration of the mechanism of IFN-gamma action.
Collapse
Affiliation(s)
- Ying Lu
- Department of Hematology/Oncology, Winship Cancer Institute, Emory University, Atlanta, Georgia 30322, USA
| | | |
Collapse
|
50
|
Abstract
The use of allogeneic hematopoietic cell transplantation (HCT) has increased as new techniques have been developed for transplantation in patients who previously would not have been considered HCT candidates. However, its efficacy continued to be limited by the development of frequent and severe acute GVHD. The complex and intricate pathophysiology of acute GVHD is a consequence of interactions between the donor and host innate and adaptive immune responses. Multiple inflammatory molecules and cell types are implicated in the development of GVHD that can be categorized as: (1) triggers that initiate GVHD by therapy-induced tissue damage and the antigen disparities between host and graft tissue; (2) sensors that detect the triggers, that is, process and present alloantigens; (3) mediators such as T-cell subsets (naive, memory, regulatory, Th17 and natural killer T cells) and (4) the effectors and amplifiers that cause damage of the target organs. These multiple inflammatory molecules and cell types that are implicated in the development of GVHD have been described with models that use stepwise cascades. Herein, we provide a novel perspective on the immunobiology of acute GVHD and briefly discuss some of the outstanding questions and limitations of the model systems.
Collapse
|