1
|
Singh RB, Cho W, Liu C, Naderi A, Surico PL, Kahale F, Dohlman TH, Chauhan SK, Dana R. Immunopathological mechanisms and clinical manifestations of ocular graft-versus-host disease following hematopoietic stem cell transplantation. Bone Marrow Transplant 2024; 59:1049-1056. [PMID: 38822141 DOI: 10.1038/s41409-024-02321-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Revised: 05/21/2024] [Accepted: 05/23/2024] [Indexed: 06/02/2024]
Abstract
Graft-versus-host disease is among the most common clinical complications following allogeneic hematopoietic stem cell transplantation. It causes inflammation-mediated destruction and dysfunction of various organ systems including ocular tissues in 60-90% of the patients and is termed ocular GVHD (oGVHD). In oGVHD, donor-derived T-cells recognize host antigens as foreign, resulting in immune dysregulation, inflammation and fibrosis of lacrimal glands, meibomian glands, cornea, and conjunctiva. The clinical presentation in oGVHD patients range from mild dry eye symptoms to catastrophic inflammation mediated pathological changes which can cause corneal perforation and blindness. In this review article, we provide detailed insights into the impact of mucosal barrier disruption, the afferent and efferent phases of immunological response involving activation of antigen presenting cells and T cells, respectively. We evaluate the evidence outlining the effector phase of the disease leading to cellular destruction and eventually fibrosis in patients with oGVHD. Finally, we discuss the well-established criteria for the diagnosis of oGVHD.
Collapse
Affiliation(s)
- Rohan Bir Singh
- Laboratory of Ocular Immunology, Transplantation and Regeneration, Schepens Eye Research Institute, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Wonkyung Cho
- Laboratory of Ocular Immunology, Transplantation and Regeneration, Schepens Eye Research Institute, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Catherine Liu
- Laboratory of Ocular Immunology, Transplantation and Regeneration, Schepens Eye Research Institute, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Amirreza Naderi
- Laboratory of Ocular Immunology, Transplantation and Regeneration, Schepens Eye Research Institute, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Pier Luigi Surico
- Laboratory of Ocular Immunology, Transplantation and Regeneration, Schepens Eye Research Institute, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Francesca Kahale
- Laboratory of Ocular Immunology, Transplantation and Regeneration, Schepens Eye Research Institute, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Thomas H Dohlman
- Laboratory of Ocular Immunology, Transplantation and Regeneration, Schepens Eye Research Institute, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Sunil K Chauhan
- Laboratory of Ocular Immunology, Transplantation and Regeneration, Schepens Eye Research Institute, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Reza Dana
- Laboratory of Ocular Immunology, Transplantation and Regeneration, Schepens Eye Research Institute, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
2
|
Ji H, Feng S, Liu Y, Cao Y, Lou H, Li Z. Effect of GVHD on the gut and intestinal microflora. Transpl Immunol 2024; 82:101977. [PMID: 38184214 DOI: 10.1016/j.trim.2023.101977] [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: 07/24/2023] [Revised: 12/08/2023] [Accepted: 12/31/2023] [Indexed: 01/08/2024]
Abstract
Graft-versus-host disease (GVHD) is one of the most important cause of death in patients undergoing allogeneic hematopoietic stem cell transplantation (allo-HSCT). The gastrointestinal tract is one of the most common sites affected by GVHD. However, there is no gold standard clinical practice for diagnosing gastrointestinal GVHD (GI-GVHD), and it is mainly diagnosed by the patient's clinical symptoms and related histological changes. Additionally, GI-GVHD causes intestinal immune system disorders, damages intestinal epithelial tissue such as intestinal epithelial cells((IEC), goblet, Paneth, and intestinal stem cells, and disrupts the intestinal epithelium's physical and chemical mucosal barriers. The use of antibiotics and diet alterations significantly reduces intestinal microbial diversity, further reducing bacterial metabolites such as short-chain fatty acids and indole, aggravating infection, and GI-GVHD. gut microbe diversity can be restored by fecal microbiota transplantation (FMT) to treat refractory GI-GVHD. This review article focuses on the clinical diagnosis of GI-GVHD and the effect of GVHD on intestinal flora and its metabolites.
Collapse
Affiliation(s)
- Hao Ji
- Department of Hematology, The First People's Hospital of Yunnan Province, Affiliated Hospital of Kunming University of Science and Technology, Kunming, China
| | - Shuai Feng
- Department of Hematology, The First People's Hospital of Yunnan Province, Affiliated Hospital of Kunming University of Science and Technology, Kunming, China; Yunnan Province Clinical Center for Hematologic Disease, The First People's Hospital of Yunnan Province, Kunming, China; Yunnan Blood Disease Hospital, The First People's Hospital of Yunnan Province, Kunming, China; National Key Clinical Specialty of Hematology, The First People's Hospital of Yunnan Province, Kunming, China; Yunnan Province Clinical Research Center for Hematologic Disease, The First People's Hospital of Yunnan Province, Kunming, China
| | - Yuan Liu
- Department of Hematology, The First People's Hospital of Yunnan Province, Affiliated Hospital of Kunming University of Science and Technology, Kunming, China
| | - Yue Cao
- Emergency of Department, Yunnan Provincial Hospital of Traditional Chinese Medicine, The First Affiliated Hospital of Yunnan University of Traditional Chinese Medicine, Kunming, China
| | - HuiQuan Lou
- Department of Oral and maxillofacial surgery, The First People's Hospital of Yunnan Province, Affiliated Hospital of Kunming University of Science and Technology, Kunming, China
| | - Zengzheng Li
- Department of Hematology, The First People's Hospital of Yunnan Province, Affiliated Hospital of Kunming University of Science and Technology, Kunming, China; Yunnan Province Clinical Center for Hematologic Disease, The First People's Hospital of Yunnan Province, Kunming, China; Yunnan Blood Disease Hospital, The First People's Hospital of Yunnan Province, Kunming, China; National Key Clinical Specialty of Hematology, The First People's Hospital of Yunnan Province, Kunming, China; Yunnan Province Clinical Research Center for Hematologic Disease, The First People's Hospital of Yunnan Province, Kunming, China.
| |
Collapse
|
3
|
Silva de Melo BM, Veras FP, Zwicky P, Lima D, Ingelfinger F, Martins TV, da Silva Prado D, Schärli S, Publio G, Hiroki CH, Melo PH, Saraiva A, Norbiato T, Lima L, Ryffel B, Vogl T, Roth J, Waisman A, Nakaya HI, da Silva Souza C, Cunha FQ, Cunha TM, Becher B, Alves-Filho JC. S100A9 Drives the Chronification of Psoriasiform Inflammation by Inducing IL-23/Type 3 Immunity. J Invest Dermatol 2023; 143:1678-1688.e8. [PMID: 36921684 DOI: 10.1016/j.jid.2023.02.026] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 01/13/2023] [Accepted: 02/09/2023] [Indexed: 03/14/2023]
Abstract
Psoriasis is a chronic inflammatory skin disorder driven by the IL-23/type 3 immune response. However, molecular mechanisms sustaining the chronicity of inflammation and psoriatic lesions remain elusive. Combining systematic analyses of several transcriptomic datasets, we delineated gene signatures across human psoriatic skin, identifying S100A9 as one of the most up-regulated genes, which was confirmed in lesioned skin from patients with psoriasis and preclinical psoriasiform skin inflammation models. Genetic ablation or pharmacologic inhibition of S100A9 alleviated Aldara-induced skin inflammation. By single-cell mapping of human psoriatic skin and bone marrow chimeric mice experiments, we identified keratinocytes as the major source of S100A9. Mechanistically, S100A9 induced IL-23 production by dendritic cells, driving the IL-23/type 3 immunity in psoriasiform skin inflammation. In addition, the cutaneous IL-23/IL-17 axis induced epidermal S100A9 expression in human and experimental psoriasis. Thus, we showed an autoregulatory circuit between keratinocyte-derived S100A9 and IL-23/type 3 immunity during psoriasiform inflammation, identifying a crucial function of S100A9 in the chronification of psoriasis.
Collapse
Affiliation(s)
- Bruno Marcel Silva de Melo
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil; Center for Research in Inflammatory Diseases, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Flávio Protásio Veras
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil; Center for Research in Inflammatory Diseases, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Pascale Zwicky
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Diógenes Lima
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Florian Ingelfinger
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Timna Varela Martins
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil; Center for Research in Inflammatory Diseases, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Douglas da Silva Prado
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil; Center for Research in Inflammatory Diseases, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Stefanie Schärli
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Gabriel Publio
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil; Center for Research in Inflammatory Diseases, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Carlos Hiroji Hiroki
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil; Center for Research in Inflammatory Diseases, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Paulo Henrique Melo
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil; Center for Research in Inflammatory Diseases, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
| | - André Saraiva
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil; Center for Research in Inflammatory Diseases, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Thainá Norbiato
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil; Center for Research in Inflammatory Diseases, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Leonardo Lima
- Center for Research in Inflammatory Diseases, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil; Department of Cell Biology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
| | | | - Thomas Vogl
- Institute of Immunology, University of Münster, Münster, Germany
| | - Johannes Roth
- Institute of Immunology, University of Münster, Münster, Germany
| | - Ari Waisman
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg, University of Mainz, Mainz, Germany
| | - Helder I Nakaya
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of Sao Paulo, Sao Paulo, Brazil; Hospital Israelita Albert Einstein, Sao Paulo, Brazil
| | - Cacilda da Silva Souza
- Department of Internal Medicine, Dermatology Division, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil, Ribeirao Preto, Sao Paulo, Brazil
| | - Fernando Q Cunha
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil; Center for Research in Inflammatory Diseases, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Thiago M Cunha
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil; Center for Research in Inflammatory Diseases, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Burkhard Becher
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - José C Alves-Filho
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil; Center for Research in Inflammatory Diseases, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil.
| |
Collapse
|
4
|
Buchanan CQ, Lawlor ML, Okafor C, Kurian SR, Philip AE, Finkle AE, McQuillan JJ, Haridas S, Koenig JM. Linked Th17 and Calgranulin Responses in Maternal-cord Blood Dyads of Preterm Gestations with Histologic Chorioamnionitis. NEWBORN (CLARKSVILLE, MD.) 2023; 2:133-141. [PMID: 37790838 PMCID: PMC10547109 DOI: 10.5005/jp-journals-11002-0064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/05/2023]
Abstract
Introduction Maternal-fetal immune crosstalk mechanisms are increasingly identified in the pathogenesis of gestational disorders, including histologic chorioamnionitis (HCA). Although an inflammatory Th17 immune phenotype has been described in preterm neonates with HCA, the associated maternal Th17 response is relatively unknown. To refine our understanding of Th17 biology in this context, we examined Th17 responses in maternal-cord blood dyads of preterm gestations. Materials and methods Paired maternal and cord blood (CB) samples were prospectively collected from preterm gestations (23-34 weeks) with HCA or controls. Th17-linked cell frequencies and plasma calgranulin (S100A8, S100A12) levels were determined by flow cytometry and enzyme-linked immunoassay, respectively. Results Analyses of 47 maternal-cord blood pairs showed striking parallel increases in Th17 cell frequencies as well as plasma calgranulin levels in the presence of fetal inflammation. Cord blood S100A12 levels were directly correlated with Th17 cell frequencies. In CB cultures, rh-S100A12 promoted in vitro propagation of Th17-type CD4+ cells. Conclusions Maternal and CB Th17-linked responses are dually amplified in gestations with HCA, supporting a biological role for maternal-fetal interactions in this disorder. In addition to advancing current knowledge of neonatal Th17 mechanisms, these data shed new light on their association with maternal inflammation.
Collapse
Affiliation(s)
- Christopher Q Buchanan
- Department of Obstetrics, Gynecology and Women’s Health, Division of Maternal–Fetal Medicine, Saint Louis University School of Medicine, St. Louis, Missouri, United States of America
| | - Megan L Lawlor
- Department of Obstetrics, Gynecology and Women’s Health, Division of Maternal–Fetal Medicine, Saint Louis University School of Medicine, St. Louis, Missouri, United States of America
| | - Chukwuebuka Okafor
- Department of Pediatrics, Division of Neonatal–Perinatal Medicine, Saint Louis University School of Medicine, St. Louis, Missouri, United States of America
| | - Shannon R Kurian
- Department of Pediatrics, Division of Neonatal–Perinatal Medicine, Saint Louis University School of Medicine, St. Louis, Missouri, United States of America
| | - Andrea E Philip
- Department of Pediatrics, Division of Neonatal–Perinatal Medicine, Saint Louis University School of Medicine, St. Louis, Missouri, United States of America
| | - Abigael E Finkle
- Department of Pediatrics, Division of Neonatal–Perinatal Medicine, Saint Louis University School of Medicine, St. Louis, Missouri, United States of America
| | - Jay J McQuillan
- Department of Pediatrics, Division of Neonatal–Perinatal Medicine, Saint Louis University School of Medicine, St. Louis, Missouri, United States of America
| | - Seema Haridas
- Department of Pediatrics, Division of Neonatal–Perinatal Medicine, Saint Louis University School of Medicine, St. Louis, Missouri, United States of America
| | - Joyce M Koenig
- Department of Pediatrics, Division of Neonatal–Perinatal Medicine, Saint Louis University School of Medicine, St. Louis, Missouri, United States of America
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, St. Louis, Missouri, United States of America
| |
Collapse
|
5
|
Li Q, Lan P. Activation of immune signals during organ transplantation. Signal Transduct Target Ther 2023; 8:110. [PMID: 36906586 PMCID: PMC10008588 DOI: 10.1038/s41392-023-01377-9] [Citation(s) in RCA: 32] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 02/10/2023] [Accepted: 02/15/2023] [Indexed: 03/13/2023] Open
Abstract
The activation of host's innate and adaptive immune systems can lead to acute and chronic graft rejection, which seriously impacts graft survival. Thus, it is particularly significant to clarify the immune signals, which are critical to the initiation and maintenance of rejection generated after transplantation. The initiation of response to graft is dependent on sensing of danger and stranger molecules. The ischemia and reperfusion of grafts lead to cell stress or death, followed by releasing a variety of damage-associated molecular patterns (DAMPs), which are recognized by pattern recognition receptors (PRRs) of host immune cells to activate intracellular immune signals and induce sterile inflammation. In addition to DAMPs, the graft exposed to 'non-self' antigens (stranger molecules) are recognized by the host immune system, stimulating a more intense immune response and further aggravating the graft damage. The polymorphism of MHC genes between different individuals is the key for host or donor immune cells to identify heterologous 'non-self' components in allogeneic and xenogeneic organ transplantation. The recognition of 'non-self' antigen by immune cells mediates the activation of immune signals between donor and host, resulting in adaptive memory immunity and innate trained immunity to the graft, which poses a challenge to the long-term survival of the graft. This review focuses on innate and adaptive immune cells receptor recognition of damage-associated molecular patterns, alloantigens and xenoantigens, which is described as danger model and stranger model. In this review, we also discuss the innate trained immunity in organ transplantation.
Collapse
Affiliation(s)
- Qingwen Li
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.,Key Laboratory of Organ Transplantation, Ministry of Education; NHC Key Laboratory of Organ Transplantation; Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, China
| | - Peixiang Lan
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China. .,Key Laboratory of Organ Transplantation, Ministry of Education; NHC Key Laboratory of Organ Transplantation; Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, China.
| |
Collapse
|
6
|
Dander E, Vinci P, Vetrano S, Recordati C, Piazza R, Fazio G, Bardelli D, Bugatti M, Sozio F, Piontini A, Bonanomi S, Bertola L, Tassistro E, Valsecchi MG, Calza S, Vermi W, Biondi A, Del Prete A, Sozzani S, D'Amico G. The chemerin/CMKLR1 axis regulates intestinal graft-versus-host disease. JCI Insight 2023; 8:154440. [PMID: 36883565 PMCID: PMC10077469 DOI: 10.1172/jci.insight.154440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Accepted: 01/23/2023] [Indexed: 03/09/2023] Open
Abstract
Gastrointestinal graft-versus-host disease (GvHD) is a major cause of mortality and morbidity following allogeneic bone marrow transplantation (allo-BMT). Chemerin is a chemotactic protein that recruits leukocytes to inflamed tissues by interacting with ChemR23/CMKLR1, a chemotactic receptor expressed by leukocytes, including macrophages. During acute GvHD, chemerin plasma levels were strongly increased in allo-BM-transplanted mice. The role of the chemerin/CMKLR1 axis in GvHD was investigated using Cmklr1-KO mice. WT mice transplanted with an allogeneic graft from Cmklr1-KO donors (t-KO) had worse survival and more severe GvHD. Histological analysis demonstrated that the gastrointestinal tract was the organ mostly affected by GvHD in t-KO mice. The severe colitis of t-KO mice was characterized by massive neutrophil infiltration and tissue damage associated with bacterial translocation and exacerbated inflammation. Similarly, Cmklr1-KO recipient mice showed increased intestinal pathology in both allogeneic transplant and dextran sulfate sodium-induced colitis. Notably, the adoptive transfer of WT monocytes into t-KO mice mitigated GvHD manifestations by decreasing gut inflammation and T cell activation. In patients, higher chemerin serum levels were predictive of GvHD development. Overall, these results suggest that CMKLR1/chemerin may be a protective pathway for the control of intestinal inflammation and tissue damage in GvHD.
Collapse
Affiliation(s)
- Erica Dander
- Tettamanti Center, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
| | - Paola Vinci
- Tettamanti Center, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
| | - Stefania Vetrano
- Laboratory of Gastrointestinal Immunopathology, Humanitas Clinical and Research Center, Rozzano, Italy.,Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy
| | - Camilla Recordati
- Department of Veterinary Medicine, University of Milan, Lodi, Italy.,Mouse and Animal Pathology Laboratory, Fondazione Unimi, Milan, Italy
| | - Rocco Piazza
- Department of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy.,Hematology Division and Bone Marrow Unit, San Gerardo Hospital, Monza, Italy
| | - Grazia Fazio
- Tettamanti Center, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
| | - Donatella Bardelli
- Tettamanti Center, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
| | - Mattia Bugatti
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Francesca Sozio
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy.,Humanitas Clinical and Research Center-IRCCS, Rozzano, Italy
| | - Andrea Piontini
- Laboratory of Gastrointestinal Immunopathology, Humanitas Clinical and Research Center, Rozzano, Italy.,Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy
| | - Sonia Bonanomi
- Pediatrics, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
| | - Luca Bertola
- Department of Veterinary Medicine, University of Milan, Lodi, Italy.,Mouse and Animal Pathology Laboratory, Fondazione Unimi, Milan, Italy
| | - Elena Tassistro
- Bicocca Center of Bioinformatics, Biostatistics and Bioimaging (B4 center), School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Maria Grazia Valsecchi
- Bicocca Center of Bioinformatics, Biostatistics and Bioimaging (B4 center), School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Stefano Calza
- Biostatistics, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - William Vermi
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Andrea Biondi
- Tettamanti Center, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy.,Pediatrics, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy.,School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Annalisa Del Prete
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy.,Humanitas Clinical and Research Center-IRCCS, Rozzano, Italy
| | - Silvano Sozzani
- Department of Molecular Medicine, Sapienza University of Rome, Laboratory affiliated to Istituto Pasteur Italia - Fondazione Cenci Bolognetti, Rome, Italy.,IRCCS Neuromed, Pozzilli, Italy
| | - Giovanna D'Amico
- Tettamanti Center, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
| |
Collapse
|
7
|
Møller DL, Kielsen K, Nielsen CH, Sengeløv H, Pedersen AE, Ryder LP, Müller K. Thymic stromal lymphopoietin levels after allogeneic hematopoietic stem cell transplantation. Immunopharmacol Immunotoxicol 2022; 44:1004-1012. [PMID: 35899395 DOI: 10.1080/08923973.2022.2102989] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND Thymic stromal lymphopoietin (TSLP) is an immunoregulatory, Th2-polarizing cytokine produced by epithelial cells. We hypothesized that TSLP affects immune reconstitution after hematopoietic stem cell transplantation (HSCT) leading to increased alloreactivity. METHODS We measured plasma TSLP by ELISA in 38 patients and assessed the immune reconstitution by flow cytometry. RESULTS TSLP levels rose after initiation of the conditioning to peak at day +21 after HSCT (p = .03), where TSLP levels correlated with counts of neutrophils (rho = 0.36, p = .04), monocytes (rho = 0.58, p = .006), and lymphocytes (rho = 0.59, p = .02). Overall absolute TSLP levels were not associated with acute or chronic graft-vs-host disease (a/cGvHD). However, patients mounting a sustained increase in TSLP levels at day +90 had a higher risk of cGvHD compared to patients who had returned to pre-conditioning levels at that stage (cumulative incidence: 77% vs. 38%, p = .01). CONCLUSION In conclusion, this study suggests a role of TSLP in immune reconstitution and alloreactivity post-HSCT. lymphopoietin (TSLP) is an immunoregulatory, Th2-polarizing cytokine produced by epithelial cells. We hypothesized that TSLP affects immune reconstitution after hematopoietic stem cell transplantation (HSCT) leading to increased alloreactivity. We measured plasma TSLP by ELISA in 38 patients and assessed the immune reconstitution by flow cytometry.
Collapse
Affiliation(s)
- Dina Leth Møller
- Department of Pediatrics and Adolescent Medicine, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark.,Institute for Inflammation Research, Department of Rheumatology and Spine Diseases, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Katrine Kielsen
- Department of Pediatrics and Adolescent Medicine, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark.,Institute for Inflammation Research, Department of Rheumatology and Spine Diseases, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Claus Henrik Nielsen
- Institute for Inflammation Research, Department of Rheumatology and Spine Diseases, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Henrik Sengeløv
- Department of Hematology, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | | | - Lars Peter Ryder
- The Tissue Typing Laboratory, Department of Clinical Immunology, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Klaus Müller
- Department of Pediatrics and Adolescent Medicine, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark.,Institute for Inflammation Research, Department of Rheumatology and Spine Diseases, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark.,Institute of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
8
|
Types of necroinflammation, the effect of cell death modalities on sterile inflammation. Cell Death Dis 2022; 13:423. [PMID: 35501340 PMCID: PMC9061831 DOI: 10.1038/s41419-022-04883-w] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 04/13/2022] [Accepted: 04/22/2022] [Indexed: 11/17/2022]
Abstract
Distinct types of immune responses are activated by infections, which cause the development of type I, II, or III inflammation, regulated by Th1, Th2, Th17 helper T cells and ILC1, ILC2 and ILC3 cells, respectively. While the classification of immune responses to different groups of pathogens is widely accepted, subtypes of the immune response elicited by sterile inflammation have not yet been detailed. Necroinflammation is associated with the release of damage-associated molecular patterns (DAMP) from dying cells. In this review, we present that the distinct molecular mechanisms activated during apoptosis, necroptosis, pyroptosis, and ferroptosis lead to the release of different patterns of DAMPs and their suppressors, SAMPs. We summarize the currently available data on how regulated cell death pathways and released DAMPs and SAMPs direct the differentiation of T helper and ILC cells. Understanding the subtypes of necroinflammation can be crucial in developing strategies for the treatment of sterile inflammatory diseases caused by cell death processes.
Collapse
|
9
|
Challenges and opportunities targeting mechanisms of epithelial injury and recovery in acute intestinal graft-versus-host disease. Mucosal Immunol 2022; 15:605-619. [PMID: 35654837 PMCID: PMC9259481 DOI: 10.1038/s41385-022-00527-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 04/21/2022] [Accepted: 05/16/2022] [Indexed: 02/04/2023]
Abstract
Despite advances in immunosuppressive prophylaxis and overall supportive care, gastrointestinal (GI) graft-versus-host disease (GVHD) remains a major, lethal side effect after allogeneic hematopoietic stem cell transplantation (allo-HSCT). It has become increasingly clear that the intestinal epithelium, in addition to being a target of transplant-related toxicity and GVHD, plays an important role in the onset of GVHD. Over the last two decades, increased understanding of the epithelial constituents and their microenvironment has led to the development of novel prophylactic and therapeutic interventions, with the potential to protect the intestinal epithelium from GVHD-associated damage and promote its recovery following insult. In this review, we will discuss intestinal epithelial injury and the role of the intestinal epithelium in GVHD pathogenesis. In addition, we will highlight possible approaches to protect the GI tract from damage posttransplant and to stimulate epithelial regeneration, in order to promote intestinal recovery. Combined treatment modalities integrating immunomodulation, epithelial protection, and induction of regeneration may hold the key to unlocking mucosal recovery and optimizing therapy for acute intestinal GVHD.
Collapse
|
10
|
Malengier-Devlies B, Metzemaekers M, Wouters C, Proost P, Matthys P. Neutrophil Homeostasis and Emergency Granulopoiesis: The Example of Systemic Juvenile Idiopathic Arthritis. Front Immunol 2021; 12:766620. [PMID: 34966386 PMCID: PMC8710701 DOI: 10.3389/fimmu.2021.766620] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2021] [Accepted: 11/23/2021] [Indexed: 12/21/2022] Open
Abstract
Neutrophils are key pathogen exterminators of the innate immune system endowed with oxidative and non-oxidative defense mechanisms. More recently, a more complex role for neutrophils as decision shaping cells that instruct other leukocytes to fine-tune innate and adaptive immune responses has come into view. Under homeostatic conditions, neutrophils are short-lived cells that are continuously released from the bone marrow. Their development starts with undifferentiated hematopoietic stem cells that pass through different immature subtypes to eventually become fully equipped, mature neutrophils capable of launching fast and robust immune responses. During severe (systemic) inflammation, there is an increased need for neutrophils. The hematopoietic system rapidly adapts to this increased demand by switching from steady-state blood cell production to emergency granulopoiesis. During emergency granulopoiesis, the de novo production of neutrophils by the bone marrow and at extramedullary sites is augmented, while additional mature neutrophils are rapidly released from the marginated pools. Although neutrophils are indispensable for host protection against microorganisms, excessive activation causes tissue damage in neutrophil-rich diseases. Therefore, tight regulation of neutrophil homeostasis is imperative. In this review, we discuss the kinetics of neutrophil ontogenesis in homeostatic conditions and during emergency myelopoiesis and provide an overview of the different molecular players involved in this regulation. We substantiate this review with the example of an autoinflammatory disease, i.e. systemic juvenile idiopathic arthritis.
Collapse
Affiliation(s)
- Bert Malengier-Devlies
- Department of Microbiology, Immunology and Transplantation, Laboratory of Immunobiology, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Mieke Metzemaekers
- Department of Microbiology, Immunology and Transplantation, Laboratory of Molecular Immunology, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Carine Wouters
- Department of Microbiology, Immunology and Transplantation, Laboratory of Immunobiology, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium.,Division of Pediatric Rheumatology, University Hospitals Leuven, Leuven, Belgium.,European Reference Network for Rare Immunodeficiency, Autoinflammatory and Autoimmune Diseases (RITA) at University Hospital Leuven, Leuven, Belgium
| | - Paul Proost
- Department of Microbiology, Immunology and Transplantation, Laboratory of Molecular Immunology, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Patrick Matthys
- Department of Microbiology, Immunology and Transplantation, Laboratory of Immunobiology, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| |
Collapse
|
11
|
Ara T, Hashimoto D. Novel Insights Into the Mechanism of GVHD-Induced Tissue Damage. Front Immunol 2021; 12:713631. [PMID: 34512636 PMCID: PMC8429834 DOI: 10.3389/fimmu.2021.713631] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2021] [Accepted: 08/10/2021] [Indexed: 12/22/2022] Open
Abstract
Prophylaxis for and treatment of graft-versus-host disease (GVHD) are essential for successful allogeneic hematopoietic stem cell transplantation (allo-SCT) and mainly consist of immunosuppressants such as calcineurin inhibitors. However, profound immunosuppression can lead to tumor relapse and infectious complications, which emphasizes the necessity of developing novel management strategies for GVHD. Emerging evidence has revealed that tissue-specific mechanisms maintaining tissue homeostasis and promoting tissue tolerance to combat GVHD are damaged after allo-SCT, resulting in exacerbation and treatment refractoriness of GVHD. In the gastrointestinal tract, epithelial regeneration derived from intestinal stem cells (ISCs), a microenvironment that maintains healthy gut microbiota, and physical and chemical mucosal barrier functions against pathogens are damaged by conditioning regimens and/or GVHD. The administration of growth factors for cells that maintain intestinal homeostasis, such as interleukin-22 (IL-22) for ISCs, R-spondin 1 (R-Spo1) for ISCs and Paneth cells, and interleukin-25 (IL-25) for goblet cells, mitigates murine GVHD. In this review, we summarize recent advances in the understanding of GVHD-induced tissue damage and emerging strategies for the management of GVHD.
Collapse
Affiliation(s)
- Takahide Ara
- Department of Hematology, Hokkaido University Faculty of Medicine, Sapporo, Japan
| | - Daigo Hashimoto
- Department of Hematology, Hokkaido University Faculty of Medicine, Sapporo, Japan
| |
Collapse
|
12
|
The development of ruxolitinib for glucocorticoid-refractory acute graft-versus-host disease. Blood Adv 2021; 4:3789-3794. [PMID: 32780849 DOI: 10.1182/bloodadvances.2020002097] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 07/13/2020] [Indexed: 02/06/2023] Open
Abstract
Acute graft-versus-host disease (aGVHD) is a life-threatening complication after allogeneic hematopoietic cell transplantation (allo-HCT). Despite the use of prophylactic immunosuppression including calcineurin inhibitors, antimetabolites, antithymocyte globulin, or posttransplant cyclophosphamide, patients still develop severe aGVHD. In particular, patients with glucocorticoid-refractory GVHD (SR-GVHD) have a dismal prognosis with a low 1-year post-allo-HCT survival rate. Most classical drugs used to prevent or treat aGVHD target 1 specific pathway such as calcineurin inhibitors or mammalian target of rapamycin inhibitors, or they interfere with fast-dividing activated cells (eg, methotrexate, mycophenolate, and cyclophosphamide). In contrast to these drugs, inhibition-of-signaling molecules, used by multiple immune cells and critical for signal transduction of multiple proinflammatory cytokines, could be more efficacious at blocking GVHD. Ruxolitinib blocks Janus kinases 1 and 2, which are required to mediate the downstream signaling of multiple cytokine receptors. Recently, a multicenter phase 3 clinical trial showed that ruxolitinib led to significant improvements in efficacy outcomes compared to best available therapy, which will lead to a paradigm shift in the treatment of SR-GVHD.
Collapse
|
13
|
Jardine L, Cytlak U, Gunawan M, Reynolds G, Green K, Wang XN, Pagan S, Paramitha M, Lamb CA, Long AK, Hurst E, Nair S, Jackson GH, Publicover A, Bigley V, Haniffa M, Simpson AJ, Collin M. Donor monocyte-derived macrophages promote human acute graft-versus-host disease. J Clin Invest 2021; 130:4574-4586. [PMID: 32453711 PMCID: PMC7456218 DOI: 10.1172/jci133909] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Accepted: 05/19/2020] [Indexed: 12/16/2022] Open
Abstract
Myelopoiesis is invariably present and contributes to pathology in animal models of graft-versus-host disease (GVHD). In humans, a rich inflammatory infiltrate bearing macrophage markers has also been described in histological studies. In order to determine the origin, functional properties, and role in pathogenesis of these cells, we isolated single-cell suspensions from acute cutaneous GVHD and subjected them to genotype, transcriptome, and in vitro functional analysis. A donor-derived population of CD11c+CD14+ cells was the dominant population of all leukocytes in GVHD. Surface phenotype and NanoString gene expression profiling indicated the closest steady-state counterpart of these cells to be monocyte-derived macrophages. In GVHD, however, there was upregulation of monocyte antigens SIRPα and S100A8/9 transcripts associated with leukocyte trafficking, pattern recognition, antigen presentation, and costimulation. Isolated GVHD macrophages stimulated greater proliferation and activation of allogeneic T cells and secreted higher levels of inflammatory cytokines than their steady-state counterparts. In HLA-matched mixed leukocyte reactions, we also observed differentiation of activated macrophages with a similar phenotype. These exhibited cytopathicity to a keratinocyte cell line and mediated pathological damage to skin explants independently of T cells. Together, these results define the origin, functional properties, and potential pathogenic roles of human GVHD macrophages.
Collapse
Affiliation(s)
- Laura Jardine
- Human Dendritic Cell Laboratory, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom.,Northern Centre for Bone Marrow Transplantation and.,NIHR Newcastle Biomedical Research Centre, Freeman Hospital, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom
| | - Urszula Cytlak
- Human Dendritic Cell Laboratory, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Merry Gunawan
- Human Dendritic Cell Laboratory, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Gary Reynolds
- NIHR Newcastle Biomedical Research Centre, Freeman Hospital, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom.,Institute of Cellular Medicine and
| | - Kile Green
- Human Dendritic Cell Laboratory, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom
| | | | - Sarah Pagan
- Human Dendritic Cell Laboratory, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Maharani Paramitha
- Human Dendritic Cell Laboratory, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Christopher A Lamb
- NIHR Newcastle Biomedical Research Centre, Freeman Hospital, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom.,Institute of Cellular Medicine and
| | - Anna K Long
- NIHR Newcastle Biomedical Research Centre, Freeman Hospital, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom.,Institute of Cellular Medicine and
| | - Erin Hurst
- Northern Centre for Bone Marrow Transplantation and
| | - Smeera Nair
- Northern Centre for Bone Marrow Transplantation and
| | - Graham H Jackson
- Northern Centre for Bone Marrow Transplantation and.,Northern Institute of Cancer Research, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Amy Publicover
- Human Dendritic Cell Laboratory, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom.,Northern Centre for Bone Marrow Transplantation and.,NIHR Newcastle Biomedical Research Centre, Freeman Hospital, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom
| | - Venetia Bigley
- Human Dendritic Cell Laboratory, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom.,Northern Centre for Bone Marrow Transplantation and.,NIHR Newcastle Biomedical Research Centre, Freeman Hospital, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom
| | - Muzlifah Haniffa
- NIHR Newcastle Biomedical Research Centre, Freeman Hospital, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom.,Institute of Cellular Medicine and
| | - A J Simpson
- NIHR Newcastle Biomedical Research Centre, Freeman Hospital, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom.,Institute of Cellular Medicine and
| | - Matthew Collin
- Human Dendritic Cell Laboratory, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom.,Northern Centre for Bone Marrow Transplantation and.,NIHR Newcastle Biomedical Research Centre, Freeman Hospital, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom
| |
Collapse
|
14
|
Giaccone L, Faraci DG, Butera S, Lia G, Di Vito C, Gabrielli G, Cerrano M, Mariotti J, Dellacasa C, Felicetti F, Brignardello E, Mavilio D, Bruno B. Biomarkers for acute and chronic graft versus host disease: state of the art. Expert Rev Hematol 2020; 14:79-96. [PMID: 33297779 DOI: 10.1080/17474086.2021.1860001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
INTRODUCTION Despite significant advances in treatment and prevention, graft-versus-host disease (GVHD) still represents the main cause of morbidity and mortality after allogeneic hematopoietic stem cell transplantation. Thus, considerable research efforts have been made to find and validate reliable biomarkers for diagnosis, prognosis, and risk stratification of GVHD. AREAS COVERED In this review the most recent evidences on different types of biomarkers studied for GVHD, such as genetic, plasmatic, cellular markers, and those associated with microbiome, were summarized. A comprehensive search of peer-review literature was performed in PubMed including meta-analysis, preclinical and clinical trials, using the terms: cellular and plasma biomarkers, graft-versus-host disease, cytokines, and allogeneic hematopoietic stem cell transplantation. EXPERT OPINION In the near future, several validated biomarkers will be available to help clinicians in the diagnosis of GVHD, the identification of patients at high risk of GVHD development and in patients' stratification according to its severity. Then, immunosuppressive treatment could be tailored to each patient's real needs. However, more efforts are needed to achieve this goal. Although most of the proposed biomarkers currently lack validation with large-scale clinical data, their study led to improved knowledge of the biological basis of GVHD, and ultimately to implementation of GHVD treatment.
Collapse
Affiliation(s)
- Luisa Giaccone
- Department of Oncology/Hematology, Stem Cell Transplant Program, A.O.U. Città Della Salute E Della Scienza Di Torino, Presidio Molinette , Torino, Italy.,Department of Molecular Biotechnology and Health Sciences, University of Torino , Torino, Italy
| | - Danilo Giuseppe Faraci
- Department of Oncology/Hematology, Stem Cell Transplant Program, A.O.U. Città Della Salute E Della Scienza Di Torino, Presidio Molinette , Torino, Italy.,Department of Molecular Biotechnology and Health Sciences, University of Torino , Torino, Italy
| | - Sara Butera
- Department of Oncology/Hematology, Stem Cell Transplant Program, A.O.U. Città Della Salute E Della Scienza Di Torino, Presidio Molinette , Torino, Italy.,Department of Molecular Biotechnology and Health Sciences, University of Torino , Torino, Italy
| | - Giuseppe Lia
- Department of Oncology/Hematology, Stem Cell Transplant Program, A.O.U. Città Della Salute E Della Scienza Di Torino, Presidio Molinette , Torino, Italy.,Department of Molecular Biotechnology and Health Sciences, University of Torino , Torino, Italy
| | - Clara Di Vito
- Unit of Clinical and Experimental Immunology, Humanitas Clinical and Research Center , Milan, Italy.,Department of Medical Biotechnologies and Translational Medicine (Biometra), University of Milan , Milan, Italy
| | - Giulia Gabrielli
- Department of Oncology/Hematology, Stem Cell Transplant Program, A.O.U. Città Della Salute E Della Scienza Di Torino, Presidio Molinette , Torino, Italy.,Department of Molecular Biotechnology and Health Sciences, University of Torino , Torino, Italy
| | - Marco Cerrano
- Department of Oncology/Hematology, Stem Cell Transplant Program, A.O.U. Città Della Salute E Della Scienza Di Torino, Presidio Molinette , Torino, Italy.,Department of Molecular Biotechnology and Health Sciences, University of Torino , Torino, Italy
| | - Jacopo Mariotti
- Bone Marrow Transplant Unit, Humanitas Clinical and Research Center, IRCCS , Rozzano, Italy
| | - Chiara Dellacasa
- Department of Oncology/Hematology, Stem Cell Transplant Program, A.O.U. Città Della Salute E Della Scienza Di Torino, Presidio Molinette , Torino, Italy
| | - Francesco Felicetti
- Transition Unit for Childhood Cancer Survivors, A.O.U. Città Della Salute E Della Scienza Di Torino , University of Torino , Torino, Italy
| | - Enrico Brignardello
- Transition Unit for Childhood Cancer Survivors, A.O.U. Città Della Salute E Della Scienza Di Torino , University of Torino , Torino, Italy
| | - Domenico Mavilio
- Unit of Clinical and Experimental Immunology, Humanitas Clinical and Research Center , Milan, Italy.,Department of Medical Biotechnologies and Translational Medicine (Biometra), University of Milan , Milan, Italy
| | - Benedetto Bruno
- Department of Oncology/Hematology, Stem Cell Transplant Program, A.O.U. Città Della Salute E Della Scienza Di Torino, Presidio Molinette , Torino, Italy.,Department of Molecular Biotechnology and Health Sciences, University of Torino , Torino, Italy
| |
Collapse
|
15
|
Rafei H, Jenq RR. Microbiome-intestine cross talk during acute graft-versus-host disease. Blood 2020; 136:401-409. [PMID: 32526029 PMCID: PMC7378453 DOI: 10.1182/blood.2019000950] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Accepted: 05/06/2020] [Indexed: 02/08/2023] Open
Abstract
Allogeneic hematopoietic stem cell transplantation (allo-SCT) offers cure for a variety of conditions, in particular, but not limited to, hematologic malignancies. However, it can be associated with life-threatening complications, including graft-versus-host disease (GVHD) and infections, which are factors limiting its widespread use. Technical advances in the field of microbiome research have allowed for a better understanding of the microbial flora of the human intestine, as well as dissection of their interactions with the host immune system in allo-SCT and posttransplant complications. There is growing evidence that the commensal microbiome is frequently dysregulated following allo-SCT and that this dysbiosis can predispose to adverse clinical outcomes, especially including acute intestinal GVHD and reduced overall survival. In this review, we discuss the interactions between the microbiome and the components of the immune system that play a major role in the pathways leading to the inflammatory state of acute intestinal GVHD. We also discuss the microbiome-centered strategies that have been devised or are actively being investigated to improve the outcomes of allo-SCT patients in regard to acute intestinal GVHD.
Collapse
Affiliation(s)
| | - Robert R Jenq
- Department of Genomic Medicine, and
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX; and
- Cancer Prevention and Research Institute of Texas, Houston, TX
| |
Collapse
|
16
|
Shabbir E, Farooq U, Yanes B, Magalhaes-Silverman M. Repeat Endoscopy Affects Patient Management in Gastrointestinal Graft- Versus-Host Disease. Clin Hematol Int 2020; 2:69-73. [PMID: 34595445 PMCID: PMC8432351 DOI: 10.2991/chi.d.200220.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Accepted: 02/08/2020] [Indexed: 11/24/2022] Open
Abstract
Graft versus host disease (GVHD) of the gut is associated with significant morbidity and mortality after allogeneic hematopoietic cell transplant (allo-HCT). No guidelines exist regarding repeat endoscopy after failure of first-line treatment with steroids. We aimed to study if repeat endoscopic biopsy can be helpful in these patients to guide treatment decisions. We retrospectively reviewed medical records of all patients who underwent repeat endoscopy for clinical suspicion of gastrointestinal (GI) GVHD after allo-HCT. Of the 318 patients, 24 underwent endoscopy twice after allo-HCT. At first endoscopy, 20 patients (80%) showed abnormal findings: 16 with GVHD alone, 1 with GVHD plus cytomegalovirus (CMV), and 3 with GVHD plus infectious colitis. On repeat endoscopy in these 20 patients with GVHD, 6 showed improvement leading to de-escalation of therapy, 8 showed worsening of GVHD including detection of CMV in 2 patients, and 2 had no histological changes. One patient with simultaneous GVHD and CMV diagnosed on first biopsy, displayed significant improvement leading to de-escalation of therapy. Three patients with GVHD along with infectious colitis on biopsy subsequently showed improvement on repeat biopsy leading to de-escalation of therapy. Among 4 patients with normal findings on first endoscopy, 3 had GVHD and 1 had epstein-barrvirus-associated post-transplant lymphoproliferative disorder (EBV-PTLD) on repeat procedures. This study supports the usefulness of repeat endoscopy in persistently symptomatic patients when there is no improvement after the initial treatment based on the results of the first endoscopy. Repeat endoscopy may guide therapy without significant complications.
Collapse
Affiliation(s)
- Ehsan Shabbir
- Department of Internal Medicine, Division of Hematology, Oncology, and Blood and Marrow Transplantation, University of Iowa Hospitals and Clinics, Iowa City, Iowa, USA.,Department of Internal Medicine and Neurology, Wright State University, Dayton, Ohio, USA
| | - Umar Farooq
- Department of Internal Medicine, Division of Hematology, Oncology, and Blood and Marrow Transplantation, University of Iowa Hospitals and Clinics, Iowa City, Iowa, USA
| | - Burhan Yanes
- Department of Hematology and Oncology, Wright State University, Dayton, Ohio, USA
| | - Margarida Magalhaes-Silverman
- Department of Internal Medicine, Division of Hematology, Oncology, and Blood and Marrow Transplantation, University of Iowa Hospitals and Clinics, Iowa City, Iowa, USA
| |
Collapse
|
17
|
Kessel C, Hedrich CM, Foell D. Innately Adaptive or Truly Autoimmune: Is There Something Unique About Systemic Juvenile Idiopathic Arthritis? Arthritis Rheumatol 2020; 72:210-219. [PMID: 31524322 DOI: 10.1002/art.41107] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Accepted: 09/10/2019] [Indexed: 12/11/2022]
Abstract
Systemic juvenile idiopathic arthritis (JIA) is a form of arthritis in childhood that is initially dominated by innate immunity-driven systemic inflammation and is thus considered a polygenic autoinflammatory disease. However, systemic JIA can progress toward an adaptive immunity-driven afebrile arthritis. Based on this observation of biphasic disease progression, a "window of opportunity" for optimal, individualized and target-directed treatment has been proposed. This hypothesis requires testing, and in this review we summarize current evidence regarding molecular factors that may contribute to the progression from an initially predominantly autoinflammatory disease phenotype to autoimmune arthritis. We consider the involvement of innately adaptive γδ T cells and natural killer T cells that express γδ or αβ T cell receptors but cannot be classified as either purely innate or adaptive cells, versus classic B and T lymphocytes in this continuum. Finally, we discuss our understanding of how and why some primarily autoinflammatory conditions can progress toward autoimmune-mediated disorders over the disease course while others do not and how this knowledge may be used to offer individualized treatment.
Collapse
|
18
|
Increased regulatory T cells and eosinophils characterize atopic dermatitis-like graft-versus-host disease compared with lichen planus-like graft-versus-host disease. J Am Acad Dermatol 2019; 83:824-831. [PMID: 31404571 DOI: 10.1016/j.jaad.2019.08.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Revised: 07/28/2019] [Accepted: 08/03/2019] [Indexed: 02/06/2023]
Abstract
BACKGROUND Graft-versus-host disease (GVHD) has various cutaneous manifestations. Little is known about the mechanisms of cutaneous GVHD with different clinical features. OBJECTIVE To characterize the immunologic features and skin barrier functions of cutaneous GVHD. METHODS The study included 19 patients with atopic dermatitis (AD)-like GVHD, 8 with lichen planus (LP)-like GVHD, 24 with AD, and 15 healthy controls. The subpopulation of T cells in peripheral blood and skin lesions was measured by flow cytometry and immunofluorescence, respectively. Filaggrin expression in skin lesions was measured by Western blot and immunohistochemistry. Transepidermal water loss was also measured using Tewameter TM 300 (Courage & Khazaka Electronic GmbH, Köln, Germany). RESULTS The number of peripheral blood eosinophils in AD-like GVHD was significantly higher than that in LP-like GVHD. Type 2 helper T cells in peripheral blood and skin lesions were increased in AD-like GVHD and LP-like GVHD. Regulatory T cells in peripheral blood and skin lesions were increased in AD-like GVHD. Filaggrin expression and transepidermal water loss were increased in skin lesions of AD-like GVHD and LP-like GVHD. LIMITATIONS The number of patients is limited. CONCLUSION Although AD-like GVHD and LP-like GVHD both had elevated type 2 helper T cells and impaired skin barrier, increased eosinophils and regulatory T cells were found only in AD-like GVHD.
Collapse
|
19
|
Han L, Zhang H, Chen S, Zhou L, Li Y, Zhao K, Huang F, Fan Z, Xuan L, Zhang X, Dai M, Lin Q, Jiang Z, Peng J, Jin H, Liu Q. Intestinal Microbiota Can Predict Acute Graft-versus-Host Disease Following Allogeneic Hematopoietic Stem Cell Transplantation. Biol Blood Marrow Transplant 2019; 25:1944-1955. [PMID: 31299215 DOI: 10.1016/j.bbmt.2019.07.006] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Revised: 06/23/2019] [Accepted: 07/03/2019] [Indexed: 02/06/2023]
Abstract
The intestinal microbiome plays an important role in the development of acute graft-versus-host disease (aGVHD). However, whether intestinal microbiota can predict the development of aGVHD has been reported only rarely. Here we conducted a prospective study of microbiota in 141 patients after allogeneic hematopoietic stem cell transplantation. We found lower microbiota diversity in the aGVHD group compared with the non-aGVHD group at day 0 and day 15 ± 1 (P = .018 and .009, respectively). Diversity was negatively associated with conditioning intensity (P = .017, day 0; P = .045, day 15) and β-lactam antibiotic administration (P = .004, day 15). Intensified conditioning and β-lactam antibiotics were associated with a lower regulatory T (Treg)/T helper 17 (Th17) cell ratio at day 15 (P = .030 and .047, respectively). At day 15, the levels of the inflammatory factors (tumor necrosis factor α, interleukin [IL]-6, IL-17A, IL-1β, and lipopolysaccharide) were higher in the intensified conditioning group compared with the standard group (P < .05). The accumulated intestinal microbiota (AIM) score was defined as microbiota diversity and gradient of the 4 bacterials (Lachnospiraceae, Peptostreptococcaceae, Erysipelotrichaceae, and Enterobacteriaceae) at day 15 post-transplantation. The AIM score was positively correlated with aGVHD grade (r = .481, P < .001), and the AIM score could be predictive of the development of aGVHD (grade II-IV aGVHD: area under the curve [AUC], .75, P < .001; grade III-IV aGVHD: AUC, .84, P < .001). These findings suggest that intestinal microbiota and conditioning might induce aGVHD by inflammatory factors and the Treg/Th17 balance. The constitution of the intestinal microbiota at neutrophil engraftment may predict the development of aGVHD.
Collapse
Affiliation(s)
- Lijie Han
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China; Department of Hematology, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Haiyan Zhang
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Shan Chen
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Lizhi Zhou
- Department of Biostatistics, School of Public Health, Southern Medical University, Guangzhou, China
| | - Yuanyuan Li
- School of Foreign Languages, Henan University of Chinese Medicine, Zhengzhou, China
| | - Ke Zhao
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Fen Huang
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zhiping Fan
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Li Xuan
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xin Zhang
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Min Dai
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Qianyun Lin
- Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zhongxing Jiang
- Department of Hematology, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jie Peng
- Department of Oncology, Second Affiliated Hospital of Guizhou Medical University, Kaili, China; Hepatology Unit and Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Hua Jin
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| | - Qifa Liu
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China; Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Southern Medical University, Guangzhou, China.
| |
Collapse
|
20
|
Mesenchymal Stem Cell Therapy Overcomes Steroid Resistance in Severe Gastrointestinal Acute Graft-Versus-Host Disease. Case Rep Transplant 2019; 2019:7890673. [PMID: 31263624 PMCID: PMC6556259 DOI: 10.1155/2019/7890673] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2019] [Accepted: 05/14/2019] [Indexed: 12/27/2022] Open
Abstract
The authors describe the high effectiveness of human mesenchymal stem cell (hMSC) therapy to treat steroid-refractory gastrointestinal acute graft-versus-host Disease (aGVHD) in a 15-year-old boy with acute lymphoblastic leukemia (ALL). He received allogeneic hematopoietic stem cell transplantation due to high-risk hypodiploid ALL. Around the time of engraftment, he developed severe diarrhea following high-grade fever and erythema. Although methylprednisolone pulse therapy was added to tacrolimus and mycophenolate mofetil, diarrhea progressed up to 5000~6000 ml/day and brought about hypocalcemia, hypoalbuminemia, and edema. Daily fresh frozen plasma (FFP), albumin, and calcium replacements were required to maintain blood circulation. After aGVHD was confirmed by colonoscopic biopsy, MSC therapy was administered. The patient received 8 biweekly intravenous infusions of 2×106 hMSCs/kg for 4 weeks, after which additional 4 weekly infusions were performed. A few weeks after initiation, diarrhea gradually resolved, and at the eighth dose of hMSC, lab data improved without replacements. MSC therapy successfully treated steroid-refractory gastrointestinal GVHD without complications. Despite life-threatening diarrhea, the regeneration potential of children and adolescents undergoing SMC therapy successfully supports restoration of gastrointestinal damage. Even with its high treatment costs, SMC therapy should be proactively considered in cases where young patients suffer from severe gastrointestinal GVHD.
Collapse
|
21
|
Zeiser R. Biology-driven developments in the therapy of acute graft-versus-host disease. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2018; 2018:236-241. [PMID: 30504316 PMCID: PMC6245989 DOI: 10.1182/asheducation-2018.1.236] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Allogeneic hematopoietic cell transplantation is a potentially curative treatment of different hematological malignancies. A major life-threatening complication is acute graft-versus-host disease (GVHD), in particular when the disease becomes steroid refractory. Based on the detection of pathogenic cytokines, chemokines, and T-cell subsets in individuals developing GVHD or experimental GVHD models, different therapeutic strategies have been developed. A potential cause why targeting individual receptors can lack efficacy could be that multiple cytokines, danger signals, and chemokine that have redundant functions are released during GVHD. To overcome this redundancy, novel strategies that do not target individual surface molecules like chemokine receptors, integrins, and cytokine receptors, but instead inhibit signaling pathways downstream of these molecules, have been tested in preclinical GVHD models and are currently being tested in clinical GVHD trials. Another important development is tissue regenerative approaches that promote healing of GVHD-related tissue damage as well as strategies that rely on microbiota modifications. These approaches are promising because they act very differently from conventional immunosuppression, instead aiming at reinstalling tissue homeostasis and microbiome diversity. This review discusses major novel developments in GVHD therapy that are based on a better understanding of GVHD biology, the repurposing of novel kinase inhibitors, microbiome modification strategies, and tissue-regenerative approaches.
Collapse
Affiliation(s)
- Robert Zeiser
- Department of Hematology, Oncology and Stem Cell Transplantation, Faculty of Medicine, Freiburg University Medical Center, Freiburg, Germany
| |
Collapse
|
22
|
Wiese F, Reinhardt-Heller K, Volz M, Gille C, Köstlin N, Billing H, Handgretinger R, Holzer U. Monocytes show immunoregulatory capacity on CD4 + T cells in a human in-vitro model of extracorporeal photopheresis. Clin Exp Immunol 2018; 195:369-380. [PMID: 30411330 DOI: 10.1111/cei.13232] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/09/2018] [Indexed: 01/02/2023] Open
Abstract
Extracorporeal photopheresis (ECP) is a widely used immunomodulatory therapy for the treatment of various T cell-mediated disorders such as cutaneous T cell lymphoma (CTCL), graft-versus-host disease (GvHD) or systemic sclerosis. Although clinical benefits of ECP are already well described, the underlying mechanism of action of ECP is not yet fully understood. Knowledge on the fate of CD14+ monocytes in the context of ECP is particularly limited and controversial. Here, we investigated the immunoregulatory function of ECP treated monocytes on T cells in an in-vitro ECP model. We show that ECP-treated monocytes significantly induce proinflammatory T cell types in co-cultured T cells, while anti-inflammatory T cells remain unaffected. Furthermore, we found significantly reduced proliferation rates of T cells after co-culture with ECP-treated monocytes. Both changes in interleukin secretion and proliferation were dependent on cell-contact between monocytes and T cells. Interestingly, blocking interactions of programmed death ligand 1 (PD-L1) to programmed death 1 (PD-1) in the in-vitro model led to a significant recovery of T cell proliferation. These results set the base for further studies on the mechanism of ECP, especially the regulatory role of ECP-treated monocytes.
Collapse
Affiliation(s)
- F Wiese
- Tuebingen University Children's Hospital, Department of Hematology and Oncology, Tuebingen, Germany
| | - K Reinhardt-Heller
- Tuebingen University Children's Hospital, Department of Hematology and Oncology, Tuebingen, Germany
| | - M Volz
- Tuebingen University Children's Hospital, Department of Hematology and Oncology, Tuebingen, Germany
| | - C Gille
- Tuebingen University Children's Hospital, Department of Neonatology, Tuebingen, Germany
| | - N Köstlin
- Tuebingen University Children's Hospital, Department of Neonatology, Tuebingen, Germany
| | - H Billing
- Tuebingen University Children's Hospital, Department of Hematology and Oncology, Tuebingen, Germany
| | - R Handgretinger
- Tuebingen University Children's Hospital, Department of Hematology and Oncology, Tuebingen, Germany
| | - U Holzer
- Tuebingen University Children's Hospital, Department of Hematology and Oncology, Tuebingen, Germany
| |
Collapse
|
23
|
Brenner AK, Bruserud Ø. S100 Proteins in Acute Myeloid Leukemia. Neoplasia 2018; 20:1175-1186. [PMID: 30366122 PMCID: PMC6215056 DOI: 10.1016/j.neo.2018.09.007] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Revised: 09/24/2018] [Accepted: 09/27/2018] [Indexed: 01/02/2023] Open
Abstract
The S100 protein family contains 20 functionally expressed members, which are commonly dysregulated in cancer. Their wide range of functions includes cell proliferation, cell differentiation, regulation of transcription factors, inflammation, chemotaxis, and angiogenesis. S100 proteins have in several types of cancer proven to be biomarkers for disease progression and prognosis. Acute myeloid leukemia (AML) is a highly heterogeneous and aggressive disease in which immature myeloblasts replace normal hematopoietic cells in the bone marrow. This review focuses on the S100 protein family members, which commonly are dysregulated in AML, and on the consequences of their dysregulation in the disorder. Like in other cancers, it appears as if S100 proteins are potential biomarkers for leukemogenesis. Furthermore, several S100 members seem to be involved in maintaining the leukemic phenotype. For these reasons, specific S100 proteins might serve as prognostic biomarkers, especially in the patient subset with intermediate/undetermined risk, and as potential targets for patient-adjusted therapy. Because the question of the most suitable candidate S100 biomarkers in AML still is under discussion, because particular AML subgroups lead to specific S100 signatures, and because downstream effects and the significance of co-expression of potential S100 binding partners in AML are not fully elucidated yet, we conclude that a panel of S100 proteins will probably be best suited for prognostic purposes.
Collapse
Affiliation(s)
- Annette K Brenner
- Department of Medicine, Haukeland University Hospital, P.O. Box 1400, 5021 Bergen, Norway; Section for Hematology, Department of Clinical Science, University of Bergen, P.O. Box 7804, 5020 Bergen, Norway
| | - Øystein Bruserud
- Department of Medicine, Haukeland University Hospital, P.O. Box 1400, 5021 Bergen, Norway; Section for Hematology, Department of Clinical Science, University of Bergen, P.O. Box 7804, 5020 Bergen, Norway.
| |
Collapse
|
24
|
Holzinger D, Foell D, Kessel C. The role of S100 proteins in the pathogenesis and monitoring of autoinflammatory diseases. Mol Cell Pediatr 2018; 5:7. [PMID: 30255357 PMCID: PMC6156694 DOI: 10.1186/s40348-018-0085-2] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2017] [Accepted: 04/27/2018] [Indexed: 01/09/2023] Open
Abstract
S100A8/A9 and S100A12 are released from activated monocytes and granulocytes and act as proinflammatory endogenous toll-like receptor (TLR)4-ligands. S100 serum concentrations correlate with disease activity, both during local and systemic inflammatory processes. In some autoinflammatory diseases such as familial Mediterranean fever (FMF) or systemic juvenile idiopathic arthritis (SJIA), dysregulation of S100 release may be involved in the pathogenesis. Moreover, S100 serum levels are a valuable supportive tool in the diagnosis of SJIA in fever of unknown origin. Furthermore, S100 levels can be used to monitor disease activity to subclinical level, as their serum concentrations decrease with successful treatment.
Collapse
Affiliation(s)
- Dirk Holzinger
- Department of Pediatric Hematology-Oncology, University of Duisburg-Essen, Hufelandstr. 55, 45122, Essen, Germany.
| | - Dirk Foell
- Department of Pediatric Rheumatology and Immunology, University Children's Hospital Muenster, Domagkstr. 3, 48149, Muenster, Germany
| | - Christoph Kessel
- Department of Pediatric Rheumatology and Immunology, University Children's Hospital Muenster, Domagkstr. 3, 48149, Muenster, Germany
| |
Collapse
|
25
|
Romano M, Fanelli G, Tan N, Nova-Lamperti E, McGregor R, Lechler RI, Lombardi G, Scottà C. Expanded Regulatory T Cells Induce Alternatively Activated Monocytes With a Reduced Capacity to Expand T Helper-17 Cells. Front Immunol 2018; 9:1625. [PMID: 30079063 PMCID: PMC6062605 DOI: 10.3389/fimmu.2018.01625] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2018] [Accepted: 07/02/2018] [Indexed: 12/29/2022] Open
Abstract
Regulatory T cells (Tregs) are essential in maintaining peripheral immunological tolerance by modulating several subsets of the immune system including monocytes. Under inflammatory conditions, monocytes migrate into the tissues, where they differentiate into dendritic cells or tissue-resident macrophages. As a result of their context-dependent plasticity, monocytes have been implicated in the development/progression of graft-vs-host disease (GvHD), autoimmune diseases and allograft rejection. In the last decade, Tregs have been exploited for their use in cell therapy with the aim to induce tolerance after solid organ transplantation and for the treatment of autoimmune diseases and GvHD. To date, safety and feasibility of Treg infusion has been demonstrated; however, many questions of how these cells induce tolerance have been raised and need to be answered. As monocytes constitute the major cellular component in inflamed tissues, we have developed an in vitro model to test how Tregs modulate their phenotype and function. We demonstrated that expanded Tregs can drive monocytes toward an alternatively activated state more efficiently than freshly isolated Tregs. The effect of expanded Tregs on monocytes led to a reduced production of pro-inflammatory cytokines (IL-6 and tumor necrosis factor-α) and NF-κB activation. Furthermore, monocytes co-cultured with expanded Tregs downregulated the expression of co-stimulatory and MHC-class II molecules with a concomitant upregulation of M2 macrophage specific markers, CD206, heme oxygenase-1, and increased interleukin-10 production. Importantly, monocytes co-cultured with expanded Tregs showed a reduced capacity to expand IL-17-producing T cells compared with monocyte cultured with freshly isolated Tregs and conventional T cells. The capacity to decrease the expansion of pro-inflammatory Th-17 was not cytokine mediated but the consequence of their lower expression of the co-stimulatory molecule CD86. Our data suggest that expanded Tregs have the capacity to induce phenotypical and functional changes in monocytes that might be crucial for tolerance induction in transplantation and the prevention/treatment of GvHD and autoimmune diseases.
Collapse
Affiliation(s)
- Marco Romano
- Immunoregulation Laboratory, MRC Centre for Transplantation, School of Immunology & Microbial Sciences, King's College London, London, United Kingdom
| | - Giorgia Fanelli
- Immunoregulation Laboratory, MRC Centre for Transplantation, School of Immunology & Microbial Sciences, King's College London, London, United Kingdom
| | - Nicole Tan
- Immunoregulation Laboratory, MRC Centre for Transplantation, School of Immunology & Microbial Sciences, King's College London, London, United Kingdom
| | - Estefania Nova-Lamperti
- Immunoregulation Laboratory, MRC Centre for Transplantation, School of Immunology & Microbial Sciences, King's College London, London, United Kingdom.,Molecular and Translational Immunology Laboratory, Department of Clinical Biochemistry and Immunology, Faculty of Pharmacy, University of Concepcion, Concepcion, Chile
| | - Reuben McGregor
- Immunoregulation Laboratory, MRC Centre for Transplantation, School of Immunology & Microbial Sciences, King's College London, London, United Kingdom
| | - Robert I Lechler
- Immunoregulation Laboratory, MRC Centre for Transplantation, School of Immunology & Microbial Sciences, King's College London, London, United Kingdom
| | - Giovanna Lombardi
- Immunoregulation Laboratory, MRC Centre for Transplantation, School of Immunology & Microbial Sciences, King's College London, London, United Kingdom
| | - Cristiano Scottà
- Immunoregulation Laboratory, MRC Centre for Transplantation, School of Immunology & Microbial Sciences, King's College London, London, United Kingdom
| |
Collapse
|
26
|
Characterization of monocyte subtypes regarding their phenotype and development in the context of graft-versus-host disease. Transpl Immunol 2018; 50:48-54. [PMID: 29906586 DOI: 10.1016/j.trim.2018.06.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Revised: 06/08/2018] [Accepted: 06/11/2018] [Indexed: 12/11/2022]
Abstract
Graft-versus-host disease (GvHD) is a major cause of morbidity and mortality after allogeneic hematopoietic cell transplantation (HCT). In this study, monocyte subtypes were characterized regarding cytokine expression pattern and development in the context of GvHD. Using inflammatory S100 proteins for monocyte stimulation, it could be demonstrated that intermediate monocytes are the main producers of inflammatory cytokines such as IL-6 and TNFα known to be involved in the development of Th17 cells pointing towards an inflammatory phenotype of this monocyte subtype. Furthermore, novel aspects regarding monocyte subtype development were found. Our data reveal that prednisolone promotes the induction of intermediate monocytes from classical monocytes which correlates with HSP70 expression levels. However, 1α,25-Dihydroxyvitamin D3 treatment results in the abrogation of the prednisolone-mediated induction of this inflammatory monocyte subset and low HSP70 expression levels. Treatment of classical monocytes with pifithrin-μ, a specific HSP70 inhibitor, also leads to an inhibited induction of intermediate monocytes in the presence of prednisolone. These data point towards a predominant role of HSP70 in the development of intermediate monocytes. Thus, HSP70 might be a promising target for GvHD therapy, especially in combination with glucocorticoids, in order to decrease intermediate monocyte subset levels.
Collapse
|
27
|
Affiliation(s)
- Robert Zeiser
- From the Department of Hematology, Oncology, and Stem Cell Transplantation, Faculty of Medicine, Freiburg University Medical Center, Freiburg, Germany (R.Z.); and the Department of Pediatrics, Division of Blood and Marrow Transplantation, University of Minnesota, Minneapolis (B.R.B.)
| | - Bruce R Blazar
- From the Department of Hematology, Oncology, and Stem Cell Transplantation, Faculty of Medicine, Freiburg University Medical Center, Freiburg, Germany (R.Z.); and the Department of Pediatrics, Division of Blood and Marrow Transplantation, University of Minnesota, Minneapolis (B.R.B.)
| |
Collapse
|
28
|
Kobayashi A, Kobayashi S, Miyai K, Osawa Y, Horiuchi T, Kato S, Maekawa T, Yamamura T, Watanabe J, Sato K, Tsuda H, Kimura F. TAK1 inhibition ameliorates survival from graft-versus-host disease in an allogeneic murine marrow transplantation model. Int J Hematol 2017; 107:222-229. [PMID: 29027124 DOI: 10.1007/s12185-017-2345-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2017] [Revised: 10/01/2017] [Accepted: 10/03/2017] [Indexed: 12/26/2022]
Abstract
Acute graft-versus-host disease (GVHD) is a major cause of morbidity and mortality in allogeneic hematopoietic cell transplantation (allo-HCT). Majority of the current immunosuppressive strategies targeting donor T cells to prevent or treat acute GVHD are only partially effective, and often require escalated immunosuppressive therapy. Recent studies have revealed that activation of antigen-presenting cells in the proinflammatory milieu is important for the priming and promotion of GVHD. This activation is mediated by innate immune signaling pathways, which therefore potentially represent new targets in addressing GVHD. Using gene expression analysis of peripheral monocytes from patients' post-allo-HCT, we detected an upregulation of TGF-β-activated kinase 1 (TAK1), a key regulator of the toll-like receptor signaling pathway. 5Z-7-oxozeaenol, a selective inhibitor of TAK1, reduced proinflammatory cytokine production by activated monocytes under lipopolysaccharide stimulation and T cell proliferation in allogeneic-mixed leukocyte reactions with monocyte-derived dendritic cells. In an experimental mouse model of GVHD, 5Z-7-oxozeaenol administration after allo-HCT ameliorated GVHD severity and mortality, with significant reduction in serum TNFα, IL-1β, and IL-12 levels. Our findings suggest that altering the activation status of innate immune cells by TAK1 inhibition may be a novel therapeutic approach for acute GVHD.
Collapse
Affiliation(s)
- Ayako Kobayashi
- Division of Hematology, Department of Internal Medicine, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama, 359-8513, Japan.
| | - Shinichi Kobayashi
- Division of Hematology, Department of Internal Medicine, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama, 359-8513, Japan
| | - Kosuke Miyai
- Department of Basic Pathology, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama, 359-8513, Japan
| | - Yukiko Osawa
- Division of Hematology, Department of Internal Medicine, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama, 359-8513, Japan
| | - Toshikatsu Horiuchi
- Division of Hematology, Department of Internal Medicine, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama, 359-8513, Japan
| | - Shoichiro Kato
- Division of Hematology, Department of Internal Medicine, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama, 359-8513, Japan
| | - Takaaki Maekawa
- Division of Hematology, Department of Internal Medicine, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama, 359-8513, Japan
| | - Takeshi Yamamura
- Division of Hematology, Department of Internal Medicine, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama, 359-8513, Japan
| | - Junichi Watanabe
- Division of Hematology, Department of Internal Medicine, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama, 359-8513, Japan
| | - Ken Sato
- Division of Hematology, Department of Internal Medicine, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama, 359-8513, Japan
| | - Hitoshi Tsuda
- Department of Basic Pathology, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama, 359-8513, Japan
| | - Fumihiko Kimura
- Division of Hematology, Department of Internal Medicine, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama, 359-8513, Japan
| |
Collapse
|
29
|
Reinhardt-Heller K, Hirschberg I, Lang P, Vogl T, Handgretinger R, Bethge WA, Holzer U. Increase of Intermediate Monocytes in Graft-versus-Host Disease: Correlation with MDR1 +Th17.1 Levels and the Effect of Prednisolone and 1α,25-Dihydroxyvitamin D3. Biol Blood Marrow Transplant 2017; 23:2057-2064. [PMID: 28807771 DOI: 10.1016/j.bbmt.2017.08.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Accepted: 08/03/2017] [Indexed: 11/30/2022]
Abstract
Graft-versus-host disease (GVHD) remains one of the major complications after allogeneic hematopoietic stem cell transplantation that is mainly treated with glucocorticoids such as prednisolone. In this study the influence of monocyte subpopulations, prednisolone, and 1α,25-dihydroxyvitamin D3 (1α,25-(OH)2D3) on the induction of a proinflammatory subset of Th17 cells (MDR+Th17.1) characterized by CCR6+CXCR3hiCCR4loCCR10-CD161+ and stable expression of the multidrug resistance protein type 1 (MDR1) was investigated. Our results demonstrate that intermediate monocytes are increased in patients with acute GVHD, promoting the induction of proinflammatory MDR1+Th17.1 cells. Furthermore, prednisolone induces the development of MDR1+Th17.1 cells, whereas 1α,25-(OH)2D3 acts as an anti-inflammatory, leading to diminished percentages of proinflammatory MDR1+Th17.1 cells in the presence of prednisolone after stimulation with the TLR4-ligand S100A8/S100A9. Moreover, 1α,25-(OH)2D3 decreased the expression level of the targets JAK2 and CD74, both associated with T cell activation, in monocytes. Thus, in steroid-resistant GVHD, 1α,25-(OH)2D3 could be an important regulator in monocyte-induced development of proinflammatory MDR1+Th17.1 cells and might therefore be a potential therapeutic agent in combination with glucocorticoids for GVHD treatment.
Collapse
Affiliation(s)
| | | | - Peter Lang
- Children's Hospital, University of Tuebingen, Germany
| | - Thomas Vogl
- Institute of Immunology, University of Muenster, Germany
| | | | | | - Ursula Holzer
- Children's Hospital, University of Tuebingen, Germany.
| |
Collapse
|
30
|
Role of the intestinal mucosa in acute gastrointestinal GVHD. Blood 2017; 128:2395-2402. [PMID: 27856471 DOI: 10.1182/blood-2016-06-716738] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Accepted: 09/14/2016] [Indexed: 12/11/2022] Open
Abstract
Intestinal graft-versus-host disease (GVHD) remains a significant obstacle to the success of allogeneic hematopoietic cell transplantation. The intestinal mucosa comprises the inner lining of the intestinal tract and maintains close proximity with commensal microbes that reside within the intestinal lumen. Recent advances have significantly improved our understanding of the interactions between the intestinal mucosa and the enteric microbiota. Changes in host mucosal tissue and commensals posttransplant have been actively investigated, and provocative insights into mucosal immunity and the enteric microbiota are now being translated into clinical trials of novel approaches for preventing and treating acute GVHD. In this review, we summarize recent findings related to aspects of the intestinal mucosa during acute GVHD.
Collapse
|
31
|
Kessel C, Lippitz K, Weinhage T, Hinze C, Wittkowski H, Holzinger D, Fall N, Grom AA, Gruen N, Foell D. Proinflammatory Cytokine Environments Can Drive Interleukin-17 Overexpression by γ/δ T Cells in Systemic Juvenile Idiopathic Arthritis. Arthritis Rheumatol 2017; 69:1480-1494. [PMID: 28296284 DOI: 10.1002/art.40099] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Accepted: 03/09/2017] [Indexed: 01/03/2023]
Abstract
OBJECTIVE Systemic-onset juvenile idiopathic arthritis (JIA) is speculated to follow a biphasic course, with an initial systemic disease phase driven by innate immune mechanisms and interleukin-1β (IL-1β) as a key cytokine and a second chronic arthritic phase that may be dominated by adaptive immunity and cytokines such as IL-17A. Although a recent mouse model points to a critical role of IL-17-expressing γ/δ T cells in disease pathology, in humans, both the prevalence of IL-17 and the role of IL-17-producing cells are still unclear. METHODS Serum samples from systemic JIA patients and healthy pediatric controls were analyzed for the levels of IL-17A and related cytokines. Whole blood samples were studied for cellular expression of IL-17 and interferon-γ (IFNγ). CD4+ and γ/δ T cells isolated from the patients and controls were assayed for cytokine secretion in different culture systems. RESULTS IL-17A was prevalent in sera from patients with active systemic JIA, while both ex vivo and in vitro experiments revealed that γ/δ T cells overexpressed this cytokine. This was not seen with CD4+ T cells, which expressed strikingly low levels of IFNγ. Therapeutic IL-1 blockade was associated with partial normalization of both cytokine expression phenotypes. Furthermore, culturing healthy donor γ/δ T cells in serum from systemic JIA patients or in medium spiked with IL-1β, IL-18, and S100A12 induced IL-17 overexpression at levels similar to those observed in the patients' cells. CONCLUSION A systemic JIA cytokine environment may prime γ/δ T cells in particular for IL-17A overexpression. Thus, our observations in systemic JIA patients strongly support a pathophysiologic role of these cells, as proposed by the recent murine model.
Collapse
Affiliation(s)
| | | | | | - Claas Hinze
- University Children's Hospital, Münster, Germany
| | | | | | - Ndate Fall
- Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Alexei A Grom
- Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Niklas Gruen
- University Children's Hospital, Münster, Germany
| | - Dirk Foell
- University Children's Hospital, Münster, Germany
| |
Collapse
|
32
|
Hueso T, Coiteux V, Joncquel Chevalier Curt M, Labreuche J, Jouault T, Yakoub-Agha I, Seguy D. Citrulline and Monocyte-Derived Macrophage Reactivity before Conditioning Predict Acute Graft-versus-Host Disease. Biol Blood Marrow Transplant 2017; 23:913-921. [PMID: 28263922 DOI: 10.1016/j.bbmt.2017.03.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Accepted: 03/01/2017] [Indexed: 12/20/2022]
Abstract
During conditioning, intestinal damage induces microbial translocation which primes macrophage reactivity and leads to donor-derived T cell stimulation. Little is known about the role of intestinal health and macrophage reactivity before conditioning in the development of acute graft-versus-host disease (aGVHD) in patients undergoing allogeneic hematopoietic cell transplantation (allo-HCT). We assessed (1) citrulline, a surrogate marker of functional enterocyte mass and (2) circulating monocyte-derived macrophage reactivity, before allo-HCT. Forty-seven consecutive patients were prospectively included. Citrulline levels from blood samples withdrawn 30 days before transplantation were assessed using liquid chromatography combined with mass spectrometry. Monocyte-derived macrophages were isolated and incubated with 5 pathogen-associated molecular patterns: lipopolysaccharide, PamCSK4, flagellin, muramyl dipeptide, and curdlan. Multiplex fluorescent immunoassay on culture supernatant assessed levels of TNF-α, IL-1β, IL-6, and IL-10 in each condition. Citrulline and cytokine levels were analyzed relatively to aGVHD onset within 100 days after transplantation. Citrulline levels were lower in the aGVHD group (n = 20) than in the no-aGVHD group (n = 27) (P = .005). Conversely, IL-6 and IL-10 were greater in aGVHD group, especially after curdlan stimulation (P = .005 and P = .012). Citrulline levels ≤20 µmol/L, IL-6 ≥ 332 pg/mL, and IL-10 ≥ 90 pg/mL were associated with aGVHD development (log-rank test, P = .002, P = .041, and P < .0001, respectively). In multivariate analysis, IL-10 ≥ 90 pg/mL, myeloablative conditioning, and citrulline ≤20 µmol/L remained independent factors of aGVHD development (hazard ratio [HR], 8.18, P = .0003; HR, 4.28, P = .006; and HR, 4.43, P = .01, respectively). Preconditioning citrulline and monocyte-derived macrophage reactivity are objective surrogate markers suitable to identify patients at risk of developing aGVHD. This work highlights the influence of preconditioning status in aGVHD development.
Collapse
Affiliation(s)
- Thomas Hueso
- LIRIC UMR 995 Inserm, University of Lille, Lille, France
| | | | | | | | | | - Ibrahim Yakoub-Agha
- LIRIC UMR 995 Inserm, University of Lille, Lille, France; Stem Cell Transplantation Unit, CHU Lille, Lille, France
| | - David Seguy
- LIRIC UMR 995 Inserm, University of Lille, Lille, France; Department of Nutrition, CHU Lille, Lille, France.
| |
Collapse
|
33
|
Peled JU, Hanash AM, Jenq RR. Role of the intestinal mucosa in acute gastrointestinal GVHD. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2016; 2016:119-127. [PMID: 27913470 PMCID: PMC5575743 DOI: 10.1182/asheducation-2016.1.119] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/10/2023]
Abstract
Intestinal graft-versus-host disease (GVHD) remains a significant obstacle to the success of allogeneic hematopoietic cell transplantation. The intestinal mucosa comprises the inner lining of the intestinal tract and maintains close proximity with commensal microbes that reside within the intestinal lumen. Recent advances have significantly improved our understanding of the interactions between the intestinal mucosa and the enteric microbiota. Changes in host mucosal tissue and commensals posttransplant have been actively investigated, and provocative insights into mucosal immunity and the enteric microbiota are now being translated into clinical trials of novel approaches for preventing and treating acute GVHD. In this review, we summarize recent findings related to aspects of the intestinal mucosa during acute GVHD.
Collapse
Affiliation(s)
- Jonathan U Peled
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY; and
- Weill Cornell Medical College, New York, NY
| | - Alan M Hanash
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY; and
| | - Robert R Jenq
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY; and
- Weill Cornell Medical College, New York, NY
| |
Collapse
|
34
|
Apostolova P, Zeiser R. The Role of Purine Metabolites as DAMPs in Acute Graft-versus-Host Disease. Front Immunol 2016; 7:439. [PMID: 27818661 PMCID: PMC5073102 DOI: 10.3389/fimmu.2016.00439] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Accepted: 10/06/2016] [Indexed: 12/15/2022] Open
Abstract
Acute graft-versus-host disease (GvHD) causes high mortality in patients undergoing allogeneic hematopoietic cell transplantation. An early event in the classical pathogenesis of acute GvHD is tissue damage caused by the conditioning treatment or infection that consecutively leads to translocation of bacterial products [pathogen-associated molecular patterns (PAMPs)] into blood or lymphoid tissue, as well as danger-associated molecular patterns (DAMPs), mostly intracellular components that act as pro-inflammatory agents, once they are released into the extracellular space. A subtype of DAMPs is nucleotides, such as adenosine triphosphate released from dying cells that can activate the innate and adaptive immune system by binding to purinergic receptors. Binding to certain purinergic receptors leads to a pro-inflammatory microenvironment and promotes allogeneic T cell priming. After priming, T cells migrate to the acute GvHD target organs, mainly skin, liver, and the gastrointestinal tract and induce cell damage that further amplifies the release of intracellular components. This review summarizes the role of different purinergic receptors in particular P2X7 and P2Y2 as well as nucleotides in the pathogenesis of GvHD.
Collapse
Affiliation(s)
- Petya Apostolova
- Department of Hematology, Oncology and Stem Cell Transplantation, University Medical Center, Albert Ludwig University of Freiburg , Freiburg , Germany
| | - Robert Zeiser
- Department of Hematology, Oncology and Stem Cell Transplantation, University Medical Center, Albert Ludwig University of Freiburg , Freiburg , Germany
| |
Collapse
|
35
|
Systems analysis uncovers inflammatory Th/Tc17-driven modules during acute GVHD in monkey and human T cells. Blood 2016; 128:2568-2579. [PMID: 27758873 DOI: 10.1182/blood-2016-07-726547] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Accepted: 09/22/2016] [Indexed: 01/30/2023] Open
Abstract
One of the central challenges of transplantation is the development of alloreactivity despite the use of multiagent immunoprophylaxis. Effective control of this immune suppression-resistant T-cell activation represents one of the key unmet needs in the fields of both solid-organ and hematopoietic stem cell transplant (HCT). To address this unmet need, we have used a highly translational nonhuman primate (NHP) model to interrogate the transcriptional signature of T cells during breakthrough acute graft-versus-host disease (GVHD) that occurs in the setting of clinically relevant immune suppression and compared this to the hyperacute GVHD, which develops in unprophylaxed or suboptimally prophylaxed transplant recipients. Our results demonstrate the complex character of the alloreactivity that develops during ongoing immunoprophylaxis and identify 3 key transcriptional hallmarks of breakthrough acute GVHD that are not observed in hyperacute GVHD: (1) T-cell persistence rather than proliferation, (2) evidence for highly inflammatory transcriptional programming, and (3) skewing toward a T helper (Th)/T cytotoxic (Tc)17 transcriptional program. Importantly, the gene coexpression profiles from human HCT recipients who developed GVHD while on immunosuppressive prophylactic agents recapitulated the patterns observed in NHP, and demonstrated an evolution toward a more inflammatory signature as time posttransplant progressed. These results strongly implicate the evolution of both inflammatory and interleukin 17-based immune pathogenesis in GVHD, and provide the first map of this evolving process in primates in the setting of clinically relevant immunomodulation. This map represents a novel transcriptomic resource for further systems-based efforts to study the breakthrough alloresponse that occurs posttransplant despite immunoprophylaxis and to develop evidence-based strategies for effective treatment of this disease.
Collapse
|
36
|
Teshima T, Reddy P, Zeiser R. Reprint of: Acute Graft-versus-Host Disease: Novel Biological Insights. Biol Blood Marrow Transplant 2016; 22:S3-8. [PMID: 26899274 DOI: 10.1016/j.bbmt.2016.01.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Accepted: 10/01/2015] [Indexed: 12/13/2022]
Abstract
Graft-versus-host disease (GVHD) continues to be a leading cause of morbidity and mortality after allogeneic hematopoietic stem cell transplantation. Recent insights into intestinal homeostasis and uncovering of new pathways and targets have greatly reconciled our understanding of GVHD pathophysiology and will reshape contemporary GVHD prophylaxis and treatment. Gastrointestinal (GI) GVHD is the major cause of mortality. Emerging data indicate that intestinal stem cells (ISCs) and their niche Paneth cells are targeted, resulting in dysregulation of the intestinal homeostasis and microbial ecology. The microbiota and their metabolites shape the immune system and intestinal homeostasis, and they may alter host susceptibility to GVHD. Protection of the ISC niche system and modification of the intestinal microbiota and metabolome to restore intestinal homeostasis may, thus, represent a novel approach to modulate GVHD and infection. Damage to the intestine plays a central role in amplifying systemic GVHD by propagating a proinflammatory cytokine milieu. Molecular targeting to inhibit kinase signaling may be a promising approach to treat GVHD, ideally via targeting the redundant effect of multiple cytokines on immune cells and enterocytes. In this review, we discuss insights on the biology of GI GVHD, interaction of microflora and metabolome with the hosts, identification of potential new target organs, and identification and targeting of novel T cell-signaling pathways. Better understanding of GVHD biology will, thus, pave a way to develop novel treatment strategies with great clinical benefits.
Collapse
Affiliation(s)
- Takanori Teshima
- Department of Hematology, Hokkaido University Graduate School of Medicine, Sapporo, Japan.
| | - Pavan Reddy
- Department of Internal Medicine, University of Michigan Comprehensive Cancer Center, Ann Arbor, Michigan
| | - Robert Zeiser
- Department of Hematology and Oncology, Freiburg University Medical Center, Albert-Ludwigs-University, Freiburg, Germany
| |
Collapse
|
37
|
The Role of Biomarkers in the Diagnosis and Risk Stratification of Acute Graft-versus-Host Disease: A Systematic Review. Biol Blood Marrow Transplant 2016; 22:1552-1564. [PMID: 27158050 DOI: 10.1016/j.bbmt.2016.04.022] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Accepted: 04/26/2016] [Indexed: 12/20/2022]
Abstract
Allogeneic hematopoietic cell transplantation (HCT) is an increasingly used curative modality for hematologic malignancies and other benign conditions. Attempts to reduce morbidity and mortality and improve survival in patients undergoing HCT are crucial. The ability to diagnose acute graft-versus-host disease (aGVHD) in a timely manner, or to even predict aGVHD before clinical manifestations, along with the accurate stratification of these patients, are critical steps to improve the treatment and outcomes of these patients. Many novel biomarkers that may help achieve these goals have been studied recently. This overview is intended to assist clinicians and investigators by providing a comprehensive review and analytical interpretation of the current knowledge concerning aGVHD and biomarkers likely to prove useful in diagnosis and risk stratification of this condition, along with the difficulties that hamper this approach.
Collapse
|
38
|
Malard F, Gaugler B, Lamarthee B, Mohty M. Translational opportunities for targeting the Th17 axis in acute graft-vs.-host disease. Mucosal Immunol 2016; 9:299-308. [PMID: 26813345 DOI: 10.1038/mi.2015.143] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Accepted: 12/02/2015] [Indexed: 02/04/2023]
Abstract
Allogeneic stem cell transplantation (allo-SCT) is a curative therapy for different life-threatening malignant and non-malignant hematologic disorders. Acute graft-vs.-host disease (aGVHD) and particularly gastrointestinal aGVHD remains a major source of morbidity and mortality following allo-SCT, which limits the use of this treatment in a broader spectrum of patients. Better understanding of aGVHD pathophysiology is indispensable to identify new therapeutic targets for aGVHD prevention and therapy. Growing amount of data suggest a role for T helper (Th)17 cells in aGVHD pathophysiology. In this review, we will discuss the current knowledge in this area in animal models and in humans. We will then describe new potential treatments for aGVHD along the Th17 axis.
Collapse
Affiliation(s)
- F Malard
- Université Pierre et Marie Curie, Paris, France.,Centre de recherche Saint-Antoine, INSERM, UMRs 938, Paris, France.,Service d'Hématologie Clinique et de Thérapie Cellulaire, Hôpital Saint Antoine, APHP, Paris, France.,INSERM, UMR 1064-Center for Research in Transplantation and Immunology, Nantes, F44093 France
| | - B Gaugler
- Université Pierre et Marie Curie, Paris, France.,Centre de recherche Saint-Antoine, INSERM, UMRs 938, Paris, France
| | - B Lamarthee
- Université Pierre et Marie Curie, Paris, France.,Centre de recherche Saint-Antoine, INSERM, UMRs 938, Paris, France
| | - M Mohty
- Université Pierre et Marie Curie, Paris, France.,Centre de recherche Saint-Antoine, INSERM, UMRs 938, Paris, France.,Service d'Hématologie Clinique et de Thérapie Cellulaire, Hôpital Saint Antoine, APHP, Paris, France
| |
Collapse
|
39
|
Apostolova P, Zeiser R. The role of danger signals and ectonucleotidases in acute graft-versus-host disease. Hum Immunol 2016; 77:1037-1047. [PMID: 26902992 DOI: 10.1016/j.humimm.2016.02.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2015] [Revised: 02/09/2016] [Accepted: 02/18/2016] [Indexed: 12/28/2022]
Abstract
Allogeneic hematopoietic cell transplantation (allo-HCT) represents the only curative treatment approach for many patients with benign or malignant diseases of the hematopoietic system. However, post-transplant morbidity and mortality are significantly increased by the development of acute graft-versus-host disease (GvHD). While alloreactive T cells act as the main cellular mediator of the GvH reaction, recent evidence suggests a critical role of the innate immune system in the early stages of GvHD initiation. Danger-associated molecular patterns released from the intracellular space as well as from the extracellular matrix activate antigen-presenting cells and set pro-inflammatory pathways in motion. This review gives an overview about danger signals representing therapeutic targets with a clinical perspective with a particular focus on extracellular nucleotides and ectonucleotidases.
Collapse
Affiliation(s)
- Petya Apostolova
- Department of Hematology, Oncology and Stem Cell Transplantation, Freiburg University Medical Center, Albert-Ludwigs-University, Freiburg, Germany.
| | - Robert Zeiser
- Department of Hematology, Oncology and Stem Cell Transplantation, Freiburg University Medical Center, Albert-Ludwigs-University, Freiburg, Germany.
| |
Collapse
|
40
|
Klämbt V, Wohlfeil SA, Schwab L, Hülsdünker J, Ayata K, Apostolova P, Schmitt-Graeff A, Dierbach H, Prinz G, Follo M, Prinz M, Idzko M, Zeiser R. A Novel Function for P2Y2 in Myeloid Recipient-Derived Cells during Graft-versus-Host Disease. THE JOURNAL OF IMMUNOLOGY 2015; 195:5795-804. [PMID: 26538394 DOI: 10.4049/jimmunol.1501357] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Accepted: 10/11/2015] [Indexed: 11/19/2022]
Abstract
Acute graft-versus-host disease (GvHD) is a life-threatening complication of allogeneic hematopoietic cell transplantation. During the initiation phase of acute GvHD, endogenous danger signals such as ATP are released and inform the innate immune system via activation of the purinergic receptor P2X7 that a noninfectious damage has occurred. A second ATP-activated purinergic receptor involved in inflammatory diseases is P2Y2. In this study, we used P2y2(-/-) mice to test the role of this receptor in GvHD. P2y2(-/-) recipients experienced reduced GvHD-related mortality, IL-6 levels, enterocyte apoptosis, and histopathology scores. Chimeric mice with P2y2 deficiency restricted to hematopoietic tissues survived longer after GvHD induction than did wild-type mice. P2y2 deficiency of the recipient was connected to lower levels of myeloperoxidase in the intestinal tract of mice developing GvHD and a reduced myeloid cell signature. Selective deficiency of P2Y2 in inflammatory monocytes decreased GvHD severity. Mechanistically, P2y2(-/-) inflammatory monocytes displayed defective ERK activation and reactive oxygen species production. Compatible with a role of P2Y2 in human GvHD, the frequency of P2Y2(+) cells in inflamed GvHD lesions correlated with histopathological GvHD severity. Our findings indicate a novel function for P2Y2 in ATP-activated recipient myeloid cells during GvHD, which could be exploited when targeting danger signals to prevent GvHD.
Collapse
Affiliation(s)
- Verena Klämbt
- Department of Hematology and Oncology, University Medical Center Freiburg, 79106 Freiburg, Germany
| | - Sebastian A Wohlfeil
- Department of Hematology and Oncology, University Medical Center Freiburg, 79106 Freiburg, Germany
| | - Lukas Schwab
- Department of Hematology and Oncology, University Medical Center Freiburg, 79106 Freiburg, Germany
| | - Jan Hülsdünker
- Department of Hematology and Oncology, University Medical Center Freiburg, 79106 Freiburg, Germany; Spemann Graduate School of Biology and Medicine, University of Freiburg, 79104 Freiburg, Germany; Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany
| | - Korcan Ayata
- Department of Pneumology, University Medical Center Freiburg, 79106 Freiburg, Germany
| | - Petya Apostolova
- Department of Hematology and Oncology, University Medical Center Freiburg, 79106 Freiburg, Germany
| | | | - Heide Dierbach
- Department of Hematology and Oncology, University Medical Center Freiburg, 79106 Freiburg, Germany
| | - Gabriele Prinz
- Department of Hematology and Oncology, University Medical Center Freiburg, 79106 Freiburg, Germany
| | - Marie Follo
- Department of Hematology and Oncology, University Medical Center Freiburg, 79106 Freiburg, Germany
| | - Marco Prinz
- Institute of Neuropathology, University Medical Center Freiburg, 79106 Freiburg, Germany; and BIOSS Centre for Biological Signalling Studies, Albert Ludwigs University of Freiburg, 79104 Freiburg, Germany
| | - Marco Idzko
- Department of Pneumology, University Medical Center Freiburg, 79106 Freiburg, Germany
| | - Robert Zeiser
- Department of Hematology and Oncology, University Medical Center Freiburg, 79106 Freiburg, Germany; BIOSS Centre for Biological Signalling Studies, Albert Ludwigs University of Freiburg, 79104 Freiburg, Germany
| |
Collapse
|
41
|
Teshima T, Reddy P, Zeiser R. Acute Graft-versus-Host Disease: Novel Biological Insights. Biol Blood Marrow Transplant 2015; 22:11-6. [PMID: 26453971 DOI: 10.1016/j.bbmt.2015.10.001] [Citation(s) in RCA: 75] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Accepted: 10/01/2015] [Indexed: 12/22/2022]
Abstract
Graft-versus-host disease (GVHD) continues to be a leading cause of morbidity and mortality after allogeneic hematopoietic stem cell transplantation. Recent insights into intestinal homeostasis and uncovering of new pathways and targets have greatly reconciled our understanding of GVHD pathophysiology and will reshape contemporary GVHD prophylaxis and treatment. Gastrointestinal (GI) GVHD is the major cause of mortality. Emerging data indicate that intestinal stem cells (ISCs) and their niche Paneth cells are targeted, resulting in dysregulation of the intestinal homeostasis and microbial ecology. The microbiota and their metabolites shape the immune system and intestinal homeostasis, and they may alter host susceptibility to GVHD. Protection of the ISC niche system and modification of the intestinal microbiota and metabolome to restore intestinal homeostasis may, thus, represent a novel approach to modulate GVHD and infection. Damage to the intestine plays a central role in amplifying systemic GVHD by propagating a proinflammatory cytokine milieu. Molecular targeting to inhibit kinase signaling may be a promising approach to treat GVHD, ideally via targeting the redundant effect of multiple cytokines on immune cells and enterocytes. In this review, we discuss insights on the biology of GI GVHD, interaction of microflora and metabolome with the hosts, identification of potential new target organs, and identification and targeting of novel T cell-signaling pathways. Better understanding of GVHD biology will, thus, pave a way to develop novel treatment strategies with great clinical benefits.
Collapse
Affiliation(s)
- Takanori Teshima
- Department of Hematology, Hokkaido University Graduate School of Medicine, Sapporo, Japan.
| | - Pavan Reddy
- Department of Internal Medicine, University of Michigan Comprehensive Cancer Center, Ann Arbor, Michigan
| | - Robert Zeiser
- Department of Hematology and Oncology, Freiburg University Medical Center, Albert-Ludwigs-University, Freiburg, Germany
| |
Collapse
|
42
|
Zhu L, Couriel DR, Chang CH. The effect of extracorporeal photopheresis on T cell response in chronic graft-versus-host disease. Leuk Lymphoma 2015; 57:376-384. [PMID: 26059058 DOI: 10.3109/10428194.2015.1057893] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Extracorporeal photopheresis (ECP) is a safe and effective immunoregulatory therapy for steroid-refractory chronic graft-versus-host disease (cGVHD) but its mechanism of action is poorly understood. In this study, we evaluated the effect of ECP in a sample of cGVHD patients. Our data showed that ECP-treated patients had lower CD4 T and B cells, and substantially higher NK cells than untreated patients. T regulatory (Treg) cells were similar between the two groups of patients. Interestingly, Treg cells were higher in ECP-treated patients and ECP-responders who had no history of aGVHD or sclerosis, than in those who had one of them or both. These findings suggest that at least one of the mechanisms of immunomodulation by ECP targets the Treg cell population and that an increase in Treg cells may be associated with response in patients with cGVHD. Together, the results of ECP are different depending on the patients' clinical condition.
Collapse
Affiliation(s)
- Lingqiao Zhu
- a Department of Microbiology and Immunology , University of Michigan Medical School , Ann Arbor , MI , USA
| | - Daniel R Couriel
- b Adult Blood and Marrow Transplant Program, University of Michigan Comprehensive Cancer Center , Ann Arbor , MI , USA
| | - Cheong-Hee Chang
- a Department of Microbiology and Immunology , University of Michigan Medical School , Ann Arbor , MI , USA
| |
Collapse
|
43
|
Ramadan A, Paczesny S. Various forms of tissue damage and danger signals following hematopoietic stem-cell transplantation. Front Immunol 2015; 6:14. [PMID: 25674088 PMCID: PMC4309199 DOI: 10.3389/fimmu.2015.00014] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2014] [Accepted: 01/08/2015] [Indexed: 12/13/2022] Open
Abstract
Hematopoietic stem-cell transplantation (HSCT) is the most potent curative therapy for many malignant and non-malignant disorders. Unfortunately, a major complication of HSCT is graft-versus-host disease (GVHD), which is mediated by tissue damage resulting from the conditioning regimens before the transplantation and the alloreaction of dual immune components (activated donor T-cells and recipient’s antigen-presenting cells). This tissue damage leads to the release of alarmins and the triggering of pathogen-recognition receptors that activate the innate immune system and subsequently the adaptive immune system. Alarmins, which are of endogenous origin, together with the exogenous pathogen-associated molecular patterns (PAMPs) elicit similar responses of danger signals and represent the group of damage-associated molecular patterns (DAMPs). Effector cells of innate and adaptive immunity that are activated by PAMPs or alarmins can secrete other alarmins and amplify the immune responses. These complex interactions and loops between alarmins and PAMPs are particularly potent at inducing and then aggravating the GVHD reaction. In this review, we highlight the role of these tissue damaging molecules and their signaling pathways. Interestingly, some DAMPs and PAMPs are organ specific and GVHD-induced and have been shown to be interesting biomarkers. Some of these molecules may represent potential targets for novel therapeutic approaches.
Collapse
Affiliation(s)
- Abdulraouf Ramadan
- Department of Pediatrics, Melvin and Bren Simon Cancer Center, Indiana University , Indianapolis, IN , USA ; Department of Microbiology and Immunology, Indiana University , Indianapolis, IN , USA
| | - Sophie Paczesny
- Department of Pediatrics, Melvin and Bren Simon Cancer Center, Indiana University , Indianapolis, IN , USA ; Department of Microbiology and Immunology, Indiana University , Indianapolis, IN , USA
| |
Collapse
|
44
|
Patterns of monocyte subpopulations and their surface expression of HLA-DR during adverse events after hematopoietic stem cell transplantation. Ann Hematol 2014; 94:825-36. [DOI: 10.1007/s00277-014-2287-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2014] [Accepted: 12/18/2014] [Indexed: 12/24/2022]
|
45
|
Hülsdünker J, Zeiser R. Insights into the pathogenesis of GvHD: what mice can teach us about man. ACTA ACUST UNITED AC 2014; 85:2-9. [PMID: 25532439 DOI: 10.1111/tan.12497] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Acute graft-vs-host disease (GvHD) is a life-threatening complication of allogeneic hematopoietic cell transplantation (allo-HCT). Most of the knowledge about the biology of GvHD is derived from mouse models of this disease and therefore a critical analysis of potential advantages and disadvantages of the murine GvHD models is important to classify and understand the findings made in these models. The central events leading up to GvHD were characterized in three phases which includes the tissue damage-phase, the T cell priming-phase and the effector-phase, when the disease becomes clinically overt. The role of individual cytokines, chemokines, transcription factor or receptors was studied in these models by using gene deficient or transgenic mice in the donor or recipient compartments. Besides, numerous studies have been performed in these models to prevent or treat GvHD. Several recent clinical trials were all based on previously reported findings from the mouse model of GvHD such as the trials on CCR5-blockade, donor statin treatment, vorinostat treatment or adoptive transfer of regulatory T cells for GvHD prevention. The different mouse models for GvHD and graft-vs-leukemia effects are critically reviewed and their impact on current clinical practice is discussed.
Collapse
Affiliation(s)
- J Hülsdünker
- Department of Hematology, Oncology and Stem Cell Transplantation, Freiburg University Medical Center, Albert Ludwigs University Freiburg, Freiburg, Germany; Spemann Graduate School of Biology and Medicine (SGBM), University of Freiburg, Freiburg, Germany; Faculty of Biology, University of Freiburg, Freiburg, Germany
| | | |
Collapse
|