1
|
Yu X, Chen Y, Lei L, Li P, Lin D, Shen Y, Hou C, Chen J, Fan Y, Jin Y, Lu H, Wu D, Xu Y. Mendelian randomization analysis of blood metabolites and immune cell mediators in relation to GVHD and relapse. BMC Med 2025; 23:201. [PMID: 40189523 PMCID: PMC11974087 DOI: 10.1186/s12916-025-04026-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Accepted: 03/19/2025] [Indexed: 04/09/2025] Open
Abstract
BACKGROUND Graft-versus-host disease (GVHD) and relapse are major complications following allogeneic hematopoietic stem cell transplantation (allo-HSCT). Metabolites play crucial roles in immune regulation, but their causal relationships with GVHD and relapse remain unclear. METHODS We utilized genetic variants from genome-wide association studies (GWAS) of 309 known metabolites as instrumental variables to evaluate their causal effects on acute GVHD (aGVHD), gut GVHD, chronic GVHD (cGVHD), and relapse in different populations. Multiple causal inference methods, heterogeneity assessments, and pleiotropy tests were conducted to ensure result robustness. Multivariable MR analysis was performed to adjust for potential confounders, and validation MR analysis further confirmed key findings. Mediation MR analysis was employed to explore indirect causal pathways. RESULTS After correction for multiple testing, we identified elevated pyridoxate and proline levels as protective factors against grade 3-4 aGVHD (aGVHD3) and relapse, respectively. Conversely, glycochenodeoxycholate increased the risk of aGVHD3, whereas 1-stearoylglycerophosphoethanolamine had a protective effect. The robustness and stability of these findings were confirmed by multiple causal inference approaches, heterogeneity, and horizontal pleiotropy analyses. Multivariable MR analysis further excluded potential confounding pleiotropic effects. Validation MR analyses supported the causal roles of pyridoxate and 1-stearoylglycerophosphoethanolamine, while mediation MR revealed that pyridoxate influences GVHD directly and indirectly via CD39 + Tregs. Pathway analyses highlighted critical biochemical alterations, including disruptions in bile acid metabolism and the regulatory roles of vitamin B6 derivatives. Finally, clinical metabolic analyses, including direct fecal metabolite measurements, confirmed the protective role of pyridoxate against aGVHD. CONCLUSIONS Our findings provide novel insights into the metabolic mechanisms underlying GVHD and relapse after allo-HSCT. Identified metabolites, particularly pyridoxate, may serve as potential therapeutic targets for GVHD prevention and management.
Collapse
Affiliation(s)
- Xinghao Yu
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Yiyin Chen
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Lei Lei
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Pengfei Li
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Dandan Lin
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Ying Shen
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Chang Hou
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Jia Chen
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Yi Fan
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Yi Jin
- Department of Pharmacy, Wujin Hospital Affiliated with Jiangsu University, Changzhou, 213000, China
| | - Huimin Lu
- Department of Outpatient and Emergency, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Depei Wu
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China.
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China.
| | - Yang Xu
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China.
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China.
| |
Collapse
|
2
|
Radhouani M, Farhat A, Hakobyan A, Zahalka S, Pimenov L, Fokina A, Hladik A, Lakovits K, Brösamlen J, Dvorak V, Nunes N, Zech A, Idzko M, Krausgruber T, Köhl J, Uluckan O, Kovarik J, Hoehlig K, Vater A, Eckhard M, Sombke A, Fortelny N, Menche J, Knapp S, Starkl P. Eosinophil innate immune memory after bacterial skin infection promotes allergic lung inflammation. Sci Immunol 2025; 10:eadp6231. [PMID: 40184438 DOI: 10.1126/sciimmunol.adp6231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 11/22/2024] [Accepted: 02/27/2025] [Indexed: 04/06/2025]
Abstract
Microbial exposure at barrier interfaces drives development and balance of the immune system, but the consequences of local infections for systemic immunity and secondary inflammation are unclear. Here, we show that skin exposure to the bacterium Staphylococcus aureus persistently shapes the immune system of mice with specific impact on progenitor and mature bone marrow neutrophil and eosinophil populations. The infection-imposed changes in eosinophils were long-lasting and associated with functional as well as imprinted epigenetic and metabolic changes. Bacterial exposure enhanced cutaneous allergic sensitization and resulted in exacerbated allergen-induced lung inflammation. Functional bone marrow eosinophil reprogramming and pulmonary allergen responses were driven by the alarmin interleukin-33 and the complement cleavage fragment C5a. Our study highlights the systemic impact of skin inflammation and reveals mechanisms of eosinophil innate immune memory and organ cross-talk that modulate systemic responses to allergens.
Collapse
Affiliation(s)
- Mariem Radhouani
- Department of Medicine I, Research Division Infection Biology, Medical University of Vienna, Vienna, Austria
- CeMM-Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Asma Farhat
- Department of Medicine I, Research Division Infection Biology, Medical University of Vienna, Vienna, Austria
- CeMM-Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Anna Hakobyan
- CeMM-Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
- Max Perutz Labs, Vienna Biocenter Campus (VBC), Vienna, Austria
- Department of Structural and Computational Biology, Center for Molecular Biology, University of Vienna, Vienna, Austria
| | - Sophie Zahalka
- Department of Medicine I, Research Division Infection Biology, Medical University of Vienna, Vienna, Austria
- CeMM-Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Lisabeth Pimenov
- Department of Medicine I, Research Division Infection Biology, Medical University of Vienna, Vienna, Austria
| | - Alina Fokina
- Department of Medicine I, Research Division Infection Biology, Medical University of Vienna, Vienna, Austria
| | - Anastasiya Hladik
- Department of Medicine I, Research Division Infection Biology, Medical University of Vienna, Vienna, Austria
| | - Karin Lakovits
- Department of Medicine I, Research Division Infection Biology, Medical University of Vienna, Vienna, Austria
| | - Jessica Brösamlen
- Department of Medicine I, Research Division Infection Biology, Medical University of Vienna, Vienna, Austria
| | | | - Natalia Nunes
- Center for Tumor Biology and Immunology, Department of Biosciences and Medical Biology, Paris-Lodron University Salzburg, Salzburg, Austria
| | - Andreas Zech
- Department of Medicine II, Department of Pulmonology, Medical University of Vienna, Vienna, Austria
| | - Marco Idzko
- Department of Medicine II, Department of Pulmonology, Medical University of Vienna, Vienna, Austria
| | - Thomas Krausgruber
- CeMM-Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
- Institute of Artificial Intelligence, Center for Medical Data Science, Medical University of Vienna, Vienna, Austria
| | - Jörg Köhl
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Ozge Uluckan
- Novartis Biomedical Research, Basel, Switzerland
| | - Jiri Kovarik
- Novartis Biomedical Research, Basel, Switzerland
| | | | | | - Margret Eckhard
- Center for Anatomy and Cell Biology, Cell and Developmental Biology, Medical University of Vienna, Vienna, Austria
| | - Andy Sombke
- Center for Anatomy and Cell Biology, Cell and Developmental Biology, Medical University of Vienna, Vienna, Austria
| | - Nikolaus Fortelny
- Center for Tumor Biology and Immunology, Department of Biosciences and Medical Biology, Paris-Lodron University Salzburg, Salzburg, Austria
| | - Jörg Menche
- CeMM-Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
- Max Perutz Labs, Vienna Biocenter Campus (VBC), Vienna, Austria
- Department of Structural and Computational Biology, Center for Molecular Biology, University of Vienna, Vienna, Austria
- Faculty of Mathematics, University of Vienna, Vienna, Austria
- Ludwig Boltzmann Institute for Network Medicine at the University of Vienna, Vienna, Austria
| | - Sylvia Knapp
- Department of Medicine I, Research Division Infection Biology, Medical University of Vienna, Vienna, Austria
- Ignaz Semmelweis Institute, Interuniversity Institute for Infection Research, Vienna, Austria
| | - Philipp Starkl
- Department of Medicine I, Research Division Infection Biology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
3
|
Klesse M, Schanz O, Heine A. Establishing a low-dose x-ray irradiation protocol for experimental acute graft-versus-host disease. Exp Hematol 2025; 146:104765. [PMID: 40164325 DOI: 10.1016/j.exphem.2025.104765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Revised: 02/27/2025] [Accepted: 03/13/2025] [Indexed: 04/02/2025]
Abstract
The investigation of graft-versus-host disease (GvHD) after allogeneic stem cell transplantation heavily relies on the use of experimental animal models and total body irradiation (TBI) as a conditioning regimen. However, 137Cs is gradually being replaced as the main source of radiation due to safety concerns, and the transfer of established irradiation protocols to x-ray irradiators has proven difficult. Here, we describe the establishment of an x-ray-based irradiation protocol in an experimental mouse model for acute GvHD (C57BL6 → BALB/c). Our data show that commonly reported dosages of 6-9 Gy did not result in a viable model. Instead, irradiation with 5 Gy led to the development of clinical symptoms of GvHD in mice after transplantation with allogeneic bone marrow and T cells. Mice with GvHD displayed altered hemograms and increased serum levels of proinflammatory cytokines compared with mice without GvHD, which was accompanied by sequestration of donor lymphocytes within organs. Donor chimerism and hemogram analyses also indicated sufficient myeloablation and hematopoietic reconstitution. Overall, we show that low-dose x-ray TBI effectively promotes acute GvHD in a mismatched mouse model. We also propose that the transfer of previously established gamma-ray TBI protocols should be carefully evaluated according to individual circumstances.
Collapse
Affiliation(s)
- Michelle Klesse
- Medical Clinic III for Oncology, Hematology, Rheumatology and Stem Cell Transplantation, University Hospital Bonn, Bonn, Germany
| | - Oliver Schanz
- Medical Clinic III for Oncology, Hematology, Rheumatology and Stem Cell Transplantation, University Hospital Bonn, Bonn, Germany
| | - Annkristin Heine
- Medical Clinic III for Oncology, Hematology, Rheumatology and Stem Cell Transplantation, University Hospital Bonn, Bonn, Germany; Clinical Division of Hematology, Department of Internal Medicine, Medical University of Graz, Auenbruggerplatz 38, 8036 Graz, Austria.
| |
Collapse
|
4
|
Kitamura W, Fujii K, Tsuge M, Mitsuhashi T, Kobayashi H, Kamoi C, Yamamoto A, Kondo T, Seike K, Fujiwara H, Asada N, Ennishi D, Matsuoka KI, Fujii N, Maeda Y. A randomized controlled trial of conventional GVHD prophylaxis with or without teprenone for the prevention of severe acute GVHD. Ann Hematol 2025; 104:1917-1929. [PMID: 39994018 PMCID: PMC12031882 DOI: 10.1007/s00277-025-06269-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Accepted: 02/16/2025] [Indexed: 02/26/2025]
Abstract
Therapies that effectively suppress graft-versus-host disease (GVHD) without compromising graft-versus-leukemia/lymphoma (GVL) effects is important in allogeneic hematopoietic stem cell transplantation (allo-HSCT) for hematopoietic malignancies. Geranylgeranylacetone (GGA) is a main component of teprenone, a gastric mucosal protectant commonly used in clinical practice. In preclinical models, GGA suppresses proinflammatory cytokines, including interleukin (IL)-1β, IL-6, and tumor necrosis factor-α (TNF-α), which are associated with GVHD as well as induces thioredoxin-1 (Trx-1), which suppresses GVHD while maintaining GVL effects. Here, we investigated whether the addition of teprenone to standard GVHD prophylaxis could reduce the cumulative incidence of severe acute GVHD (aGVHD) without attenuating GVL effects. This open-label, randomized clinical trial enrolled 40 patients (21 control and 19 teprenone group) who received allo-HSCT between May 2022 and February 2023 in our institution. Patients in the teprenone group received 50 mg of teprenone orally thrice daily for 21 days from the initiation of the conditioning regimen. The cumulative incidence of severe aGVHD by day 100 after allo-HSCT was not significantly different in the two groups (27.9 vs. 16.1%, p = 0.25). The exploratory studies revealed no obvious changes in Trx-1 levels, but the alternations from baseline in IL-1β and TNF-α levels at day 28 after allo-HSCT tended to be lower in the teprenone group. In conclusion, we could not demonstrate that teprenone significantly prevented the development of severe aGVHD. Discrepancy with preclinical model suggests that appropriate dose of teprenone may be necessary to induce the expression of antioxidant enzymes that suppress severe aGVHD. Clinical Trial Registration number:jRCTs 061210072.
Collapse
Affiliation(s)
- Wataru Kitamura
- Department of Hematology and Oncology, Okayama University Hospital, 2-5-1 Shikata-Cho, Kita-Ku, Okayama, 700-8558, Japan
- Division of Transfusion and Cell Therapy, Okayama University Hospital, Okayama, Japan
| | - Keiko Fujii
- Department of Hematology and Oncology, Okayama University Hospital, 2-5-1 Shikata-Cho, Kita-Ku, Okayama, 700-8558, Japan.
- Division of Clinical Laboratory, Okayama University Hospital, Okayama, Japan.
| | - Mitsuru Tsuge
- Department of Pediatric Acute Diseases, Okayama University Academic Field of Medicine Dentistry, and Pharmaceutical Sciences, Okayama, Japan
| | - Toshiharu Mitsuhashi
- Center for Innovative Clinical Medicine, Okayama University Hospital, Okayama, Japan
| | - Hiroki Kobayashi
- Department of Hematology and Oncology, Okayama University Hospital, 2-5-1 Shikata-Cho, Kita-Ku, Okayama, 700-8558, Japan
| | - Chihiro Kamoi
- Department of Hematology and Oncology, Okayama University Hospital, 2-5-1 Shikata-Cho, Kita-Ku, Okayama, 700-8558, Japan
- Division of Transfusion and Cell Therapy, Okayama University Hospital, Okayama, Japan
| | - Akira Yamamoto
- Department of Hematology and Oncology, Okayama University Hospital, 2-5-1 Shikata-Cho, Kita-Ku, Okayama, 700-8558, Japan
| | - Takumi Kondo
- Department of Hematology and Oncology, Okayama University Hospital, 2-5-1 Shikata-Cho, Kita-Ku, Okayama, 700-8558, Japan
| | - Keisuke Seike
- Department of Hematology and Oncology, Okayama University Hospital, 2-5-1 Shikata-Cho, Kita-Ku, Okayama, 700-8558, Japan
| | - Hideaki Fujiwara
- Department of Hematology and Oncology, Okayama University Hospital, 2-5-1 Shikata-Cho, Kita-Ku, Okayama, 700-8558, Japan
| | - Noboru Asada
- Department of Hematology and Oncology, Okayama University Hospital, 2-5-1 Shikata-Cho, Kita-Ku, Okayama, 700-8558, Japan
| | - Daisuke Ennishi
- Department of Hematology and Oncology, Okayama University Hospital, 2-5-1 Shikata-Cho, Kita-Ku, Okayama, 700-8558, Japan
- Center for Comprehensive Genomic Medicine, Okayama University Hospital, Okayama, Japan
| | - Ken-Ichi Matsuoka
- Department of Hematology and Oncology, Okayama University Hospital, 2-5-1 Shikata-Cho, Kita-Ku, Okayama, 700-8558, Japan
| | - Nobuharu Fujii
- Department of Hematology and Oncology, Okayama University Hospital, 2-5-1 Shikata-Cho, Kita-Ku, Okayama, 700-8558, Japan
- Division of Transfusion and Cell Therapy, Okayama University Hospital, Okayama, Japan
| | - Yoshinobu Maeda
- Department of Hematology and Oncology, Okayama University Hospital, 2-5-1 Shikata-Cho, Kita-Ku, Okayama, 700-8558, Japan
| |
Collapse
|
5
|
Rayasam A, Moe A, Kudek M, Shah RK, Yuan CY, Miller JM, Rau M, Patton M, Wanat K, Colonna M, Zamora AE, Drobyski WR. Intestinal epithelium-derived IL-34 reprograms macrophages to mitigate gastrointestinal tract graft-versus-host disease. Sci Transl Med 2025; 17:eadn3963. [PMID: 39937882 DOI: 10.1126/scitranslmed.adn3963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 10/10/2024] [Accepted: 01/08/2025] [Indexed: 02/14/2025]
Abstract
Gastrointestinal (GI) tract graft-versus-host disease (GVHD) is a major complication after allogeneic hematopoietic stem cell transplantation and is attributable to dysregulation that occurs between the effector and regulatory arms of the immune system. Whereas regulatory T cells have a primary role in counterbalancing GVHD-induced inflammation, identifying and harnessing other pathways that promote immune tolerance remain major goals in this disease. Herein, we identified interleukin-34 (IL-34) as an intestinal epithelium-derived cytokine that was able to mitigate the severity of GVHD within the GI tract. Specifically, we observed that the absence of recipient IL-34 production exacerbated GVHD lethality, promoted intestinal epithelial cell death, and compromised barrier integrity. Mechanistically, the absence of host IL-34 skewed donor macrophages toward a proinflammatory phenotype and augmented the accumulation of pathogenic CD4+ granulocyte-macrophage colony-stimulating factor (GM-CSF)+ T cells within the colon. Conversely, the administration of recombinant IL-34 substantially reduced GVHD mortality and inflammation, which was dependent on the expression of apolipoprotein E in donor macrophages. Complementary genetic and imaging approaches in mice demonstrated that intestinal epithelial cells were the relevant source of IL-34. These results were supported by colonic biopsies from patients with GVHD, which displayed IL-34 expression in intestinal epithelial cells and apolipoprotein E in lamina propria macrophages, validating similar cellular localization in humans. These studies indicate that IL-34 acts as a tissue-intrinsic cytokine that regulates GVHD severity in the GI tract and could serve as a potential therapeutic target for amelioration of this disease.
Collapse
Affiliation(s)
- Aditya Rayasam
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Alison Moe
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Matthew Kudek
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Ravi K Shah
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Cheng-Yin Yuan
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - James M Miller
- Department of Pathology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Mary Rau
- Department of Pathology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Mollie Patton
- Department of Pathology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Karolyn Wanat
- Department of Dermatology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Marco Colonna
- Department of Pathology and Immunology, Washington University, Saint Louis, MO 63110, USA
| | - Anthony E Zamora
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - William R Drobyski
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| |
Collapse
|
6
|
Atamas SP, Lockatell V, Todd NW, Papadimitriou JC, Rus V, Lugkey KN, Vogel SN, Toshchakov VY, Luzina IG. Therapeutic targeting of full-length interleukin-33 protein levels with cell-permeable decoy peptides attenuates fibrosis in the bleomycin model in vivo. J Pharmacol Exp Ther 2025; 392:100008. [PMID: 39893007 DOI: 10.1124/jpet.123.002050] [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: 12/01/2023] [Revised: 05/08/2024] [Accepted: 05/20/2024] [Indexed: 06/12/2024] Open
Abstract
Interleukin (IL)-33 has been shown to centrally regulate, among other processes, inflammation and fibrosis. Both intracellular full-length (FLIL33) precursor and extracellular mature cytokine (MIL33) forms exert such regulation, albeit differentially. Drug development efforts to target the IL-33 pathway have focused mostly on MIL33 and its specific cell-surface receptor, ST2, with limited attempts to negotiate the pathophysiological contributions from FLIL33. Furthermore, even a successful strategy for targeting MIL33 effects would arguably benefit from a simultaneous attenuation of the levels of FLIL33, which remains the continuous source of MIL33 supply. We therefore sought to develop an approach to depleting FLIL33 protein levels. We previously reported that the steady-state levels of FLIL33 are controlled in part through its proteasomal degradation and that such regulation can be mapped to a segment in the N-terminal portion of FLIL33. We hypothesized that disruption of this regulation would lead to a decrease in FLIL33 levels, thus inducing a beneficial therapeutic effect in an IL-33-dependent pathology. To test this hypothesis, we designed and tested cell-permeable decoy peptides, which mimic the target N-terminal FLIL33 region. We argued that such mimic peptides would compete with FLIL33 for the components of the native FLIL33 production and maintenance molecular machinery. Administered in the therapeutic regimen to bleomycin-challenged mice, the tested cell-permeable decoy peptides alleviated the overall severity of the disease by restoring body weight loss and attenuating accumulation of collagen in the lungs. This proof-of-principle study lays the foundation for future work toward the development of this prospective therapeutic approach. SIGNIFICANCE STATEMENT: An antifibrotic therapeutic approach is proposed and preclinically tested in mice in vivo based on targeting the full-length IL-33 precursor protein. Peptide fusion constructs consisted of a cell-permeable sequence fused with a sequence mimicking an N-terminal segment of IL-33 precursor that is responsible for this protein's stability. Systemic administration of such peptides to mice in either the acute intratracheal or chronic systemic bleomycin challenge models leads to a decrease in the bleomycin-induced elevations of pulmonary IL-33 and collagen.
Collapse
Affiliation(s)
- Sergei P Atamas
- Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland; Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Virginia Lockatell
- Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland
| | - Nevins W Todd
- Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland; Research Service, Baltimore VA Medical Center, Baltimore, Maryland
| | - John C Papadimitriou
- Department of Pathology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Violeta Rus
- Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland
| | - Katerina N Lugkey
- Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland
| | - Stefanie N Vogel
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Vladimir Y Toshchakov
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Irina G Luzina
- Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland; Research Service, Baltimore VA Medical Center, Baltimore, Maryland.
| |
Collapse
|
7
|
Lema DA, Jakobsson G, Daoud A, Elias D, Talor MV, Rattik S, Grönberg C, Kalinoski H, Jaensson Gyllenbäck E, Wang N, Liberg D, Schiopu A, Čiháková D. IL1RAP Blockade With a Monoclonal Antibody Reduces Cardiac Inflammation and Preserves Heart Function in Viral and Autoimmune Myocarditis. Circ Heart Fail 2024; 17:e011729. [PMID: 39513273 PMCID: PMC11643131 DOI: 10.1161/circheartfailure.124.011729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Accepted: 09/24/2024] [Indexed: 11/15/2024]
Abstract
BACKGROUND Currently, there are no therapies targeting specific pathogenic pathways in myocarditis. IL (interleukin)-1 blockade has shown promise in preclinical studies and case reports. We hypothesized that blockade of IL1RAP (IL-1 receptor accessory protein), a shared subunit of the IL-1, IL-33, and IL-36 receptors, could be more efficient than IL-1 blockade alone. METHODS We induced coxsackievirus B3 (CVB3)-mediated or experimental autoimmune myocarditis (EAM) in BALB/c mice, followed by treatment with an Fc (fragment crystallizable)-modified mIgG2a mouse anti-mouse IL1RAP monoclonal antibody (mCAN10). Myocarditis severity and immune infiltration were assessed by histology and flow cytometry. Cardiac function was measured by echocardiography. We used spatial transcriptomics (Visium 10× Genomics) to compare the gene expression landscape in the hearts of mCAN10-treated versus control mice. RESULTS IL1RAP blockade reduced CVB3 and EAM severity. In EAM, the treatment prevented deterioration of cardiac function, measured on day 42 post-disease induction (left ventricular ejection fraction: 56.5% versus 51.0% in isotype controls [P=0.002] and versus 51.4% in mice treated with anti-IL-1β antibodies alone [P=0.003]; n=10-11 mice per group). In the CVB3 model, mCAN10 did not impede viral clearance from the heart and significantly lowered the numbers of CD4+ (cluster of differentiation 4) T cells (P=0.025), inflammatory Ly6C+CCR2+ (lymphocyte antigen 6 complex, locus C/C-C motif chemokine receptor 2) monocytes (P=0.038), neutrophils (P=0.001) and eosinophils (P<0.001) infiltrating the myocardium. The spatial transcriptomic analysis revealed reduced canonical IL-1 signaling and chemokine expression in cardiac immune foci in CVB3-infected mice treated with IL1RAP blockade. CONCLUSIONS Blocking IL1RAP reduces acute CVB3 myocarditis and EAM severity and preserves cardiac function in EAM. We conclude that IL1RAP blockade is a potential therapeutic strategy in viral and autoimmune myocarditis.
Collapse
Affiliation(s)
- Diego A. Lema
- Department of Pathology, School of Medicine (D.A.L., D.E., M.V.T., D.Č.), Johns Hopkins University, Baltimore, MD
| | - Gabriel Jakobsson
- Department of Translational Medicine, Lund University, Malmö, Sweden (G.J., A.S.)
| | - Abdel Daoud
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health (A.D., H.K., D.Č.), Johns Hopkins University, Baltimore, MD
| | - David Elias
- Department of Pathology, School of Medicine (D.A.L., D.E., M.V.T., D.Č.), Johns Hopkins University, Baltimore, MD
| | - Monica V. Talor
- Department of Pathology, School of Medicine (D.A.L., D.E., M.V.T., D.Č.), Johns Hopkins University, Baltimore, MD
| | - Sara Rattik
- Cantargia AB, Lund, Sweden (S.R., C.G., E.J.G., D.L.)
| | | | - Hannah Kalinoski
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health (A.D., H.K., D.Č.), Johns Hopkins University, Baltimore, MD
| | | | - Nadan Wang
- Department of Cardiology, School of Medicine (N.W.), Johns Hopkins University, Baltimore, MD
| | - David Liberg
- Cantargia AB, Lund, Sweden (S.R., C.G., E.J.G., D.L.)
| | - Alexandru Schiopu
- Department of Translational Medicine, Lund University, Malmö, Sweden (G.J., A.S.)
- Internal Medicine Clinic, Skåne University Hospital, Lund, Sweden (A.S.)
- “Nicolae Simionescu” Institute of Cellular Biology and Pathology, Bucharest, Romania (A.S.)
| | - Daniela Čiháková
- Department of Pathology, School of Medicine (D.A.L., D.E., M.V.T., D.Č.), Johns Hopkins University, Baltimore, MD
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health (A.D., H.K., D.Č.), Johns Hopkins University, Baltimore, MD
| |
Collapse
|
8
|
Wu Y, Sun G, Tang B, Song K, Cheng Y, Tu M, Zhu X. Ruxolitinib Plus Basiliximab Therapy for Steroid-Refractory Acute Graft-Versus-Host Disease in Unrelated Cord Blood Transplantation: A Large-Scale Study. Transplant Cell Ther 2024; 30:916.e1-916.e12. [PMID: 38971463 DOI: 10.1016/j.jtct.2024.06.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Revised: 06/11/2024] [Accepted: 06/25/2024] [Indexed: 07/08/2024]
Abstract
Steroid-refractory (SR) acute graft-versus-host disease (aGVHD) is a major cause of mortality after allogeneic hematopoietic stem cell transplantation. We aimed to evaluate the effectiveness and safety of ruxolitinib plus basiliximab for treating SR-aGVHD after unrelated cord blood transplantation (UCBT). Among the 1154 patients with hematological malignancies who underwent UCBT between February 2014 and May 2022, 198 patients with grade II to IV SR-aGVHD were enrolled, 112 of whom were treated with basiliximab alone (basiliximab group) and 86 of whom received basiliximab plus ruxolitinib (combined therapy group). The combined therapy group demonstrated a significantly higher complete response rate (CRR) on day 28 (36.0%) than did the basiliximab group (12.5%, P < .001). SR-aGVHD patients were further stratified into standard-risk and high-risk groups using the refined Minnesota aGVHD risk score. For standard-risk patients, combined therapy significantly improved the CRR (51.1% versus 13.6%, P < .001) and 3-year overall survival (74.5% versus 52.4%, P = .033). However, high-risk patients did not exhibit the same benefits. Compared with basiliximab monotherapy, ruxolitinib plus basiliximab therapy was an effective therapy for patients with standard-risk SR-aGVHD following UCBT. The effectiveness of combined therapy in high-risk patients was not apparent, indicating the need for other treatments.
Collapse
Affiliation(s)
- Yue Wu
- Department of Hematology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China; Anhui Provincial Key Laboratory of Blood Research and Applications, Hefei, China; Blood and Cell Therapy Institute, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Guangyu Sun
- Department of Hematology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Baolin Tang
- Department of Hematology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China; Anhui Provincial Key Laboratory of Blood Research and Applications, Hefei, China; Blood and Cell Therapy Institute, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Kaidi Song
- Department of Hematology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Yaxin Cheng
- Department of Hematology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Meijuan Tu
- Department of Hematology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Xiaoyu Zhu
- Department of Hematology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China; Anhui Provincial Key Laboratory of Blood Research and Applications, Hefei, China; Blood and Cell Therapy Institute, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.
| |
Collapse
|
9
|
Vijayan V, Yan H, Lohmeyer JK, Prentiss KA, Patil RV, Barbarito G, Lopez I, Elezaby A, Peterson K, Baker J, Ostberg NP, Bertaina A, Negrin RS, Mochly-Rosen D, Weinberg K, Haileselassie B. Extracellular release of damaged mitochondria induced by prehematopoietic stem cell transplant conditioning exacerbates GVHD. Blood Adv 2024; 8:3691-3704. [PMID: 38701354 PMCID: PMC11284707 DOI: 10.1182/bloodadvances.2023012328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 03/07/2024] [Accepted: 04/12/2024] [Indexed: 05/05/2024] Open
Abstract
ABSTRACT Despite therapeutic advancements, graft-versus-host disease (GVHD) is a major complication of hematopoietic stem cell transplantation (HSCT). In current models of GVHD, tissue injury induced by cytotoxic conditioning regimens, along with translocation of microbes expressing pathogen-associated molecular patterns, result in activation of host antigen-presenting cells (APCs) to stimulate alloreactive donor T lymphocytes. Recent studies have demonstrated that in many pathologic states, tissue injury results in the release of mitochondria from the cytoplasm to the extracellular space. We hypothesized that extracellular mitochondria, which are related to archaebacteria, could also trigger GVHD by stimulation of host APCs. We found that clinically relevant doses of radiation or busulfan induced extracellular release of mitochondria by various cell types, including cultured intestinal epithelial cells. Conditioning-mediated mitochondrial release was associated with mitochondrial damage and impaired quality control but did not affect the viability of the cells. Extracellular mitochondria directly stimulated host APCs to express higher levels of major histocompatibility complex II (MHC-II), costimulatory CD86, and proinflammatory cytokines, resulting in increased donor T-cell activation, and proliferation in mixed lymphocyte reactions. Analyses of plasma from both experimental mice and a cohort of children undergoing HSCT demonstrated that conditioning induced extracellular mitochondrial release in vivo. In mice undergoing MHC-mismatched HSCT, administration of purified syngeneic extracellular mitochondria increased host APC activation and exacerbated GVHD. Our data suggest that pre-HSCT conditioning results in extracellular release of damaged mitochondria, which increase alloreactivity and exacerbate GVHD. Therefore, decreasing the extracellular release of damaged mitochondria after conditioning could serve as a novel strategy for GVHD prevention.
Collapse
Affiliation(s)
- Vijith Vijayan
- Division of Critical Care Medicine, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA
| | - Hao Yan
- Division of Blood and Marrow Transplantation and Cellular Therapy, Department of Medicine, Stanford University School of Medicine, Stanford, CA
| | - Juliane K. Lohmeyer
- Division of Blood and Marrow Transplantation and Cellular Therapy, Department of Medicine, Stanford University School of Medicine, Stanford, CA
| | - Kaylin A. Prentiss
- Division of Hematology, Oncology, Stem Cell Transplantation and Regenerative Medicine, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA
| | - Rachna V. Patil
- Division of Hematology, Oncology, Stem Cell Transplantation and Regenerative Medicine, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA
| | - Giulia Barbarito
- Division of Hematology, Oncology, Stem Cell Transplantation and Regenerative Medicine, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA
| | - Ivan Lopez
- Division of Critical Care Medicine, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA
| | - Aly Elezaby
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, CA
| | - Kolten Peterson
- Division of Hematology, Oncology, Stem Cell Transplantation and Regenerative Medicine, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA
| | - Jeanette Baker
- Division of Blood and Marrow Transplantation and Cellular Therapy, Department of Medicine, Stanford University School of Medicine, Stanford, CA
| | - Nicolai P. Ostberg
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, CA
| | - Alice Bertaina
- Division of Hematology, Oncology, Stem Cell Transplantation and Regenerative Medicine, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA
| | - Robert S. Negrin
- Division of Blood and Marrow Transplantation and Cellular Therapy, Department of Medicine, Stanford University School of Medicine, Stanford, CA
| | - Daria Mochly-Rosen
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, CA
| | - Kenneth Weinberg
- Division of Hematology, Oncology, Stem Cell Transplantation and Regenerative Medicine, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA
| | - Bereketeab Haileselassie
- Division of Critical Care Medicine, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA
| |
Collapse
|
10
|
DeFilipp Z, Kim HT, Spyrou N, Katsivelos N, Kowalyk S, Eng G, Kasikis S, Beheshti R, Baez J, Akahoshi Y, Ayuk F, Choe H, Etra A, Grupp SA, Hexner EO, Hogan WJ, Kitko CL, Qayed M, Reshef R, Vasova I, Zeiser R, Young R, Holler E, Ferrara JLM, Nakamura R, Levine JE, Chen YB. The MAGIC algorithm probability predicts treatment response and long-term outcomes to second-line therapy for acute GVHD. Blood Adv 2024; 8:3488-3496. [PMID: 38640197 PMCID: PMC11260849 DOI: 10.1182/bloodadvances.2024012561] [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: 01/16/2024] [Revised: 04/02/2024] [Accepted: 04/02/2024] [Indexed: 04/21/2024] Open
Abstract
ABSTRACT The significance of biomarkers in second-line treatment for acute graft-versus-host disease (GVHD) has not been well characterized. We analyzed clinical data and serum samples at the initiation of second-line systemic treatment of acute GVHD from 167 patients from 17 centers of the Mount Sinai Acute GVHD International Consortium (MAGIC) between 2016 and 2021. Sixty-two patients received ruxolitinib-based therapy, whereas 102 received other systemic agents. In agreement with prospective trials, ruxolitinib resulted in a higher day 28 (D28) overall response Frate than nonruxolitinib therapies (55% vs 31%, P = .003) and patients who received ruxolitinib had significantly lower nonrelapse mortality (NRM) than those who received nonruxolitinib therapies (point estimates at 2-year: 35% vs 61%, P = .002). Biomarker analyses demonstrated that the benefit from ruxolitinib was observed only in patients with low MAGIC algorithm probabilities (MAPs) at the start of second-line treatment. Among patients with a low MAP, those who received ruxolitinib experienced significantly lower NRM than those who received nonruxolitinib therapies (point estimates at 2-year: 12% vs 41%, P = .016). However, patients with high MAP experienced high NRM regardless of treatment with ruxolitinib or nonruxolitinib therapies (point estimates at 2-year: 67% vs 80%, P = .65). A landmark analysis demonstrated that the relationship between the D28 response and NRM largely depends on the MAP level at the initiation of second-line therapy. In conclusion, MAP measured at second-line systemic treatment for acute GVHD predicts treatment response and NRM. The outcomes of patients with high MAP are poor regardless of treatment choice, and ruxolitinib appears to primarily benefit patients with low MAP.
Collapse
Affiliation(s)
- Zachariah DeFilipp
- Hematopoietic Cell Transplant and Cellular Therapy Program, Massachusetts General Hospital, Boston, MA
| | - Haesook T. Kim
- Department of Data Science, Dana-Farber Cancer Institute and Harvard T.H. Chan School of Public Health, Boston, MA
| | - Nikolaos Spyrou
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Nikolaos Katsivelos
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Steven Kowalyk
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Gilbert Eng
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Stelios Kasikis
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Rahnuma Beheshti
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Janna Baez
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Yu Akahoshi
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Francis Ayuk
- Department of Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Hannah Choe
- Division of Hematology, James Cancer Center, The Ohio State University, Columbus, OH
| | - Aaron Etra
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Stephan A. Grupp
- Division of Oncology, Children’s Hospital of Philadelphia, Philadelphia, PA
| | - Elizabeth O. Hexner
- Blood and Marrow Transplantation Program, Abramson Cancer Center, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | | | - Carrie L. Kitko
- Pediatric Stem Cell Transplant Program, Vanderbilt University Medical Center, Nashville, TN
| | - Muna Qayed
- Aflac Cancer and Blood Disorders Center, Emory University, Atlanta, GA
| | - Ran Reshef
- Blood and Marrow Transplantation Program, Columbia University Medical Center, New York, NY
| | - Ingrid Vasova
- Department of Internal Medicine 5, Hematology/Oncology, University Hospital Erlangen, Erlangen, Germany
| | - Robert Zeiser
- Department of Medicine I - Medical Centre, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Rachel Young
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Ernst Holler
- Department of Hematology and Oncology, Internal Medicine III, University of Regensburg, Regensburg, Germany
| | - James L. M. Ferrara
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Ryotaro Nakamura
- Hematology/Hematopoietic Cell Transplant, City of Hope National Medical Center, Duarte, CA
| | - John E. Levine
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Yi-Bin Chen
- Hematopoietic Cell Transplant and Cellular Therapy Program, Massachusetts General Hospital, Boston, MA
| |
Collapse
|
11
|
Etra A, El Jurdi N, Katsivelos N, Kwon D, Gergoudis S, Morales G, Spyrou N, Kowalyk S, Aguayo-Hiraldo P, Akahoshi Y, Ayuk F, Baez J, Betts BC, Chanswangphuwana C, Chen YB, Choe H, DeFilipp Z, Gleich S, Hexner E, Hogan WJ, Holler E, Kitko CL, Kraus S, Al Malki M, MacMillan M, Pawarode A, Quagliarella F, Qayed M, Reshef R, Schechter T, Vasova I, Weisdorf D, Wölfl M, Young R, Nakamura R, Ferrara JLM, Levine JE, Holtan S. Amphiregulin, ST2, and REG3α biomarker risk algorithms as predictors of nonrelapse mortality in patients with acute GVHD. Blood Adv 2024; 8:3284-3292. [PMID: 38640195 PMCID: PMC11226972 DOI: 10.1182/bloodadvances.2023011049] [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: 06/27/2023] [Revised: 03/29/2024] [Accepted: 03/29/2024] [Indexed: 04/21/2024] Open
Abstract
ABSTRACT Graft-versus-host disease (GVHD) is a major cause of nonrelapse mortality (NRM) after allogeneic hematopoietic cell transplantation. Algorithms containing either the gastrointestinal (GI) GVHD biomarker amphiregulin (AREG) or a combination of 2 GI GVHD biomarkers (suppressor of tumorigenicity-2 [ST2] + regenerating family member 3 alpha [REG3α]) when measured at GVHD diagnosis are validated predictors of NRM risk but have never been assessed in the same patients using identical statistical methods. We measured the serum concentrations of ST2, REG3α, and AREG by enzyme-linked immunosorbent assay at the time of GVHD diagnosis in 715 patients divided by the date of transplantation into training (2004-2015) and validation (2015-2017) cohorts. The training cohort (n = 341) was used to develop algorithms for predicting the probability of 12-month NRM that contained all possible combinations of 1 to 3 biomarkers and a threshold corresponding to the concordance probability was used to stratify patients for the risk of NRM. Algorithms were compared with each other based on several metrics, including the area under the receiver operating characteristics curve, proportion of patients correctly classified, sensitivity, and specificity using only the validation cohort (n = 374). All algorithms were strong discriminators of 12-month NRM, whether or not patients were systemically treated (n = 321). An algorithm containing only ST2 + REG3α had the highest area under the receiver operating characteristics curve (0.757), correctly classified the most patients (75%), and more accurately risk-stratified those who developed Minnesota standard-risk GVHD and for patients who received posttransplant cyclophosphamide-based prophylaxis. An algorithm containing only AREG more accurately risk-stratified patients with Minnesota high-risk GVHD. Combining ST2, REG3α, and AREG into a single algorithm did not improve performance.
Collapse
Affiliation(s)
- Aaron Etra
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Najla El Jurdi
- Hematology, Oncology and Transplant, University of Minnesota, Minneapolis, MN
| | - Nikolaos Katsivelos
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Deukwoo Kwon
- Department of Population Health Science and Policy, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Stephanie Gergoudis
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - George Morales
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Nikolaos Spyrou
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Steven Kowalyk
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Paibel Aguayo-Hiraldo
- Division of Hematology, Oncology, and Blood and Marrow Transplantation, Children's Hospital Los Angeles, Los Angeles, CA
| | - Yu Akahoshi
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Francis Ayuk
- Department of Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Janna Baez
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Brian C. Betts
- Hematology, Oncology and Transplant, University of Minnesota, Minneapolis, MN
| | | | - Yi-Bin Chen
- Hematopoietic Cell Transplant and Cellular Therapy Program, Massachusetts General Hospital, Boston, MA
| | - Hannah Choe
- Division of Hematology, James Cancer Center, The Ohio State University, Columbus, OH
| | - Zachariah DeFilipp
- Hematopoietic Cell Transplant and Cellular Therapy Program, Massachusetts General Hospital, Boston, MA
| | - Sigrun Gleich
- Department of Hematology and Oncology, Internal Medicine III, University of Regensburg, Regensburg, Germany
| | - Elizabeth Hexner
- Blood and Marrow Transplantation Program, Abramson Cancer Center, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | | | - Ernst Holler
- Department of Hematology and Oncology, Internal Medicine III, University of Regensburg, Regensburg, Germany
| | - Carrie L. Kitko
- Pediatric Stem Cell Transplant Program, Vanderbilt University Medical Center, Nashville, TN
| | - Sabrina Kraus
- Department of Internal Medicine II, University Hospital of Würzburg, Würzburg, Germany
| | - Monzr Al Malki
- Hematology/Hematopoietic Cell Transplant, City of Hope National Medical Center, Duarte, CA
| | - Margaret MacMillan
- Hematology, Oncology and Transplant, University of Minnesota, Minneapolis, MN
| | - Attaphol Pawarode
- Blood and Marrow Transplantation Program, University of Michigan, Ann Arbor, MI
| | | | - Muna Qayed
- Aflac Cancer and Blood Disorders Center, Emory University, Atlanta, GA
| | - Ran Reshef
- Blood and Marrow Transplantation Program, Columbia University Medical Center, New York, NY
| | - Tal Schechter
- Division of Hematology/Oncology/BMT, The Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
| | - Ingrid Vasova
- Med. Klinik III/Poliklinik, Universitatsklinik Erlangen, Erlangen, Germany
| | - Daniel Weisdorf
- Hematology, Oncology and Transplant, University of Minnesota, Minneapolis, MN
| | - Matthias Wölfl
- Pediatric Blood and Marrow Transplantation Program, Children’s Hospital, University of Würzburg, Würzburg, Germany
| | - Rachel Young
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Ryotaro Nakamura
- Hematology/Hematopoietic Cell Transplant, City of Hope National Medical Center, Duarte, CA
| | - James L. M. Ferrara
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - John E. Levine
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Shernan Holtan
- Hematology, Oncology and Transplant, University of Minnesota, Minneapolis, MN
| |
Collapse
|
12
|
Hong S, Sarantopoulos S. A More MAGICal Alogrithm in Acute GVHD. Transplant Cell Ther 2024; 30:347-348. [PMID: 38604718 DOI: 10.1016/j.jtct.2024.03.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/13/2024]
Affiliation(s)
- Sanghee Hong
- Division of Hematologic Malignancies and Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215; Duke Cancer Institute, Duke University Medical Center, Durham NC 27710
| | - Stefanie Sarantopoulos
- Division of Hematologic Malignancies and Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215; Duke Cancer Institute, Duke University Medical Center, Durham NC 27710.
| |
Collapse
|
13
|
Oravecz-Wilson K, Lauder E, Taylor A, Maneix L, Van Nostrand JL, Sun Y, Li L, Zhao D, Liu C, Reddy P. Autophagy differentially regulates tissue tolerance of distinct target organs in graft-versus-host disease models. J Clin Invest 2024; 134:e167369. [PMID: 38426503 PMCID: PMC10904048 DOI: 10.1172/jci167369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 01/11/2024] [Indexed: 03/02/2024] Open
Abstract
Tissue-intrinsic mechanisms that regulate severity of systemic pathogenic immune-mediated diseases, such as acute graft-versus-host disease (GVHD), remain poorly understood. Following allogeneic hematopoietic stem cell transplantation, autophagy, a cellular stress protective response, is induced in host nonhematopoietic cells. To systematically address the role of autophagy in various host nonhematopoietic tissues, both specific classical target organs of acute GVHD (intestines, liver, and skin) and organs conventionally not known to be targets of GVHD (kidneys and heart), we generated mice with organ-specific knockout of autophagy related 5 (ATG5) to specifically and exclusively inhibit autophagy in the specific organs. When compared with wild-type recipients, animals that lacked ATG5 in the gastrointestinal tract or liver showed significantly greater tissue injury and mortality, while autophagy deficiency in the skin, kidneys, or heart did not affect mortality. Treatment with the systemic autophagy inducer sirolimus only partially mitigated GVHD mortality in intestine-specific autophagy-deficient hosts. Deficiency of autophagy increased MHC class I on the target intestinal epithelial cells, resulting in greater susceptibility to damage by alloreactive T cells. Thus, autophagy is a critical cell-intrinsic protective response that promotes tissue tolerance and regulates GVHD severity.
Collapse
Affiliation(s)
- Katherine Oravecz-Wilson
- Department of Internal Medicine, Division of Hematology and Oncology, University of Michigan, Rogel Cancer Center, Ann Arbor, Michigan, USA
| | - Emma Lauder
- Department of Internal Medicine, Division of Hematology and Oncology, University of Michigan, Rogel Cancer Center, Ann Arbor, Michigan, USA
- Dan L. Duncan Comprehensive Cancer Center and
| | - Austin Taylor
- Department of Internal Medicine, Division of Hematology and Oncology, University of Michigan, Rogel Cancer Center, Ann Arbor, Michigan, USA
| | | | - Jeanine L. Van Nostrand
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, USA
| | - Yaping Sun
- Dan L. Duncan Comprehensive Cancer Center and
| | - Lu Li
- Dan L. Duncan Comprehensive Cancer Center and
| | | | - Chen Liu
- Department of Pathology, Yale School of Medicine, Yale University, New Haven, Connecticut, USA
| | - Pavan Reddy
- Department of Internal Medicine, Division of Hematology and Oncology, University of Michigan, Rogel Cancer Center, Ann Arbor, Michigan, USA
- Dan L. Duncan Comprehensive Cancer Center and
| |
Collapse
|
14
|
Brunner TM, Serve S, Marx AF, Fadejeva J, Saikali P, Dzamukova M, Durán-Hernández N, Kommer C, Heinrich F, Durek P, Heinz GA, Höfer T, Mashreghi MF, Kühn R, Pinschewer DD, Löhning M. A type 1 immunity-restricted promoter of the IL-33 receptor gene directs antiviral T-cell responses. Nat Immunol 2024; 25:256-267. [PMID: 38172258 PMCID: PMC10834369 DOI: 10.1038/s41590-023-01697-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Accepted: 11/02/2023] [Indexed: 01/05/2024]
Abstract
The pleiotropic alarmin interleukin-33 (IL-33) drives type 1, type 2 and regulatory T-cell responses via its receptor ST2. Subset-specific differences in ST2 expression intensity and dynamics suggest that transcriptional regulation is key in orchestrating the context-dependent activity of IL-33-ST2 signaling in T-cell immunity. Here, we identify a previously unrecognized alternative promoter in mice and humans that is located far upstream of the curated ST2-coding gene and drives ST2 expression in type 1 immunity. Mice lacking this promoter exhibit a selective loss of ST2 expression in type 1- but not type 2-biased T cells, resulting in impaired expansion of cytotoxic T cells (CTLs) and T-helper 1 cells upon viral infection. T-cell-intrinsic IL-33 signaling via type 1 promoter-driven ST2 is critical to generate a clonally diverse population of antiviral short-lived effector CTLs. Thus, lineage-specific alternative promoter usage directs alarmin responsiveness in T-cell subsets and offers opportunities for immune cell-specific targeting of the IL-33-ST2 axis in infections and inflammatory diseases.
Collapse
Affiliation(s)
- Tobias M Brunner
- Experimental Immunology and Osteoarthritis Research, Department of Rheumatology and Clinical Immunology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany.
- Pitzer Laboratory of Osteoarthritis Research, German Rheumatism Research Center (DRFZ), a Leibniz Institute, Berlin, Germany.
| | - Sebastian Serve
- Experimental Immunology and Osteoarthritis Research, Department of Rheumatology and Clinical Immunology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Pitzer Laboratory of Osteoarthritis Research, German Rheumatism Research Center (DRFZ), a Leibniz Institute, Berlin, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, BIH Biomedical Innovation Academy, Berlin, Germany
| | - Anna-Friederike Marx
- Division of Experimental Virology, Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Jelizaveta Fadejeva
- Experimental Immunology and Osteoarthritis Research, Department of Rheumatology and Clinical Immunology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Pitzer Laboratory of Osteoarthritis Research, German Rheumatism Research Center (DRFZ), a Leibniz Institute, Berlin, Germany
| | - Philippe Saikali
- Experimental Immunology and Osteoarthritis Research, Department of Rheumatology and Clinical Immunology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Pitzer Laboratory of Osteoarthritis Research, German Rheumatism Research Center (DRFZ), a Leibniz Institute, Berlin, Germany
| | - Maria Dzamukova
- Experimental Immunology and Osteoarthritis Research, Department of Rheumatology and Clinical Immunology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Pitzer Laboratory of Osteoarthritis Research, German Rheumatism Research Center (DRFZ), a Leibniz Institute, Berlin, Germany
| | - Nayar Durán-Hernández
- Experimental Immunology and Osteoarthritis Research, Department of Rheumatology and Clinical Immunology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Pitzer Laboratory of Osteoarthritis Research, German Rheumatism Research Center (DRFZ), a Leibniz Institute, Berlin, Germany
| | - Christoph Kommer
- Division of Theoretical Systems Biology, German Cancer Research Center (DKFZ), Heidelberg, Germany
- BioQuant Center, University of Heidelberg, Heidelberg, Germany
| | - Frederik Heinrich
- Therapeutic Gene Regulation, German Rheumatism Research Center (DRFZ), a Leibniz Institute, Berlin, Germany
| | - Pawel Durek
- Therapeutic Gene Regulation, German Rheumatism Research Center (DRFZ), a Leibniz Institute, Berlin, Germany
| | - Gitta A Heinz
- Therapeutic Gene Regulation, German Rheumatism Research Center (DRFZ), a Leibniz Institute, Berlin, Germany
| | - Thomas Höfer
- Division of Theoretical Systems Biology, German Cancer Research Center (DKFZ), Heidelberg, Germany
- BioQuant Center, University of Heidelberg, Heidelberg, Germany
| | - Mir-Farzin Mashreghi
- Therapeutic Gene Regulation, German Rheumatism Research Center (DRFZ), a Leibniz Institute, Berlin, Germany
| | - Ralf Kühn
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Daniel D Pinschewer
- Division of Experimental Virology, Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Max Löhning
- Experimental Immunology and Osteoarthritis Research, Department of Rheumatology and Clinical Immunology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany.
- Pitzer Laboratory of Osteoarthritis Research, German Rheumatism Research Center (DRFZ), a Leibniz Institute, Berlin, Germany.
| |
Collapse
|
15
|
Essomba RG, Mbe RM, Ngogang MP, Ekono CB, Bitoungui VJN, Seni N, Nguwoh PS, Ateba PT, Kamdem SD, Nono JK, Ambomo MS, Assoumou MCO, Mbopi-Kéou FX. Plasma IL-33 levels and immune activation in HIV-TB coinfection: a cross-sectional study in Yaoundé, Cameroon. Pan Afr Med J 2023; 46:13. [PMID: 38035159 PMCID: PMC10683167 DOI: 10.11604/pamj.2023.46.13.41152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Accepted: 08/22/2023] [Indexed: 12/02/2023] Open
Abstract
Introduction HIV-1 and Mtb are characterized by immune activation and unbalances production of cytokines, but the expression of IL33 in HIV/TB coinfection remain understudied. This study aimed to evaluate the level of IL-33 in plasma of HIV and M. tuberculosis (HIV/TB) coinfected patients compared to patients with respective mono infections in Yaoundé. Methods a cross-sectional study was conducted among patients attending the pneumology service and HIV treatment center of the Yaoundé Jamot Hospital. Plasma samples of 157 HIV/TB coinfected patients (n =26, 50% males and 50% females, mean age 39), HIV-1 monoinfected patients (n = 41, 41% males and 59% females, mean age 35), TB monoinfected patients (n = 48, 56% males and 44% females, mean age 37) and healthy controls (n = 42, 29% males and 71% females, mean age 32) were examined by enzyme-linked immunoassay (ELISA) to detect the levels of IL-33 cytokine. Results plasma level of IL-33 were higher in HIV/TB coinfected (33.1±30.9 pg/ml) and TB monoinfected individuals (15.1±2.9 pg/ml) compared to healthy controls (14.0±3.4 pg/ml) and could not be detected in most of the HIV-1 monoinfected individuals (12.6±8.7 pg/ml). Interestingly, the increased plasma level of IL-33 in HIV/TB coinfected patients showed a statistically significant difference between healthy controls (33.1±30.9 pg/ml vs 14.0±3.4 pg/ml, P<0.0001) and HIV-1 monoinfected patients (33.1±30.9 pg/ml vs 12.6±8.7 pg/ml, P=0.0002). We further found that IL-33 was higher in patients with high viral load group (40.6±59.7 pg/ml vs 12.6±1.8 pg/ml), P= 0.47) whereas patients under highly active antiretroviral therapy (HAART) showed decreased level of IL-33 concentration as the number of years under ART increased. Our data showed a positive association between plasma IL-33 and viral load in the context of HIV/TB coinfection in our study population with a positive Pearson coefficient of r=0.21. Conclusion this study indicates that plasma level of IL-33 differs among HIV/TB coinfected patients and respective monoinfections patients. The increased level of plasma IL-33 reveals that IL-33 measurement in HIV-1 monoinfected patients may represent an early predictor of development of tuberculosis.
Collapse
Affiliation(s)
- René Ghislain Essomba
- National Public Health Laboratory (NPHL), Ministry of Public Health, Yaoundé, Cameroon
- Faculty of Medicine and Biomedical Sciences (FMBS), University of Yaoundé I, Yaoundé, Cameroon
- School of Health Sciences, Catholic University of Central Africa, Yaoundé, Cameroon
| | - Rostand Munkam Mbe
- School of Health Sciences, Catholic University of Central Africa, Yaoundé, Cameroon
| | - Marie Paule Ngogang
- Faculty of Medicine and Biomedical Sciences (FMBS), University of Yaoundé I, Yaoundé, Cameroon
| | - Claire Bitchong Ekono
- Faculty of Medicine and Pharmaceutical Sciences (FMPS), University of Douala, Douala, Cameroon
- Pneumology Service, Jamot Hospital of Yaoundé, Yaoundé, Cameroon
| | | | - Nassif Seni
- School of Health Sciences, Catholic University of Central Africa, Yaoundé, Cameroon
| | | | | | - Severin Donald Kamdem
- Faculty of Medicine and Pharmaceutical Sciences (FMPS), University of Dschang, Dschang, Cameroon
- Institute of Medical Research and Medicinal Plant Studies (IMPM), Ministry of Scientific Research and Innovation, Yaoundé, Cameroon
| | - Justin Komguep Nono
- Faculty of Medicine and Pharmaceutical Sciences (FMPS), University of Dschang, Dschang, Cameroon
- Division of Immunology, Health Science Faculty, University of Cape Town, Cape Town, South Africa
| | - Myriam Sylvie Ambomo
- School of Health Sciences, Catholic University of Central Africa, Yaoundé, Cameroon
| | - Marie Claire Okomo Assoumou
- National Public Health Laboratory (NPHL), Ministry of Public Health, Yaoundé, Cameroon
- Faculty of Medicine and Biomedical Sciences (FMBS), University of Yaoundé I, Yaoundé, Cameroon
| | | |
Collapse
|
16
|
Cassano A, Chong AS, Alegre ML. Tregs in transplantation tolerance: role and therapeutic potential. FRONTIERS IN TRANSPLANTATION 2023; 2:1217065. [PMID: 38993904 PMCID: PMC11235334 DOI: 10.3389/frtra.2023.1217065] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Accepted: 08/14/2023] [Indexed: 07/13/2024]
Abstract
CD4+ Foxp3+ regulatory T cells (Tregs) are indispensable for preventing autoimmunity, and they play a role in cancer and transplantation settings by restraining immune responses. In this review, we describe evidence for the importance of Tregs in the induction versus maintenance of transplantation tolerance, discussing insights into mechanisms of Treg control of the alloimmune response. Further, we address the therapeutic potential of Tregs as a clinical intervention after transplantation, highlighting engineered CAR-Tregs as well as expansion of donor and host Tregs.
Collapse
Affiliation(s)
- Alexandra Cassano
- Department of Medicine, University of Chicago, Chicago, IL, United States
| | - Anita S. Chong
- Department of Surgery, University of Chicago, Chicago, IL, United States
| | - Maria-Luisa Alegre
- Department of Medicine, University of Chicago, Chicago, IL, United States
| |
Collapse
|
17
|
Cao Y, Wang J, Jiang S, Lyu M, Zhao F, Liu J, Wang M, Pei X, Zhai W, Feng X, Feng S, Han M, Xu Y, Jiang E. JAK1/2 inhibitor ruxolitinib promotes the expansion and suppressive action of polymorphonuclear myeloid-derived suppressor cells via the JAK/STAT and ROS-MAPK/NF-κB signalling pathways in acute graft-versus-host disease. Clin Transl Immunology 2023; 12:e1441. [PMID: 36855558 PMCID: PMC9968240 DOI: 10.1002/cti2.1441] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 11/27/2022] [Accepted: 02/07/2023] [Indexed: 02/27/2023] Open
Abstract
Objectives Ruxolitinib, a Janus kinase (JAK) 1/2 inhibitor, demonstrates efficacy for treating steroid-resistant acute graft-versus-host disease (SR-aGVHD) following allogeneic stem cell transplantation (allo-HSCT). Myeloid-derived suppressor cells (MDSCs) have a protective effect on aGVHD via suppressing T cell function. However, the precise features and mechanism of JAK inhibitor-mediated immune modulation on MDSCs subsets remain poorly understood. Methods A total of 74 SR-aGVHD patients treated with allo-HSCT and ruxolitinib were enrolled in the present study. The alterations of MDSC and regulatory T cell (Treg) populations were monitored during ruxolitinib treatment in responders and nonresponders. A mouse model of aGVHD was used to evaluate the immunosuppressive activity of MDSCs and related signalling pathways in response to ruxolitinib administration in vivo and in vitro. Results Patients with SR-aGVHD who received ruxolitinib treatment achieved satisfactory outcomes. Elevation proportions of MDSCs before treatment, especially polymorphonuclear-MDSCs (PMN-MDSCs) were better to reflect the response to ruxolitinib than those in Tregs. In the mouse model of aGVHD, the administration of ruxolitinib resulted in the expansion and functional enhancement of PMN-MDSCs and the effects could be partially reversed by an anti-Gr-1 antibody in vivo. Ruxolitinib treatment significantly elevated the suppressive function of PMN-MDSCs through reactive oxygen species (ROS) production by Nox2 upregulation as well as bypassing the activated MAPK/NF-κB signalling pathway. Additionally, ex vivo experiments demonstrated that ruxolitinib prevented the differentiation of mature myeloid cells and promoted the accumulation of MDSCs by inhibiting STAT5. Conclusions Ruxolitinib enhances PMN-MDSCs functions through JAK/STAT and ROS-MAPK/NF-κB signalling pathways. Monitoring frequencies and functions of MDSCs can help evaluate treatment responses to ruxolitinib.
Collapse
Affiliation(s)
- Yigeng Cao
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases HospitalChinese Academy of Medical Sciences & Peking Union Medical CollegeTianjinChina,Tianjin Institutes of Health ScienceTianjinChina,Hematopoietic Stem Cell Transplantation Center, Institute of Hematology & Blood Diseases HospitalChinese Academy of Medical Sciences & Peking Union Medical CollegeTianjinChina
| | - Jiali Wang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases HospitalChinese Academy of Medical Sciences & Peking Union Medical CollegeTianjinChina,Tianjin Institutes of Health ScienceTianjinChina,Hematopoietic Stem Cell Transplantation Center, Institute of Hematology & Blood Diseases HospitalChinese Academy of Medical Sciences & Peking Union Medical CollegeTianjinChina
| | - Shan Jiang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases HospitalChinese Academy of Medical Sciences & Peking Union Medical CollegeTianjinChina,Tianjin Institutes of Health ScienceTianjinChina
| | - Mengnan Lyu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases HospitalChinese Academy of Medical Sciences & Peking Union Medical CollegeTianjinChina,Tianjin Institutes of Health ScienceTianjinChina,Hematopoietic Stem Cell Transplantation Center, Institute of Hematology & Blood Diseases HospitalChinese Academy of Medical Sciences & Peking Union Medical CollegeTianjinChina
| | - Fei Zhao
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases HospitalChinese Academy of Medical Sciences & Peking Union Medical CollegeTianjinChina,Tianjin Institutes of Health ScienceTianjinChina,Hematopoietic Stem Cell Transplantation Center, Institute of Hematology & Blood Diseases HospitalChinese Academy of Medical Sciences & Peking Union Medical CollegeTianjinChina
| | - Jia Liu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases HospitalChinese Academy of Medical Sciences & Peking Union Medical CollegeTianjinChina,Tianjin Institutes of Health ScienceTianjinChina,Hematopoietic Stem Cell Transplantation Center, Institute of Hematology & Blood Diseases HospitalChinese Academy of Medical Sciences & Peking Union Medical CollegeTianjinChina
| | - Mingyang Wang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases HospitalChinese Academy of Medical Sciences & Peking Union Medical CollegeTianjinChina,Tianjin Institutes of Health ScienceTianjinChina,Hematopoietic Stem Cell Transplantation Center, Institute of Hematology & Blood Diseases HospitalChinese Academy of Medical Sciences & Peking Union Medical CollegeTianjinChina
| | - Xiaolei Pei
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases HospitalChinese Academy of Medical Sciences & Peking Union Medical CollegeTianjinChina,Tianjin Institutes of Health ScienceTianjinChina,Hematopoietic Stem Cell Transplantation Center, Institute of Hematology & Blood Diseases HospitalChinese Academy of Medical Sciences & Peking Union Medical CollegeTianjinChina
| | - Weihua Zhai
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases HospitalChinese Academy of Medical Sciences & Peking Union Medical CollegeTianjinChina,Tianjin Institutes of Health ScienceTianjinChina,Hematopoietic Stem Cell Transplantation Center, Institute of Hematology & Blood Diseases HospitalChinese Academy of Medical Sciences & Peking Union Medical CollegeTianjinChina
| | - Xiaoming Feng
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases HospitalChinese Academy of Medical Sciences & Peking Union Medical CollegeTianjinChina,Tianjin Institutes of Health ScienceTianjinChina
| | - Sizhou Feng
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases HospitalChinese Academy of Medical Sciences & Peking Union Medical CollegeTianjinChina,Tianjin Institutes of Health ScienceTianjinChina,Hematopoietic Stem Cell Transplantation Center, Institute of Hematology & Blood Diseases HospitalChinese Academy of Medical Sciences & Peking Union Medical CollegeTianjinChina
| | - Mingzhe Han
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases HospitalChinese Academy of Medical Sciences & Peking Union Medical CollegeTianjinChina,Tianjin Institutes of Health ScienceTianjinChina,Hematopoietic Stem Cell Transplantation Center, Institute of Hematology & Blood Diseases HospitalChinese Academy of Medical Sciences & Peking Union Medical CollegeTianjinChina
| | - Yuanfu Xu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases HospitalChinese Academy of Medical Sciences & Peking Union Medical CollegeTianjinChina,Tianjin Institutes of Health ScienceTianjinChina
| | - Erlie Jiang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases HospitalChinese Academy of Medical Sciences & Peking Union Medical CollegeTianjinChina,Tianjin Institutes of Health ScienceTianjinChina,Hematopoietic Stem Cell Transplantation Center, Institute of Hematology & Blood Diseases HospitalChinese Academy of Medical Sciences & Peking Union Medical CollegeTianjinChina
| |
Collapse
|
18
|
Marx AF, Kallert SM, Brunner TM, Villegas JA, Geier F, Fixemer J, Abreu-Mota T, Reuther P, Bonilla WV, Fadejeva J, Kreutzfeldt M, Wagner I, Aparicio-Domingo P, Scarpellino L, Charmoy M, Utzschneider DT, Hagedorn C, Lu M, Cornille K, Stauffer K, Kreppel F, Merkler D, Zehn D, Held W, Luther SA, Löhning M, Pinschewer DD. The alarmin interleukin-33 promotes the expansion and preserves the stemness of Tcf-1 + CD8 + T cells in chronic viral infection. Immunity 2023; 56:813-828.e10. [PMID: 36809763 DOI: 10.1016/j.immuni.2023.01.029] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 12/22/2022] [Accepted: 01/27/2023] [Indexed: 02/22/2023]
Abstract
T cell factor 1 (Tcf-1) expressing CD8+ T cells exhibit stem-like self-renewing capacity, rendering them key for immune defense against chronic viral infection and cancer. Yet, the signals that promote the formation and maintenance of these stem-like CD8+ T cells (CD8+SL) remain poorly defined. Studying CD8+ T cell differentiation in mice with chronic viral infection, we identified the alarmin interleukin-33 (IL-33) as pivotal for the expansion and stem-like functioning of CD8+SL as well as for virus control. IL-33 receptor (ST2)-deficient CD8+ T cells exhibited biased end differentiation and premature loss of Tcf-1. ST2-deficient CD8+SL responses were restored by blockade of type I interferon signaling, suggesting that IL-33 balances IFN-I effects to control CD8+SL formation in chronic infection. IL-33 signals broadly augmented chromatin accessibility in CD8+SL and determined these cells' re-expansion potential. Our study identifies the IL-33-ST2 axis as an important CD8+SL-promoting pathway in the context of chronic viral infection.
Collapse
Affiliation(s)
- Anna-Friederike Marx
- Department of Biomedicine, Division of Experimental Virology, University of Basel, 4055 Basel, Switzerland.
| | - Sandra M Kallert
- Department of Biomedicine, Division of Experimental Virology, University of Basel, 4055 Basel, Switzerland
| | - Tobias M Brunner
- Experimental Immunology and Osteoarthritis Research, Department of Rheumatology and Clinical Immunology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany; Pitzer Laboratory of Osteoarthritis Research, German Rheumatism Research Center (DRFZ), a Leibniz Institute, 10117 Berlin, Germany
| | - José A Villegas
- Department of Immunobiology, University of Lausanne, 1066 Epalinges, Switzerland
| | - Florian Geier
- Department of Biomedicine, Bioinformatics Core Facility, University Hospital Basel, 4031 Basel, Switzerland; Swiss Institute of Bioinformatics, Basel, Switzerland
| | - Jonas Fixemer
- Department of Biomedicine, Division of Experimental Virology, University of Basel, 4055 Basel, Switzerland
| | - Tiago Abreu-Mota
- Department of Biomedicine, Division of Experimental Virology, University of Basel, 4055 Basel, Switzerland
| | - Peter Reuther
- Department of Biomedicine, Division of Experimental Virology, University of Basel, 4055 Basel, Switzerland
| | - Weldy V Bonilla
- Department of Biomedicine, Division of Experimental Virology, University of Basel, 4055 Basel, Switzerland
| | - Jelizaveta Fadejeva
- Experimental Immunology and Osteoarthritis Research, Department of Rheumatology and Clinical Immunology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany; Pitzer Laboratory of Osteoarthritis Research, German Rheumatism Research Center (DRFZ), a Leibniz Institute, 10117 Berlin, Germany
| | - Mario Kreutzfeldt
- Department of Pathology and Immunology University of Geneva, Geneva, Switzerland; Division of Clinical Pathology, Geneva University Hospital, 1211 Geneva, Switzerland
| | - Ingrid Wagner
- Department of Pathology and Immunology University of Geneva, Geneva, Switzerland; Division of Clinical Pathology, Geneva University Hospital, 1211 Geneva, Switzerland
| | | | - Leo Scarpellino
- Department of Immunobiology, University of Lausanne, 1066 Epalinges, Switzerland
| | - Mélanie Charmoy
- Department of Oncology, University of Lausanne, 1066 Epalinges, Switzerland
| | - Daniel T Utzschneider
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC 3000, Australia
| | - Claudia Hagedorn
- Witten/Herdecke University (UW/H), Faculty of Health/School of Medicine, Stockumer Str. 10, 58453 Witten, Germany
| | - Min Lu
- Department of Biomedicine, Division of Experimental Virology, University of Basel, 4055 Basel, Switzerland
| | - Karen Cornille
- Department of Biomedicine, Division of Experimental Virology, University of Basel, 4055 Basel, Switzerland
| | - Karsten Stauffer
- Department of Biomedicine, Division of Experimental Virology, University of Basel, 4055 Basel, Switzerland
| | - Florian Kreppel
- Witten/Herdecke University (UW/H), Faculty of Health/School of Medicine, Stockumer Str. 10, 58453 Witten, Germany
| | - Doron Merkler
- Department of Pathology and Immunology University of Geneva, Geneva, Switzerland; Division of Clinical Pathology, Geneva University Hospital, 1211 Geneva, Switzerland
| | - Dietmar Zehn
- Division of Animal Physiology and Immunology, School of Life Sciences Weihenstephan, Technical University of Munich, 85354 Freising, Germany
| | - Werner Held
- Department of Oncology, University of Lausanne, 1066 Epalinges, Switzerland
| | - Sanjiv A Luther
- Department of Immunobiology, University of Lausanne, 1066 Epalinges, Switzerland
| | - Max Löhning
- Experimental Immunology and Osteoarthritis Research, Department of Rheumatology and Clinical Immunology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany; Pitzer Laboratory of Osteoarthritis Research, German Rheumatism Research Center (DRFZ), a Leibniz Institute, 10117 Berlin, Germany.
| | - Daniel D Pinschewer
- Department of Biomedicine, Division of Experimental Virology, University of Basel, 4055 Basel, Switzerland.
| |
Collapse
|
19
|
Balakrishnan B, Kulkarni UP, Pai AA, Illangeswaran RSS, Mohanan E, Mathews V, George B, Balasubramanian P. Biomarkers for early complications post hematopoietic cell transplantation: Insights and challenges. Front Immunol 2023; 14:1100306. [PMID: 36817455 PMCID: PMC9932777 DOI: 10.3389/fimmu.2023.1100306] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Accepted: 01/23/2023] [Indexed: 02/05/2023] Open
Abstract
Hematopoietic cell transplantation is an established curative treatment option for various hematological malignant, and non-malignant diseases. However, the success of HCT is still limited by life-threatening early complications post-HCT, such as Graft Versus Host Disease (GVHD), Sinusoidal Obstruction Syndrome (SOS), and transplant-associated microangiopathy, to name a few. A decade of research in the discovery and validation of novel blood-based biomarkers aims to manage these early complications by using them for diagnosis or prognosis. Advances in this field have also led to predictive biomarkers to identify patients' likelihood of response to therapy. Although biomarkers have been extensively evaluated for different complications, these are yet to be used in routine clinical practice. This review provides a detailed summary of various biomarkers for individual early complications post-HCT, their discovery, validation, ongoing clinical trials, and their limitations. Furthermore, this review also provides insights into the biology of biomarkers and the challenge of obtaining a universal cut-off value for biomarkers.
Collapse
Affiliation(s)
- Balaji Balakrishnan
- Department of Integrative Biology, School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore, India
| | | | - Aswin Anand Pai
- Department of Haematology, Christian Medical College, Vellore, India
| | | | | | - Vikram Mathews
- Department of Haematology, Christian Medical College, Vellore, India
| | - Biju George
- Department of Haematology, Christian Medical College, Vellore, India
| | | |
Collapse
|
20
|
Abstract
When discovered in the early 2000s, interleukin-33 (IL-33) was characterized as a potent driver of type 2 immunity and implicated in parasite clearance, as well as asthma, allergy, and lung fibrosis. Yet research in other models has since revealed that IL-33 is a highly pleiotropic molecule with diverse functions. These activities are supported by elusive release mechanisms and diverse expression of the IL-33 receptor, STimulation 2 (ST2), on both immune and stromal cells. Interestingly, IL-33 also supports type 1 immune responses during viral and tumor immunity and after allogeneic hematopoietic stem cell transplantation. Yet the IL-33-ST2 axis is also critical to the establishment of systemic homeostasis and tissue repair and regeneration. Despite these recent findings, the mechanisms by which IL-33 governs the balance between immunity and homeostasis or can support both effective repair and pathogenic fibrosis are poorly understood. As such, ongoing research is trying to understand the potential reparative and regulatory versus pro-inflammatory and pro-fibrotic roles for IL-33 in transplantation. This review provides an overview of the emerging regenerative role of IL-33 in organ homeostasis and tissue repair as it relates to transplantation immunology. It also outlines the known impacts of IL-33 in commonly transplanted solid organs and covers the envisioned roles for IL-33 in ischemia-reperfusion injury, rejection, and tolerance. Finally, we give a comprehensive summary of its effects on different cell populations involved in these processes, including ST2 + regulatory T cells, innate lymphoid cell type 2, as well as significant myeloid cell populations.
Collapse
|
21
|
Jin J, Wan Y, Shu Q, Liu J, Lai D. Knowledge mapping and research trends of IL-33 from 2004 to 2022: a bibliometric analysis. Front Immunol 2023; 14:1158323. [PMID: 37153553 PMCID: PMC10157155 DOI: 10.3389/fimmu.2023.1158323] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 03/30/2023] [Indexed: 05/09/2023] Open
Abstract
Background IL-33 has been studied widely but its comprehensive and systematic bibliometric analysis is yet available. The present study is to summarize the research progress of IL-33 through bibliometric analysis. Methods The publications related to IL-33 were identified and selected from the Web of Science Core Collection (WoSCC) database on 7 December 2022. The downloaded data was analyzed with bibliometric package in R software. CiteSpace and VOSviewer were used to conduct IL-33 bibliometric and knowledge mapping analysis. Results From 1 January 2004 to 7 December 2022, 4711 articles on IL-33 research published in 1009 academic journals by 24652 authors in 483 institutions from 89 countries were identified. The number of articles had grown steadily over this period. The United States of America(USA) and China are the major contributors in the field of research while University of Tokyo and University of Glasgow are the most active institutions. The most prolific journal is Frontiers in Immunology, while the Journal of Immunity is the top 1 co-cited journal. Andrew N. J. Mckenzie published the most significant number of articles and Jochen Schmitz was co-cited most. The major fields of these publications are immunology, cell biology, and biochemistry & molecular biology. After analysis, the high-frequency keywords of IL-33 research related to molecular biology (sST2, IL-1), immunological effects (type 2 immunity, Th2 cells), and diseases (asthma, cancer, cardiovascular diseases). Among these, the involvement of IL-33 in the regulation of type 2 inflammation has strong research potential and is a current research hotspot. Conclusion The present study quantifies and identifies the current research status and trends of IL-33 using bibliometric and knowledge mapping analysis. This study may offer the direction of IL-33-related research for scholars.
Collapse
Affiliation(s)
- Jingyi Jin
- Department of Neonatal Surgery, Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Yantong Wan
- Guangdong Provincial Key Laboratory of Proteomics, Department of Pathophysiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Qiang Shu
- Department of Thoracic and Cardiovascular Surgery, Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Jinghua Liu
- Guangdong Provincial Key Laboratory of Proteomics, Department of Pathophysiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
- *Correspondence: Dengming Lai, ; Jinghua Liu,
| | - Dengming Lai
- Department of Neonatal Surgery, Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
- *Correspondence: Dengming Lai, ; Jinghua Liu,
| |
Collapse
|
22
|
Guo H, Bossila EA, Ma X, Zhao C, Zhao Y. Dual Immune Regulatory Roles of Interleukin-33 in Pathological Conditions. Cells 2022; 11:cells11203237. [PMID: 36291105 PMCID: PMC9600220 DOI: 10.3390/cells11203237] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 10/07/2022] [Accepted: 10/11/2022] [Indexed: 11/20/2022] Open
Abstract
Interleukin-33 (IL-33), a member of the IL-1 cytokine family and a multifunctional cytokine, plays critical roles in maintaining host homeostasis and in pathological conditions, such as allergy, infectious diseases, and cancer, by acting on multiple types of immune cells and promoting type 1 and 2 immune responses. IL-33 is rapidly released by immune and non-immune cells upon stimulation by stress, acting as an “alarmin” by binding to its receptor, suppression of tumorigenicity 2 (ST2), to trigger downstream signaling pathways and activate inflammatory and immune responses. It has been recognized that IL-33 displays dual-functioning immune regulatory effects in many diseases and has both pro- and anti-tumorigenic effects, likely depending on its primary target cells, IL-33/sST2 expression levels, cellular context, and the cytokine microenvironment. Herein, we summarize our current understanding of the biological functions of IL-33 and its roles in the pathogenesis of various conditions, including inflammatory and autoimmune diseases, infections, cancers, and cases of organ transplantation. We emphasize the nature of context-dependent dual immune regulatory functions of IL-33 in many cells and diseases and review systemic studies to understand the distinct roles of IL-33 in different cells, which is essential to the development of more effective diagnoses and therapeutic approaches for IL-33-related diseases.
Collapse
Affiliation(s)
- Han Guo
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 101499, China
| | - Elhusseny A. Bossila
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 101499, China
- Biotechnology Department, Faculty of Agriculture Al-Azhar University, Cairo 11311, Egypt
| | - Xinran Ma
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 101499, China
| | - Chenxu Zhao
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 101499, China
| | - Yong Zhao
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 101499, China
- Beijing Institute for Stem Cell and Regeneration, Beijing 100101, China
- Correspondence: ; Tel.: +86-10-64807302; Fax: +86-10-64807313
| |
Collapse
|
23
|
Cramer M, Pineda Molina C, Hussey G, Turnquist HR, Badylak SF. Transcriptomic Regulation of Macrophages by Matrix-Bound Nanovesicle-Associated Interleukin-33. Tissue Eng Part A 2022; 28:867-878. [PMID: 35770892 PMCID: PMC9634988 DOI: 10.1089/ten.tea.2022.0006] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Accepted: 06/14/2022] [Indexed: 11/12/2022] Open
Abstract
The innate immune response, particularly the phenotype of responding macrophages, has significant clinical implications in the remodeling outcome following implantation of biomaterials and engineered tissues. In general, facilitation of an anti-inflammatory (M2-like) phenotype is associated with tissue repair and favorable outcomes, whereas pro-inflammatory (M1-like) activation can contribute to chronic inflammation and a classic foreign body response. Biologic scaffolds composed of extracellular matrix (ECM) and, more recently, matrix-bound nanovesicles (MBV) embedded within the ECM are known to direct macrophages toward an anti-inflammatory phenotype and stimulate a constructive remodeling outcome. The mechanisms of MBV-mediated macrophage activation are not fully understood, but interleukin-33 (IL-33) within the MBV appears critical for M2-like activation. Previous work has shown that IL-33 is encapsulated within the lumen of MBV and stimulates phenotypical changes in macrophages independent of its canonical surface receptor stimulation-2 (ST2). In the present study, we used next-generation RNA sequencing to determine the gene signature of macrophages following exposure to MBV with and without intraluminal IL-33. MBV-associated IL-33 instructed an anti-inflammatory phenotype in both wild-type and st2-/- macrophages by upregulating M2-like and downregulating M1-like genes. The repertoire of genes regulated by ST2-independent IL-33 signaling were broadly related to the inflammatory response and crosstalk between cells of both the innate and adaptive immune systems. These results signify the importance of the MBV intraluminal protein IL-33 in stimulating a pro-remodeling M2-like phenotype in macrophages and provides guidance for the designing of next-generation biomaterials and tissue engineering strategies. Impact statement The phenotype of responding macrophages is predictive of the downstream remodeling response to an implanted biomaterial. The clinical impact of macrophage phenotype has motivated studies to investigate the factors that regulate macrophage activation. Matrix-bound nanovesicles (MBV) embedded within the extracellular matrix direct macrophages toward an anti-inflammatory (M2)-like phenotype that is indicative of a favorable remodeling response. Although the mechanisms of MBV-mediated macrophage activation are not fully understood, the intraluminal protein interleukin-33 (IL-33) is clearly a contributing signaling molecule. The present study identifies those genes regulated by MBV-associated IL-33 that promote a pro-remodeling M2-like macrophage activation state and can guide future therapies in regenerative medicine.
Collapse
Affiliation(s)
- Madeline Cramer
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Catalina Pineda Molina
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - George Hussey
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Surgery and School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Heth R. Turnquist
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Surgery and School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Immunology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Thomas E. Starzl Transplantation Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Stephen F. Badylak
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Surgery and School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
24
|
Nagamura-Inoue T, Kato S, Najima Y, Isobe M, Doki N, Yamamoto H, Uchida N, Takahashi A, Hori A, Nojima M, Ohashi K, Nagamura F, Tojo A. Immunological influence of serum-free manufactured umbilical cord-derived mesenchymal stromal cells for steroid-resistant acute graft-versus-host disease. Int J Hematol 2022; 116:754-769. [PMID: 35908021 DOI: 10.1007/s12185-022-03408-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Accepted: 06/13/2022] [Indexed: 12/31/2022]
Abstract
This study investigated the safety, efficacy, and immunological influence of allogeneic umbilical cord-derived mesenchymal stromal cells (IMSUT-CORD) processed in serum-free medium and cryoprotectant, for treating steroid-resistant acute graft-versus-host disease (aGVHD). In a phase I dose-escalation trial, IMSUT-CORD were infused intravenously twice weekly over two cycles with up to two additional cycles. Four patients received a dose of 1 × 106 cells/kg, while three received 2 × 106/kg. Of 76 total adverse events, fourteen associated or possibly associated adverse events included 2 cases of a hot flash, headache, and peripheral neuropathy, 1 each of upper abdominal pain, hypoxia, increased γ-GTP, somnolence, peripheral vascular pain at the injection site, thrombocytopenia, hypertension, and decreased fibrinogen. At 16 weeks after the initial IMSUT-CORD infusion, three patients showed complete response (CR), two partial response (PR), one mixed response, and one no response. The overall response rate was 71.4%, and the continuous CR/PR rate was 100% for over 28 days after CR/PR. NK cell count significantly increased and correlated with treatment response, whereas IL-12, IL-17, and IL-33 levels decreased, but did not correlate with treatment response. CCL2 and CCL11 levels increased during IMSUT-CORD therapy. IMSUT-CORD are usable in patients with steroid-resistant aGVHD (UMIN000032819: https://www.umin.ac.jp/ctr ).
Collapse
Affiliation(s)
- Tokiko Nagamura-Inoue
- Department of Cell Processing and Transfusion, Research Hospital, The Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo, 108-8639, Japan. .,Department of Hematology and Oncology, Research Hospital, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan. .,IMSUT CORD, Research Hospital, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan. .,Department of Global Clinical Research, Graduate School of Medicine, Chiba University, Chiba, Japan.
| | - Seiko Kato
- Department of Hematology and Oncology, Research Hospital, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Yuho Najima
- Hematology Division, Tokyo Metropolitan Cancer and Infectious Diseases Center, Komagome Hospital, Tokyo, Japan
| | - Masamichi Isobe
- Department of Hematology and Oncology, Research Hospital, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Noriko Doki
- Hematology Division, Tokyo Metropolitan Cancer and Infectious Diseases Center, Komagome Hospital, Tokyo, Japan
| | | | - Naoyuki Uchida
- Department of Hematology, Toranomon Hospital, Tokyo, Japan
| | - Atsuko Takahashi
- Department of Cell Processing and Transfusion, Research Hospital, The Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo, 108-8639, Japan.,IMSUT CORD, Research Hospital, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Akiko Hori
- Department of Cell Processing and Transfusion, Research Hospital, The Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo, 108-8639, Japan.,IMSUT CORD, Research Hospital, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Masanori Nojima
- Division of Advanced Medicine Promotion, The Advanced Clinical Center, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Kazuteru Ohashi
- Hematology Division, Tokyo Metropolitan Cancer and Infectious Diseases Center, Komagome Hospital, Tokyo, Japan
| | - Fumitaka Nagamura
- Department of Global Clinical Research, Graduate School of Medicine, Chiba University, Chiba, Japan.,Division of Advanced Medicine Promotion, The Advanced Clinical Center, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Arinobu Tojo
- Department of Hematology and Oncology, Research Hospital, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan.,Institute of Innovation Advancement, Tokyo Medical and Dental University, Tokyo, Japan
| |
Collapse
|
25
|
Dwyer GK, Mathews LR, Villegas JA, Lucas A, Gonzalez de Peredo A, Blazar BR, Girard JP, Poholek AC, Luther SA, Shlomchik W, Turnquist HR. IL-33 acts as a costimulatory signal to generate alloreactive Th1 cells in graft-versus-host disease. J Clin Invest 2022; 132:e150927. [PMID: 35503257 PMCID: PMC9197517 DOI: 10.1172/jci150927] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 04/28/2022] [Indexed: 12/02/2022] Open
Abstract
Antigen-presenting cells (APCs) integrate signals emanating from local pathology and program appropriate T cell responses. In allogeneic hematopoietic stem cell transplantation (alloHCT), recipient conditioning releases damage-associated molecular patterns (DAMPs) that generate proinflammatory APCs that secrete IL-12, which is a driver of donor Th1 responses, causing graft-versus-host disease (GVHD). Nevertheless, other mechanisms exist to initiate alloreactive T cell responses, as recipients with disrupted DAMP signaling or lacking IL-12 develop GVHD. We established that tissue damage signals are perceived directly by donor CD4+ T cells and promoted T cell expansion and differentiation. Specifically, the fibroblastic reticular cell-derived DAMP IL-33 is increased by recipient conditioning and is critical for the initial activation, proliferation, and differentiation of alloreactive Th1 cells. IL-33 stimulation of CD4+ T cells was not required for lymphopenia-induced expansion, however. IL-33 promoted IL-12-independent expression of Tbet and generation of Th1 cells that infiltrated GVHD target tissues. Mechanistically, IL-33 augmented CD4+ T cell TCR-associated signaling pathways in response to alloantigen. This enhanced T cell expansion and Th1 polarization, but inhibited the expression of regulatory molecules such as IL-10 and Foxp3. These data establish an unappreciated role for IL-33 as a costimulatory signal for donor Th1 generation after alloHCT.
Collapse
Affiliation(s)
- Gaelen K. Dwyer
- Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Thomas E. Starzl Transplantation Institute and
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Lisa R. Mathews
- Thomas E. Starzl Transplantation Institute and
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - José A. Villegas
- Department of Biochemistry, University of Lausanne, Epalinges, Switzerland
| | - Anna Lucas
- Thomas E. Starzl Transplantation Institute and
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Anne Gonzalez de Peredo
- Institut de Pharmacologie et de Biologie Structurale, Université de Toulouse, Centre National de la Recherche Scientifique, Université Paul Sabatier, Toulouse, France
| | - Bruce R. Blazar
- Department of Pediatrics, University of Minnesota Medical School, Minneapolis, Minnesota, USA
| | - Jean-Philippe Girard
- Institut de Pharmacologie et de Biologie Structurale, Université de Toulouse, Centre National de la Recherche Scientifique, Université Paul Sabatier, Toulouse, France
| | - Amanda C. Poholek
- Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Division of Pediatric Rheumatology, and
| | - Sanjiv A. Luther
- Department of Biochemistry, University of Lausanne, Epalinges, Switzerland
| | - Warren Shlomchik
- Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Thomas E. Starzl Transplantation Institute and
- Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Hēth R. Turnquist
- Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Thomas E. Starzl Transplantation Institute and
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
26
|
Ferrara J, Prado-Acosta M. Graft-versus-host disease: establishing IL-33 as an important costimulatory molecule. J Clin Invest 2022; 132:160692. [PMID: 35703182 PMCID: PMC9197507 DOI: 10.1172/jci160692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Approximately half of patients with hematologic malignancy who are treated with allogeneic hematopoietic stem cell transplantation (alloHCT) experience graft-versus-host disease (GVHD), which has high mortality rates despite immunosuppressive therapy. IL-12 is known to drive donor T cells toward an inflammatory Th1 lineage in GVHD, but other mechanisms also promote pathological Th1 alloimmune responses. In this issue of the JCI, Dwyer et al. report on their use of transgenic mice and alloHCT models of GVHD to demonstrate that IL-33 acts directly on donor T cells to increase Tbet expression independently of IL-12. Notably, IL-33 amplified T cell receptor–signaling pathways and inhibited production of regulatory molecules. These findings firmly establish IL-33 as an important costimulatory molecule for Th1 cells during GVHD and provide a target for reducing GVHD, especially in the gastrointestinal (GI) tract, where damage drives mortality.
Collapse
|
27
|
Piper C, Hainstock E, Yin-Yuan C, Chen Y, Khatun A, Kasmani MY, Evans J, Miller JA, Gorski J, Cui W, Drobyski WR. Single-cell immune profiling reveals a developmentally distinct CD4+ GM-CSF+ T-cell lineage that induces GI tract GVHD. Blood Adv 2022; 6:2791-2804. [PMID: 35015822 PMCID: PMC9092418 DOI: 10.1182/bloodadvances.2021006084] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Accepted: 11/29/2021] [Indexed: 11/20/2022] Open
Abstract
Gastrointestinal (GI) tract involvement is a major determinant for subsequent morbidity and mortality arising during graft-versus-host disease (GVHD). CD4+ T cells that produce granulocyte-macrophage colony stimulating factor (GM-CSF) have emerged as central mediators of inflammation in this tissue site as GM-CSF serves as a critical cytokine link between the adaptive and innate arms of the immune system. However, cellular heterogeneity within the CD4+ GM-CSF+ T-cell population due to the concurrent production of other inflammatory cytokines has raised questions as to whether these cells have a common ontology or if a unique CD4+ GM-CSF+ subset exists that differs from other defined T helper subtypes. Using single-cell RNA sequencing analysis (scRNAseq), we identified two CD4+ GM-CSF+ T-cell populations that arose during GVHD and were distinguishable according to the presence or absence of interferon-γ (IFN-γ) coexpression. CD4+ GM-CSF+ IFN-γ- T cells, which emerged preferentially in the colon, had a distinct transcriptional profile, used unique gene regulatory networks, and possessed a nonoverlapping T-cell receptor repertoire compared with CD4+ GM-CSF+ IFN-γ+ T cells as well as all other transcriptionally defined CD4+ T-cell populations in the colon. Functionally, this CD4+ GM-CSF+ T-cell population contributed to pathologic damage in the GI tract that was critically dependent on signaling through the interleukin-17 (IL-7) receptor but was independent of type 1 interferon signaling. Thus, these studies help to unravel heterogeneity within CD4+ GM-CSF+ T cells that arise during GVHD and define a developmentally distinct colitogenic T helper subtype GM-CSF+ subset that mediates immunopathology.
Collapse
Affiliation(s)
- Clint Piper
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI
| | - Emma Hainstock
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI
| | - Cheng Yin-Yuan
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI
| | - Yao Chen
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI
- Blood Research Institute, Versiti, Milwaukee, WI; and
| | - Achia Khatun
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI
- Blood Research Institute, Versiti, Milwaukee, WI; and
| | - Moujtaba Y. Kasmani
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI
- Blood Research Institute, Versiti, Milwaukee, WI; and
| | | | | | - Jack Gorski
- Blood Research Institute, Versiti, Milwaukee, WI; and
| | - Weiguo Cui
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI
- Blood Research Institute, Versiti, Milwaukee, WI; and
| | - William R. Drobyski
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI
| |
Collapse
|
28
|
Abstract
Our understanding of the functions of the IL-1 superfamily cytokine and damage-associated molecular pattern IL-33 continues to evolve with our understanding of homeostasis and immunity. The early findings that IL-33 is a potent driver of type 2 immune responses promoting parasite expulsion, but also inflammatory diseases like allergy and asthma, have been further supported. Yet, as the importance of a type 2 response in tissue repair and homeostasis has emerged, so has the fundamental importance of IL-33 to these processes. In this review, we outline an evolving understanding of IL-33 immunobiology, paying particular attention to how IL-33 directs a network of ST2+ regulatory T cells, reparative and regulatory macrophages, and type 2 innate lymphoid cells that are fundamental to tissue development, homeostasis, and repair. Expected final online publication date for the Annual Review of Immunology, Volume 40 is April 2022. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Gaelen K. Dwyer
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Thomas E. Starzl Transplantation Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Louise M. D'Cruz
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Hēth R. Turnquist
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Thomas E. Starzl Transplantation Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
29
|
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: 3.3] [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
|
30
|
Makris S, de Winde CM, Horsnell HL, Cantoral-Rebordinos JA, Finlay RE, Acton SE. Immune function and dysfunction are determined by lymphoid tissue efficacy. Dis Model Mech 2022; 15:dmm049256. [PMID: 35072206 PMCID: PMC8807573 DOI: 10.1242/dmm.049256] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Lymphoid tissue returns to a steady state once each immune response is resolved, and although this occurs multiple times throughout life, its structural integrity and functionality remain unaffected. Stromal cells orchestrate cellular interactions within lymphoid tissue, and any changes to the microenvironment can have detrimental outcomes and drive disease. A breakdown in lymphoid tissue homeostasis can lead to a loss of tissue structure and function that can cause aberrant immune responses. This Review highlights recent advances in our understanding of lymphoid tissue function and remodelling in adaptive immunity and in disease states. We discuss the functional role of lymphoid tissue in disease progression and explore the changes to lymphoid tissue structure and function driven by infection, chronic inflammatory conditions and cancer. Understanding the role of lymphoid tissues in immune responses to a wide range of pathologies allows us to take a fuller systemic view of disease progression.
Collapse
Affiliation(s)
- Spyridon Makris
- Stromal Immunology Group, MRC Laboratory for Molecular Cell Biology, University College London, Gower Street, London WC1E 6BT, UK
| | - Charlotte M. de Winde
- Department for Molecular Cell Biology and Immunology, Amsterdam UMC, location VUmc, De Boelelaan 1108, 1081 HZ Amsterdam, Netherlands
| | - Harry L. Horsnell
- Stromal Immunology Group, MRC Laboratory for Molecular Cell Biology, University College London, Gower Street, London WC1E 6BT, UK
| | - Jesús A. Cantoral-Rebordinos
- Stromal Immunology Group, MRC Laboratory for Molecular Cell Biology, University College London, Gower Street, London WC1E 6BT, UK
| | - Rachel E. Finlay
- Division of Immunology, Immunity to Infection and Respiratory Medicine, School of Biological Sciences, The University of Manchester, Manchester M13 9PL, UK
| | - Sophie E. Acton
- Stromal Immunology Group, MRC Laboratory for Molecular Cell Biology, University College London, Gower Street, London WC1E 6BT, UK
| |
Collapse
|
31
|
Single Nucleotide Polymorphisms of IL-33 Gene Correlated with Renal Allograft Fibrosis in Kidney Transplant Recipients. J Immunol Res 2021; 2021:8029180. [PMID: 34950738 PMCID: PMC8689233 DOI: 10.1155/2021/8029180] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Accepted: 11/08/2021] [Indexed: 11/17/2022] Open
Abstract
Background Nowadays, renal allograft survival is confined by the development of allograft fibrosis. Previous studies have reported interleukin-33 (IL-33) upregulated significantly in patients with chronic renal allograft dysfunction, and it could induce renal tubular epithelial to mesenchymal transition (EMT), which eventually contributed to renal allograft fibrosis. Our study intended to detect the underlying association between single nucleotide polymorphisms (SNPs) of IL-33 gene and renal allograft fibrosis in kidney transplant recipients. Methods We collected blood samples from 200 renal transplant recipients for the identification of SNPs and transplanted kidney tissue samples for identifying differentially expressed genes (DEGs). Intersection of SNP-related genes and DEGs was conducted for further analysis. Relationships between these SNPs and renal allograft fibrosis were evaluated by the inheritance models. Immunohistochemical (IHC) staining and western blotting (WB) were used to detect the expression of IL-33 and the markers of EMT in human kidney tissues obtained from control and chronic renal allograft dysfunction (CAD) patients. In vitro, we detected the progressions of EMT-related markers and the levels of MAPK signaling pathway mediators after transfecting IL-33 mutant plasmids in HK2 cells. Results Three intersected genes including IL-33 genes were significantly expressed. IL-33 expression was validated in kidney tissues by IHC and WB. Thirty-nine IL-33-related SNPs were identified in targeted sequencing, in which 26 tagger SNPs were found by linkage disequilibrium analysis for further analysis. General linear models indicated sirolimus administration significantly influenced renal allograft fibrosis (P < 0.05), adjustment of which was conducted in the following analysis. By multiple inheritance model analyses, SNP rs10975519 of IL-33 gene was found closely related to renal allograft fibrosis (P < 0.005). Furthermore, HK2 cells transfected with mutated plasmid of rs10975519 showed stronger mobility and migration ability. Moreover, IL-33 mutant plasmids could promote the IL-33-induced EMT through the sustained activation of p38 MAPK signaling pathway in HK2 cells. Conclusion In our study, rs10975519 on the IL-33 gene was found to be statistically associated with the development of renal allograft fibrosis in kidney transplant recipients. This process may be related to the IL-33-induced EMT and sustained activation of p38 MAPK signaling pathway.
Collapse
|
32
|
Abstract
PURPOSE OF REVIEW Graft-versus-host disease (GVHD) following hematopoietic cell transplant (HCT) has a significant impact on morbidity and mortality among recipients. Predicting the long-term outcomes at the time of diagnosis of GVHD or even after response to up-front therapy can be challenging and only has modest accuracy. With biomarkers available to help guide decision-making, the landscape of GVHD is evolving. RECENT FINDINGS Several acute GVHD biomarkers have been identified, with some better able to categorize patients based on their GVHD severity and potential for refractory disease than standard clinical staging or response criteria. SUMMARY Biomarkers are now being incorporated into the clinical trial design for both high and low-risk GVHD. These findings will likely impact how clinical care is delivered in the future as improved risk stratification has the potential to improve outcomes by providing individualized treatment plans for affected patients.
Collapse
Affiliation(s)
- Saara Kaviany
- Monroe Carell Jr Children's Hospital, Vanderbilt University Medical Center, Department of Pediatrics, Division of Hematology/Oncology, Nashville, Tennessee, USA
| | | |
Collapse
|
33
|
Coron E, Esnaud E, Chevallier P, Bessard A, Perez Cuadrado‐Robles E, David G, Bossard C, Brégéon J, Jarry A, Neunlist M, Quénéhervé L. Early remodeling of the colonic mucosa after allogeneic hematopoietic stem cells transplantation: An open-label controlled pilot study on 19 patients. United European Gastroenterol J 2021; 9:955-963. [PMID: 34431618 PMCID: PMC8498402 DOI: 10.1002/ueg2.12128] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Accepted: 02/12/2020] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Graft-versus-host disease (GVHD), particularly acute digestive GVHD (aDGVHD), is a severe complication of allogeneic hematopoietic stem cell transplantation (allo-HSCT). It is necessary to identify predictive factors of GVHD to adapt prophylactic treatment. OBJECTIVE In this context, our pilot study aimed (i) to determine whether an early remodeling of the colonic mucosa occurred after allo-HSCT and (ii) to identify potential predictive mucosal markers of aDGVHD after allo-HSCT. METHODS Between day 21 and day 28 after the allo-HSCT, 19 allo-HSCT patients were included and had a rectosigmoidoscopy with probe-based confocal laser endomicroscopy (pCLE) recording and biopsies. Sixteen patients were included in the control group. Morphological (pCLE), functional (intestinal permeability), and inflammatory parameters (cytokine multiplex immunoassay) were assessed. RESULTS Among allo-HSCT patients, 11 patients developed GVHD, and 6 of them developed aDGVHD. Morphological and functional changes of the colonic mucosa occurred after allo-HSCT. Indeed, the perimeter of colonic crypts was significantly increased in allo-HSCT patients compared to controls as well as crypt lumen fluorescein leakage (53% vs. 9%), whereas crypts sphericity, roundness, Feret diameter, and mean vessel area were significantly decreased in allo-HSCT patients compared to the control group. In addition, interleukin-6 (IL-6), IL-33, and IL-15 levels in the supernatants of 24 h explant cultures of colonic biopsies were significantly increased in allo-HSCT patients compared to controls. Finally, there was no difference in pCLE parameters, intestinal permeability, and inflammatory cytokines between patients who developed aDGVHD and those who did not. CONCLUSION This pilot study identified early colonic mucosa remodeling after allo-HSCT conditioning therapy, that is morphological and functional mucosal alterations as well as mucosal inflammation. As to whether these changes are first steps in GVHD initiation and could be considered as predictive biomarkers of aDGVHD need to be determined in a larger cohort of patients.
Collapse
Affiliation(s)
- Emmanuel Coron
- Université de NantesINSERMThe Enteric Nervous System in Gut and Brain DisordersIMADNantesFrance
- Institut des Maladies de l'Appareil DigestifIMADCHU NantesHôpital Hôtel‐DieuNantesFrance
| | - Elise Esnaud
- Institut des Maladies de l'Appareil DigestifIMADCHU NantesHôpital Hôtel‐DieuNantesFrance
| | | | - Anne Bessard
- Université de NantesINSERMThe Enteric Nervous System in Gut and Brain DisordersIMADNantesFrance
| | | | - Grégoire David
- Université de NantesINSERMThe Enteric Nervous System in Gut and Brain DisordersIMADNantesFrance
| | - Céline Bossard
- Service d'Anatomie et Cytologie PathologiqueUniversité de NantesCHU NantesInsermCRCINANantesFrance
| | - Jérémy Brégéon
- Université de NantesINSERMThe Enteric Nervous System in Gut and Brain DisordersIMADNantesFrance
| | - Anne Jarry
- Université de NantesInsermCRCINANantesFrance
| | - Michel Neunlist
- Université de NantesINSERMThe Enteric Nervous System in Gut and Brain DisordersIMADNantesFrance
- Institut des Maladies de l'Appareil DigestifIMADCHU NantesHôpital Hôtel‐DieuNantesFrance
| | - Lucille Quénéhervé
- Université de NantesINSERMThe Enteric Nervous System in Gut and Brain DisordersIMADNantesFrance
- Institut des Maladies de l'Appareil DigestifIMADCHU NantesHôpital Hôtel‐DieuNantesFrance
| |
Collapse
|
34
|
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: 17] [Impact Index Per Article: 4.3] [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
|
35
|
Maas-Bauer K, Lohmeyer JK, Hirai T, Ramos TL, Fazal FM, Litzenburger UM, Yost KE, Ribado JV, Kambham N, Wenokur AS, Lin PY, Alvarez M, Mavers M, Baker J, Bhatt AS, Chang HY, Simonetta F, Negrin RS. Invariant natural killer T-cell subsets have diverse graft-versus-host-disease-preventing and antitumor effects. Blood 2021; 138:858-870. [PMID: 34036317 PMCID: PMC8432044 DOI: 10.1182/blood.2021010887] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Accepted: 04/22/2021] [Indexed: 11/20/2022] Open
Abstract
Invariant natural killer T (iNKT) cells are a T-cell subset with potent immunomodulatory properties. Experimental evidence in mice and observational studies in humans indicate that iNKT cells have antitumor potential as well as the ability to suppress acute and chronic graft-versus-host-disease (GVHD). Murine iNKT cells differentiate during thymic development into iNKT1, iNKT2, and iNKT17 sublineages, which differ transcriptomically and epigenomically and have subset-specific developmental requirements. Whether distinct iNKT sublineages also differ in their antitumor effect and their ability to suppress GVHD is currently unknown. In this work, we generated highly purified murine iNKT sublineages, characterized their transcriptomic and epigenomic landscape, and assessed specific functions. We show that iNKT2 and iNKT17, but not iNKT1, cells efficiently suppress T-cell activation in vitro and mitigate murine acute GVHD in vivo. Conversely, we show that iNKT1 cells display the highest antitumor activity against murine B-cell lymphoma cells both in vitro and in vivo. Thus, we report for the first time that iNKT sublineages have distinct and different functions, with iNKT1 cells having the highest antitumor activity and iNKT2 and iNKT17 cells having immune-regulatory properties. These results have important implications for the translation of iNKT cell therapies to the clinic for cancer immunotherapy as well as for the prevention and treatment of GVHD.
Collapse
Affiliation(s)
- Kristina Maas-Bauer
- Division of Blood and Marrow Transplantation, Stanford University, Stanford, CA
- Department of Hematology, Oncology, and Stem Cell Transplantation, University of Freiburg Medical Center, Freiburg, Germany
| | - Juliane K Lohmeyer
- Division of Blood and Marrow Transplantation, Stanford University, Stanford, CA
| | - Toshihito Hirai
- Division of Blood and Marrow Transplantation, Stanford University, Stanford, CA
| | - Teresa Lopes Ramos
- Division of Blood and Marrow Transplantation, Stanford University, Stanford, CA
| | | | | | | | | | | | - Arielle S Wenokur
- Division of Blood and Marrow Transplantation, Stanford University, Stanford, CA
| | - Po-Yu Lin
- Division of Blood and Marrow Transplantation, Stanford University, Stanford, CA
| | - Maite Alvarez
- Division of Blood and Marrow Transplantation, Stanford University, Stanford, CA
| | - Melissa Mavers
- Division of Blood and Marrow Transplantation, Stanford University, Stanford, CA
- Division of Stem Cell Transplantation and Regenerative Medicine, Bass Center for Childhood Cancer and Blood Diseases, Department of Pediatrics, Stanford University School of Medicine, Palo Alto, CA
| | - Jeanette Baker
- Division of Blood and Marrow Transplantation, Stanford University, Stanford, CA
| | - Ami S Bhatt
- Division of Blood and Marrow Transplantation, Stanford University, Stanford, CA
- Department of Genetics, and
- Division of Hematology and
| | - Howard Y Chang
- Center for Personal Dynamic Regulomes
- Howard Hughes Medical Institute, Stanford University, Stanford, CA
| | - Federico Simonetta
- Division of Blood and Marrow Transplantation, Stanford University, Stanford, CA
- Division of Hematology, Department of Oncology, Geneva University Hospitals, Geneva, Switzerland; and
- Translational Research Center for Oncohematology, Department of Internal Medicine Specialties, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Robert S Negrin
- Division of Blood and Marrow Transplantation, Stanford University, Stanford, CA
| |
Collapse
|
36
|
Zewde MG, Morales G, Gandhi I, Özbek U, Aguayo-Hiraldo P, Ayuk F, Baez J, Chanswangphuwana C, Choe H, DeFilipp Z, Etra A, Grupp S, Hexner EO, Hogan W, Javorniczky NR, Kasikis S, Kitko CL, Kowalyk S, Meedt E, Merli P, Nakamura R, Qayed M, Reshef R, Rösler W, Schechter T, Weber D, Wölfl M, Yanik G, Young R, Levine JE, Ferrara JLM, Chen YB. Evaluation of Elafin as a Prognostic Biomarker in Acute Graft-versus-Host Disease. Transplant Cell Ther 2021; 27:988.e1-988.e7. [PMID: 34474163 DOI: 10.1016/j.jtct.2021.08.021] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Revised: 08/23/2021] [Accepted: 08/24/2021] [Indexed: 11/17/2022]
Abstract
Acute graft-versus-host disease (GVHD) is a major cause of mortality in patients undergoing hematopoietic cell transplantation (HCT) for hematologic malignancies. The skin is the most commonly involved organ in GVHD. Elafin, a protease inhibitor overexpressed in inflamed epidermis, was previously identified as a diagnostic biomarker of skin GVHD; however, this finding was restricted to a subset of patients with isolated skin GVHD. The main driver of nonrelapse mortality (NRM) in HCT recipients is gastrointestinal (GI) GVHD. Two biomarkers, Regenerating islet-derived 3a (REG3α) and Suppressor of tumorigenesis 2 (ST2), have been validated as biomarkers of GI GVHD that predict long-term outcomes in patients treated for GVHD. We undertook this study to determine the utility of elafin as a prognostic biomarker in the general population of acute GVHD patients in whom GVHD may develop in multiple organs. We analyzed serum elafin concentrations as a predictive biomarker of acute GVHD outcomes and compared it with ST2 and REG3α in a large group of patients treated at multiple centers. A total of 526 patients from the Mount Sinai Acute GVHD International Consortium (MAGIC) who had received corticosteroid treatment for skin GVHD and who had not been previously studied were analyzed. Serum concentrations of elafin, ST2, and REG3α were measured by ELISA in all patients. The patients were divided at random into equal training and validation sets, and a competing-risk regression model was developed to model 6-month NRM using elafin concentration in the training set. Additional models were developed using concentrations of ST2 and REG3α or the combination of all 3 biomarkers as predictors. Receiver operating characteristic (ROC) curves were constructed using the validation set to evaluate the predictive accuracy of each model and to stratify patients into high- and low-risk biomarker groups. The cumulative incidence of 6-month NRM, overall survival (OS), and 4-week treatment response were compared between the risk groups. Unexpectedly, patients in the low-risk elafin group demonstrated a higher incidence of 6-month NRM, although the difference was not statistically significant (17% versus 11%; P = .19). OS at 6 months (68% versus 68%; P > .99) and 4-week response (78% versus 78%; P = .98) were similar in the low-risk and high-risk elafin groups. The area under the ROC curve (AUC) was 0.55 for elafin and 0.75 for the combination of ST2 and REG3α. The addition of elafin to the other 2 biomarkers did not improve the AUC. Our data indicate that serum elafin concentrations measured at the initiation of systemic treatment for acute GVHD did not predict 6-month NRM, OS, or treatment response in a multicenter population of patients treated systemically for acute GVHD. As seen in previous studies, serum concentrations of the GI GVHD biomarkers ST2 and REG3α were significant predictors of NRM, and the addition of elafin levels did not improve their accuracy. These results underscore the importance of GI disease in driving NRM in patients who develop acute GVHD.
Collapse
Affiliation(s)
- Makda Getachew Zewde
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - George Morales
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Isha Gandhi
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Umut Özbek
- Biostatistics Shared Resource Facility, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Paibel Aguayo-Hiraldo
- Children's Center for Cancer and Blood Diseases, Blood and Marrow Transplantation Section, Children's Hospital Los Angeles, Los Angeles, California
| | - Francis Ayuk
- Department of Stem Cell Transplantation, University Medical Center, Hamburg-Eppendorf, Germany
| | - Janna Baez
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | | | - Hannah Choe
- Blood and Marrow Transplantation Program, Ohio State University, Columbus, Ohio
| | - Zachariah DeFilipp
- Hematopoietic Cell Transplant and Cellular Therapy Program, Massachusetts General Hospital, Boston, Massachusetts
| | - Aaron Etra
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Stephan Grupp
- Division of Oncology, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Elizabeth O Hexner
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania
| | - William Hogan
- Blood and Marrow Transplantation Program, Mayo Clinic, Rochester, Minnesota
| | - Nora Rebeka Javorniczky
- Department of Medicine I, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
| | - Stelios Kasikis
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Carrie L Kitko
- Pediatric Blood and Marrow Transplantation Program, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Steven Kowalyk
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Elisabeth Meedt
- Blood and Marrow Transplantation Program, University of Regensburg, Regensburg, Germany
| | - Pietro Merli
- Department of Pediatric Hematology and Oncology, Bambino Gesù Children's Hospital, Rome, Italy
| | - Ryotaro Nakamura
- Hematology and Hematopoietic Cell Transplantation, City of Hope Medical Center, Duarte, California
| | - Muna Qayed
- Pediatric Blood and Marrow Transplantation Program, Aflac Cancer and Blood Disorders Center, Emory University and Children's Healthcare of Atlanta, Atlanta
| | - Ran Reshef
- Blood and Marrow Transplantation Program, Columbia University, New York, New York
| | - Wolf Rösler
- Department of Internal Medicine 5, Hematology/Oncology, University Hospital Erlangen, Erlangen, Germany
| | - Tal Schechter
- Division of Hematology/Oncology, Department of Pediatrics, University of Toronto, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Daniela Weber
- Blood and Marrow Transplantation Program, University of Regensburg, Regensburg, Germany
| | - Matthias Wölfl
- Pediatric Blood and Marrow Transplantation Program, Children's Hospital, University of Würzburg, Würzburg, Germany
| | - Gregory Yanik
- Blood and Marrow Transplantation Program, University of Michigan, Ann Arbor, Michigan
| | - Rachel Young
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - John E Levine
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - James L M Ferrara
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York.
| | - Yi-Bin Chen
- Hematopoietic Cell Transplant and Cellular Therapy Program, Massachusetts General Hospital, Boston, Massachusetts
| |
Collapse
|
37
|
Liang Y, Wang X, Wang H, Yang W, Yi P, Soong L, Cong Y, Cai J, Fan X, Sun J. IL-33 activates mTORC1 and modulates glycolytic metabolism in CD8 + T cells. Immunology 2021; 165:61-73. [PMID: 34411293 DOI: 10.1111/imm.13404] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 07/26/2021] [Accepted: 08/12/2021] [Indexed: 12/15/2022] Open
Abstract
Interleukin (IL)-33, a member in the IL-1 family, plays a central role in innate and adaptive immunity; however, how IL-33 mediates cytotoxic T-cell regulation and the downstream signals remain elusive. In this study, we found increased mouse IL-33 expression in CD8+ T cells following cell activation via anti-CD3/CD28 stimulation in vitro or lymphocytic choriomeningitis virus (LCMV) infection in vivo. Our cell adoptive transfer experiment demonstrated that extracellular, but not nuclear, IL-33 contributed to the activation and proliferation of CD8+ , but not CD4+ T effector cells in LCMV infection. Importantly, IL-33 induced mTORC1 activation in CD8+ T cells as evidenced by increased phosphorylated S6 ribosomal protein (p-S6) levels both in vitro and in vivo. Meanwhile, this IL-33-induced CD8+ T-cell activation was suppressed by mTORC1 inhibitors. Furthermore, IL-33 elevated glucose uptake and lactate production in CD8+ T cells in both dose- and time-dependent manners. The results of glycolytic rate assay demonstrated the increased glycolytic capacity of IL-33-treated CD8+ T cells compared with that of control cells. Our mechanistic study further revealed the capacity of IL-33 in promoting the expression of glucose transporter 1 (Glut1) and glycolytic enzymes via mTORC1, leading to accelerated aerobic glucose metabolism Warburg effect and increased effector T-cell activation. Together, our data provide new insights into IL-33-mediated regulation of CD8+ T cells, which might be beneficial for therapeutic strategies of inflammatory and infectious diseases in the future.
Collapse
Affiliation(s)
- Yuejin Liang
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA.,Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX, USA
| | - Xiaofang Wang
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA.,Department of Infectious Diseases, Key Laboratory of Viral Hepatitis of Hunan, Xiangya Hospital, Central South University, Changsha, China
| | - Hui Wang
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, USA
| | - Wenjing Yang
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA
| | - Panpan Yi
- Department of Infectious Diseases, Key Laboratory of Viral Hepatitis of Hunan, Xiangya Hospital, Central South University, Changsha, China
| | - Lynn Soong
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA.,Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX, USA.,Department of Pathology, University of Texas Medical Branch, Galveston, TX, USA
| | - Yingzi Cong
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA.,Department of Pathology, University of Texas Medical Branch, Galveston, TX, USA
| | - Jiyang Cai
- Department of Ophthalmology, University of Texas Medical Branch, Galveston, TX, USA
| | - Xuegong Fan
- Department of Infectious Diseases, Key Laboratory of Viral Hepatitis of Hunan, Xiangya Hospital, Central South University, Changsha, China
| | - Jiaren Sun
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA.,Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX, USA.,Department of Pathology, University of Texas Medical Branch, Galveston, TX, USA
| |
Collapse
|
38
|
Wang D, Liu Y, Lai X, Chen J, Cheng Q, Ma X, Lin Z, Wu D, Xu Y. Efficiency and Toxicity of Ruxolitinib as a Salvage Treatment for Steroid-Refractory Chronic Graft-Versus-Host Disease. Front Immunol 2021; 12:673636. [PMID: 34276662 PMCID: PMC8278571 DOI: 10.3389/fimmu.2021.673636] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Accepted: 06/21/2021] [Indexed: 11/13/2022] Open
Abstract
Graft-versus-host disease (GVHD), especially steroid-refractory GVHD, remains a life-threatening complication after hematopoietic stem cell transplantation (HSCT). The effect of the JAK1/2 kinase inhibitor ruxolitinib on treating steroid-refractory acute GVHD has been verified by the REACH1/2 study; however, its safety and efficacy in patients with steroid-refractory chronic GVHD (SR-cGVHD) remain unclear. In this retrospective study, 70 patients received ruxolitinib as a salvage therapy for SR-cGVHD. Twenty-four weeks after ruxolitinib treatment, the overall response rate (ORR) was 74.3% (52/70), including 34 patients who achieved complete remission (CR) and 18 who achieved partial remission (PR). The main adverse event was cytopenia, which occurred in 51.4% (36/70) of patients. After ruxolitinib treatment, the percentage of CD4 cells increased from 18.20% to 23.22% (P<0.001), while the percentages of NK (CD16+CD56+) cells and regulatory T cells (CD4+CD127 ± CD25+) decreased (P<0.001, P<0.001). Among the B cell subsets, the proportion of total B cells approximately tripled from 3.69% to 11.16% (P<0.001). Moreover, we observed a significant increase in IL-10 levels after ruxolitinib treatment (P=0.025) and a remarkable decrease in levels of suppression of tumorigenicity 2 (ST2) from 229.90 ng/ml to 72.65 ng/ml. The median follow-up after the initiation of ruxolitinib treatment was 401 (6-1076) days. The estimated one-year overall survival rate of the whole group was 66.0% (54.4-77.6%, 95% CI), and the one-year overall survival rate of patients with mild and moderate cGVHD was 69.6% (57.4-81.8%, 95% CI), which was better than that of patients with severe cGVHD (31.3%, 0.0-66.2%, 95% CI) (P=0.002). Patients who achieved a CR and PR achieved better survival outcomes (84.5%, 73.9-95.1%, 95% CI) than those who showed NR to ruxolitinib treatments (16.7%, 0-34.3%, 95% CI) (P<0.001). At the final follow-up, cGVHD relapse occurred in six patients after they reduced or continued their ruxolitinib doses. Collectively, our results suggest that ruxolitinib is potentially a safe and effective treatment for SR-cGVHD.
Collapse
Affiliation(s)
- Dong Wang
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, Key Laboratory of Thrombosis and Hemostasis of Ministry of Health, The First Affiliated Hospital of Soochow University, Suzhou, China.,Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Yin Liu
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, Key Laboratory of Thrombosis and Hemostasis of Ministry of Health, The First Affiliated Hospital of Soochow University, Suzhou, China.,Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Xiaoxuan Lai
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, Key Laboratory of Thrombosis and Hemostasis of Ministry of Health, The First Affiliated Hospital of Soochow University, Suzhou, China.,Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Jia Chen
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, Key Laboratory of Thrombosis and Hemostasis of Ministry of Health, The First Affiliated Hospital of Soochow University, Suzhou, China.,Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Qiao Cheng
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, Key Laboratory of Thrombosis and Hemostasis of Ministry of Health, The First Affiliated Hospital of Soochow University, Suzhou, China.,Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Xiao Ma
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, Key Laboratory of Thrombosis and Hemostasis of Ministry of Health, The First Affiliated Hospital of Soochow University, Suzhou, China.,Department of Hematology, Soochow Hopes Hematonosis Hospital, Suzhou, China
| | - Zhihong Lin
- Soochow Yongding Hospital, Department of Affiliated Renji Hospital of Shanghai Jiao Tong University of Medicine, Suzhou, China
| | - Depei Wu
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, Key Laboratory of Thrombosis and Hemostasis of Ministry of Health, The First Affiliated Hospital of Soochow University, Suzhou, China.,Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Yang Xu
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, Key Laboratory of Thrombosis and Hemostasis of Ministry of Health, The First Affiliated Hospital of Soochow University, Suzhou, China.,Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| |
Collapse
|
39
|
The Janus Face of IL-33 Signaling in Tumor Development and Immune Escape. Cancers (Basel) 2021; 13:cancers13133281. [PMID: 34209038 PMCID: PMC8268428 DOI: 10.3390/cancers13133281] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 06/06/2021] [Accepted: 06/25/2021] [Indexed: 12/15/2022] Open
Abstract
Simple Summary Interleukin-33 (IL-33) is often released from damaged cells, acting as a danger signal. IL-33 exerts its function by interacting with its receptor suppression of tumorigenicity 2 (ST2) that is constitutively expressed on most immune cells. Therefore, IL-33/ST2 signaling can modulate immune responses to participate actively in a variety of pathological conditions, such as cancer. Like a two-faced Janus, which faces opposite directions, IL-33/ST2 signaling may play contradictory roles on its impact on cancer progression through both immune and nonimmune cellular components. Accumulating evidence demonstrates both pro- and anti-tumorigenic properties of IL-33, depending on the complex nature of different tumor immune microenvironments. We summarize and discuss the most recent studies on the contradictory effects of IL-33 on cancer progression and treatment, with a goal to better understanding the various ways for IL-33 as a therapeutic target. Abstract Interleukin-33 (IL-33), a member of the IL-1 cytokine family, plays a critical role in maintaining tissue homeostasis as well as pathological conditions, such as allergy, infectious disease, and cancer, by promoting type 1 and 2 immune responses. Through its specific receptor ST2, IL-33 exerts multifaceted functions through the activation of diverse intracellular signaling pathways. ST2 is expressed in different types of immune cells, including Th2 cells, Th1 cells, CD8+ T cells, regulatory T cells (Treg), cytotoxic NK cells, group 2 innate lymphoid cells (ILC2s), and myeloid cells. During cancer initiation and progression, the aberrant regulation of the IL-33/ST2 axis in the tumor microenvironment (TME) extrinsically and intrinsically mediates immune editing via modulation of both innate and adaptive immune cell components. The summarized results in this review suggest that IL-33 exerts dual-functioning, pro- as well as anti-tumorigenic effects depending on the tumor type, expression levels, cellular context, and cytokine milieu. A better understanding of the distinct roles of IL-33 in epithelial, stromal, and immune cell compartments will benefit the development of a targeting strategy for this IL-33/ST2 axis for cancer immunotherapy.
Collapse
|
40
|
Khuat LT, Le CT, Pai CCS, Shields-Cutler RR, Holtan SG, Rashidi A, Parker SL, Knights D, Luna JI, Dunai C, Wang Z, Sturgill IR, Stoffel KM, Merleev AA, More SK, Maverakis E, Raybould HE, Chen M, Canter RJ, Monjazeb AM, Dave M, Ferrara JLM, Levine JE, Longo DL, Abedi M, Blazar BR, Murphy WJ. Obesity induces gut microbiota alterations and augments acute graft-versus-host disease after allogeneic stem cell transplantation. Sci Transl Med 2021; 12:12/571/eaay7713. [PMID: 33239390 DOI: 10.1126/scitranslmed.aay7713] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 01/22/2020] [Accepted: 06/02/2020] [Indexed: 12/22/2022]
Abstract
The efficacy of allogeneic hematopoietic stem cell transplantation (allo-HSCT) is limited by acute and chronic graft-versus-host disease (GVHD). The impact of obesity on allo-HSCT outcomes is poorly understood. Here, we report that obesity had a negative and selective impact on acute gut GVHD after allo-HSCT in mice with diet-induced obesity (DIO). These animals exhibited increased gut permeability, endotoxin translocation across the gut, and radiation-induced gastrointestinal damage after allo-HSCT. After allo-HSCT, both male and female DIO mouse recipients showed increased proinflammatory cytokine production and expression of the GVHD marker ST2 (IL-33R) and MHC class II molecules; they also exhibited decreased survival associated with acute severe gut GVHD. This rapid-onset, obesity-associated gut GVHD depended on donor CD4+ T cells and occurred even with a minor MHC mismatch between donor and recipient animals. Retrospective analysis of clinical cohorts receiving allo-HSCT transplants from unrelated donors revealed that recipients with a high body mass index (BMI, >30) had reduced survival and higher serum ST2 concentrations compared with nonobese transplant recipients. Assessment of both DIO mice and allo-HSCT recipients with a high BMI revealed reduced gut microbiota diversity and decreased Clostridiaceae abundance. Prophylactic antibiotic treatment protected DIO mouse recipients from endotoxin translocation across the gut and increased inflammatory cytokine production, as well as gut pathology and mortality, but did not protect against later development of chronic skin GVHD. These results suggest that obesity-induced alterations of the gut microbiota may affect GVHD after allo-HSCT in DIO mice, which could be ameliorated by prophylactic antibiotic treatment.
Collapse
Affiliation(s)
- Lam T Khuat
- Department of Dermatology, School of Medicine, University of California, Davis, Sacramento, CA 95817, USA
| | - Catherine T Le
- Department of Dermatology, School of Medicine, University of California, Davis, Sacramento, CA 95817, USA
| | - Chien-Chun Steven Pai
- Department of Dermatology, School of Medicine, University of California, Davis, Sacramento, CA 95817, USA
| | | | - Shernan G Holtan
- Blood and Marrow Transplant Program, University of Minnesota, Minneapolis, MN 55455, USA
| | - Armin Rashidi
- Blood and Marrow Transplant Program, University of Minnesota, Minneapolis, MN 55455, USA
| | - Sarah L Parker
- Department of Internal Medicine, University of Minnesota, Minneapolis, MN 55455, USA
| | - Dan Knights
- Department of Computer Science and Engineering, Biotechnology Institute, University of Minnesota, Minneapolis, MN 55455, USA
| | - Jesus I Luna
- Department of Dermatology, School of Medicine, University of California, Davis, Sacramento, CA 95817, USA
| | - Cordelia Dunai
- Department of Dermatology, School of Medicine, University of California, Davis, Sacramento, CA 95817, USA
| | - Ziming Wang
- Department of Dermatology, School of Medicine, University of California, Davis, Sacramento, CA 95817, USA
| | - Ian R Sturgill
- Department of Dermatology, School of Medicine, University of California, Davis, Sacramento, CA 95817, USA
| | - Kevin M Stoffel
- Department of Dermatology, School of Medicine, University of California, Davis, Sacramento, CA 95817, USA
| | - Alexander A Merleev
- Department of Dermatology, School of Medicine, University of California, Davis, Sacramento, CA 95817, USA
| | - Shyam K More
- Division of Gastroenterology, Department of Internal Medicine, School of Medicine, University of California, Davis, Sacramento, CA 95817, USA
| | - Emanual Maverakis
- Department of Dermatology, School of Medicine, University of California, Davis, Sacramento, CA 95817, USA
| | - Helen E Raybould
- Department of Anatomy, Physiology, and Cell Biology, School of Veterinary Medicine, University of California, Davis, Davis, CA 95616, USA
| | - Mingyi Chen
- Department of Pathology and Laboratory Medicine, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Robert J Canter
- Division of Surgical Oncology, Department of Surgery, School of Medicine, University of California, Davis, Sacramento, CA 95817, USA
| | - Arta M Monjazeb
- Department of Radiation Oncology, School of Medicine, University of California, Davis, Sacramento, CA 95817, USA
| | - Maneesh Dave
- Division of Gastroenterology, Department of Internal Medicine, School of Medicine, University of California, Davis, Sacramento, CA 95817, USA
| | - James L M Ferrara
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - John E Levine
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Dan L Longo
- Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Mehrdad Abedi
- Department of Internal Medicine, School of Medicine, University of California, Davis, Sacramento, CA 95817, USA
| | - Bruce R Blazar
- Masonic Cancer Center and Division of Blood and Marrow Transplantation, Department of Pediatrics, University of Minnesota, Minneapolis, MN 55455, USA
| | - William J Murphy
- Department of Dermatology, School of Medicine, University of California, Davis, Sacramento, CA 95817, USA. .,Department of Internal Medicine, School of Medicine, University of California, Davis, Sacramento, CA 95817, USA
| |
Collapse
|
41
|
Liu O, Xu J, Wang F, Jin W, Zanvit P, Wang D, Goldberg N, Cain A, Guo N, Han Y, Bynum A, Ma G, Wang S, Tang Z, Chen W. Adipose-mesenchymal stromal cells suppress experimental Sjögren syndrome by IL-33-driven expansion of ST2 + regulatory T cells. iScience 2021; 24:102446. [PMID: 33997712 PMCID: PMC8105666 DOI: 10.1016/j.isci.2021.102446] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 03/11/2021] [Accepted: 04/14/2021] [Indexed: 02/06/2023] Open
Abstract
Adipose-derived mesenchymal stromal cells (ADSCs) play important roles in the alleviation of inflammation and autoimmune diseases. Interleukin-33 (IL-33), a member of the IL-1 family, has been shown to regulate innate and adaptive immunity. However, it is still unknown whether ADSCs regulate immune responses via IL-33. We show here that ADSCs produced IL-33 in response to IL-1β stimulation, which depended on TAK1, ERK, and p38 pathways. ADSCs-derived IL-33 drove the proliferation of CD4+Foxp3+ST2+ regulatory T cells (Tregs) and alleviated experimental autoimmune Sjögren syndrome in mice. Importantly, human ADSCs also produced IL-33 in response to IL-1β. Thus, we have revealed a previously unrecognized immunoregulatory function of ADSCs by IL-33 production in experimental autoimmunity, which may have clinical applications for human immunopathology. Human and mouse ADSCs express IL-33 in response to IL-β stimulation mADSC-derived IL-33 inhibits inflammation in salivary glands in SS model mADSC-derived IL-33 expand ST2+ Tregs in vitro and in SS model
Collapse
Affiliation(s)
- Ousheng Liu
- Mucosal Immunology Section, NIDCR, NIH, Bethesda, MD 20892, USA
- Hunan Key Laboratory of Oral Health Research & Hunan 3D Printing Engineering Research Center of Oral Care & Hunan Clinical Research Center of Oral Major Diseases and Oral Health & Xiangya Stomatological Hospital & Xiangya School of Stomatology, Central South University, Changsha 410008, China
| | - Junji Xu
- Mucosal Immunology Section, NIDCR, NIH, Bethesda, MD 20892, USA
| | - Fu Wang
- Mucosal Immunology Section, NIDCR, NIH, Bethesda, MD 20892, USA
- Dalian Medical University, School of Stomatology, Dalian 116044, China
| | - Wenwen Jin
- Mucosal Immunology Section, NIDCR, NIH, Bethesda, MD 20892, USA
| | - Peter Zanvit
- Mucosal Immunology Section, NIDCR, NIH, Bethesda, MD 20892, USA
| | - Dandan Wang
- Mucosal Immunology Section, NIDCR, NIH, Bethesda, MD 20892, USA
| | - Nathan Goldberg
- Mucosal Immunology Section, NIDCR, NIH, Bethesda, MD 20892, USA
| | - Alexander Cain
- Mucosal Immunology Section, NIDCR, NIH, Bethesda, MD 20892, USA
| | - Nancy Guo
- Mucosal Immunology Section, NIDCR, NIH, Bethesda, MD 20892, USA
| | - Yichen Han
- Mucosal Immunology Section, NIDCR, NIH, Bethesda, MD 20892, USA
| | - Andrew Bynum
- Mucosal Immunology Section, NIDCR, NIH, Bethesda, MD 20892, USA
| | - Guowu Ma
- Dalian Medical University, School of Stomatology, Dalian 116044, China
| | - Songlin Wang
- Molecular Laboratory for Gene Therapy and Tooth Regeneration, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, School of Stomatology, Capital Medical University, Beijing 100050, China
| | - Zhangui Tang
- Hunan Key Laboratory of Oral Health Research & Hunan 3D Printing Engineering Research Center of Oral Care & Hunan Clinical Research Center of Oral Major Diseases and Oral Health & Xiangya Stomatological Hospital & Xiangya School of Stomatology, Central South University, Changsha 410008, China
| | - Wanjun Chen
- Mucosal Immunology Section, NIDCR, NIH, Bethesda, MD 20892, USA
- Corresponding author
| |
Collapse
|
42
|
Strattan E, Hildebrandt GC. Mast Cell Involvement in Fibrosis in Chronic Graft-Versus-Host Disease. Int J Mol Sci 2021; 22:2385. [PMID: 33673565 PMCID: PMC7956846 DOI: 10.3390/ijms22052385] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 02/21/2021] [Indexed: 02/07/2023] Open
Abstract
Allogeneic hematopoietic stem cell transplantation (HSCT) is most commonly a treatment for inborn defects of hematopoiesis or acute leukemias. Widespread use of HSCT, a potentially curative therapy, is hampered by onset of graft-versus-host disease (GVHD), classified as either acute or chronic GVHD. While the pathology of acute GVHD is better understood, factors driving GVHD at the cellular and molecular level are less clear. Mast cells are an arm of the immune system that are known for atopic disease. However, studies have demonstrated that they can play important roles in tissue homeostasis and wound healing, and mast cell dysregulation can lead to fibrotic disease. Interestingly, in chronic GVHD, aberrant wound healing mechanisms lead to pathological fibrosis, but the cellular etiology driving this is not well-understood, although some studies have implicated mast cells. Given this novel role, we here review the literature for studies of mast cell involvement in the context of chronic GVHD. While there are few publications on this topic, the papers excellently characterized a niche for mast cells in chronic GVHD. These findings may be extended to other fibrosing diseases in order to better target mast cells or their mediators for treatment of fibrotic disease.
Collapse
Affiliation(s)
| | - Gerhard Carl Hildebrandt
- Division of Hematology and Blood & Marrow Transplant, Markey Cancer Center, University of Kentucky, Lexington, KY 40536, USA;
| |
Collapse
|
43
|
Dwyer GK, Turnquist HR. Untangling Local Pro-Inflammatory, Reparative, and Regulatory Damage-Associated Molecular-Patterns (DAMPs) Pathways to Improve Transplant Outcomes. Front Immunol 2021; 12:611910. [PMID: 33708206 PMCID: PMC7940545 DOI: 10.3389/fimmu.2021.611910] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 01/05/2021] [Indexed: 12/28/2022] Open
Abstract
Detrimental inflammatory responses after solid organ transplantation are initiated when immune cells sense pathogen-associated molecular patterns (PAMPs) and certain damage-associated molecular patterns (DAMPs) released or exposed during transplant-associated processes, such as ischemia/reperfusion injury (IRI), surgical trauma, and recipient conditioning. These inflammatory responses initiate and propagate anti-alloantigen (AlloAg) responses and targeting DAMPs and PAMPs, or the signaling cascades they activate, reduce alloimmunity, and contribute to improved outcomes after allogeneic solid organ transplantation in experimental studies. However, DAMPs have also been implicated in initiating essential anti-inflammatory and reparative functions of specific immune cells, particularly Treg and macrophages. Interestingly, DAMP signaling is also involved in local and systemic homeostasis. Herein, we describe the emerging literature defining how poor outcomes after transplantation may result, not from just an over-abundance of DAMP-driven inflammation, but instead an inadequate presence of a subset of DAMPs or related molecules needed to repair tissue successfully or re-establish tissue homeostasis. Adverse outcomes may also arise when these homeostatic or reparative signals become dysregulated or hijacked by alloreactive immune cells in transplant niches. A complete understanding of the critical pathways controlling tissue repair and homeostasis, and how alloimmune responses or transplant-related processes disrupt these will lead to new immunotherapeutics that can prevent or reverse the tissue pathology leading to lost grafts due to chronic rejection.
Collapse
Affiliation(s)
- Gaelen K Dwyer
- Departments of Surgery and Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States.,Thomas E. Starzl Transplantation Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Hēth R Turnquist
- Departments of Surgery and Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States.,Thomas E. Starzl Transplantation Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States.,McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| |
Collapse
|
44
|
Aarstad HH, Moe SEE, Bruserud Ø, Lybak S, Aarstad HJ, Tvedt THA. The Acute Phase Reaction and Its Prognostic Impact in Patients with Head and Neck Squamous Cell Carcinoma: Single Biomarkers Including C-Reactive Protein Versus Biomarker Profiles. Biomedicines 2020; 8:biomedicines8100418. [PMID: 33066437 PMCID: PMC7602291 DOI: 10.3390/biomedicines8100418] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2020] [Revised: 09/26/2020] [Accepted: 10/13/2020] [Indexed: 12/12/2022] Open
Abstract
C-reactive protein (CRP) has a prognostic impact in head and neck squamous cell carcinoma (HNSCC). However, the acute phase reaction involves many other proteins depending on its inducing events, including various cytokines that can function as reaction inducers. In the present study, we compared the pretreatment acute phase cytokine profile for 144 patients with potentially curative HNSCC. We investigated the systemic levels of interleukin (IL)6 family mediators (glycoprotein (gp130), IL6 receptor (R)α, IL6, IL27, IL31, oncostatin M (OSM), ciliary neurotrophic factor (CNTF)), IL1 subfamily members (IL1R antagonist (A), IL33Rα), and tumor necrosis factor (TNF)α. Patient subsets identified from this 10-mediator profile did not differ with regard to disease stage, human papilloma virus (HPV) status, CRP levels, or death cause. Increased CRP, IL6, and IL1RA levels were independent markers for HNSCC-related death in the whole patient population. Furthermore, gp130, IL6Rα, and IL31 were suggested to predict prognosis among tumor HPV-negative patients. Only IL6 predicted survival in HPV-positive patients. Finally, we did a clustering analysis of HPV-negative patients based on six acute phase mediators that showed significant or borderline association with prognosis in Kaplan–Meier analyses; three subsets could then be identified, and they differed in survival (p < 0.001). To conclude, (i) HPV-negative and HPV-positive HNSCC patients show similar variations of their systemic acute phase profiles; (ii) the prognostic impact of single mediators differs between these two patient subsets; and (iii) for HPV-negative patients, acute phase profiling identifies three patient subsets that differ significantly in survival.
Collapse
Affiliation(s)
- Helene Hersvik Aarstad
- Department of Clinical Science, Faculty of Medicine, University of Bergen, 5021 Bergen, Norway; (H.H.A.); (Ø.B.)
- Department of Otolaryngology/Head and Neck Surgery, Haukeland University Hospital, 5021 Bergen, Norway; (S.E.E.M.); (S.L.)
| | - Svein Erik Emblem Moe
- Department of Otolaryngology/Head and Neck Surgery, Haukeland University Hospital, 5021 Bergen, Norway; (S.E.E.M.); (S.L.)
| | - Øystein Bruserud
- Department of Clinical Science, Faculty of Medicine, University of Bergen, 5021 Bergen, Norway; (H.H.A.); (Ø.B.)
- Section for Hematology, Department of Medicine, Haukeland University Hospital, 5021 Bergen, Norway;
| | - Stein Lybak
- Department of Otolaryngology/Head and Neck Surgery, Haukeland University Hospital, 5021 Bergen, Norway; (S.E.E.M.); (S.L.)
| | - Hans Jørgen Aarstad
- Department of Otolaryngology/Head and Neck Surgery, Haukeland University Hospital, 5021 Bergen, Norway; (S.E.E.M.); (S.L.)
- Department of Clinical Medicine, Faculty of Medicine, University of Bergen, 5021 Bergen, Norway
- Correspondence: ; Tel.: +47-5597-2664
| | | |
Collapse
|
45
|
Olson KE, Namminga KL, Schwab AD, Thurston MJ, Lu Y, Woods A, Lei L, Shen W, Wang F, Joseph SB, Gendelman HE, Mosley RL. Neuroprotective Activities of Long-Acting Granulocyte-Macrophage Colony-Stimulating Factor (mPDM608) in 1-Methyl-4-Phenyl-1,2,3,6-Tetrahydropyridine-Intoxicated Mice. Neurotherapeutics 2020; 17:1861-1877. [PMID: 32638217 PMCID: PMC7851309 DOI: 10.1007/s13311-020-00877-8] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Loss of dopaminergic neurons along the nigrostriatal axis, neuroinflammation, and peripheral immune dysfunction are the pathobiological hallmarks of Parkinson's disease (PD). Granulocyte-macrophage colony-stimulating factor (GM-CSF) has been successfully tested for PD treatment. GM-CSF is a known immune modulator that induces regulatory T cells (Tregs) and serves as a neuronal protectant in a broad range of neurodegenerative diseases. Due to its short half-life, limited biodistribution, and potential adverse effects, alternative long-acting treatment schemes are of immediate need. A long-acting mouse GM-CSF (mPDM608) was developed through Calibr, a Division of Scripps Research. Following mPDM608 treatment, complete hematologic and chemistry profiles and T-cell phenotypes and functions were determined. Neuroprotective and anti-inflammatory capacities of mPDM608 were assessed in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-intoxicated mice that included transcriptomic immune profiles. Treatment with a single dose of mPDM608 resulted in dose-dependent spleen and white blood cell increases with parallel enhancements in Treg numbers and immunosuppressive function. A shift in CD4+ T-cell gene expression towards an anti-inflammatory phenotype corresponded with decreased microgliosis and increased dopaminergic neuronal cell survival. mPDM608 elicited a neuroprotective peripheral immune transformation. The observed phenotypic shift and neuroprotective response was greater than observed with recombinant GM-CSF (rGM-CSF) suggesting human PDM608 as a candidate for PD treatment.
Collapse
Affiliation(s)
- Katherine E. Olson
- Department of Pharmacology and Experimental Neuroscience, Center for Neurodegenerative Disorders, University of Nebraska Medical Center, Omaha, NE 68198 USA
| | - Krista L. Namminga
- Department of Pharmacology and Experimental Neuroscience, Center for Neurodegenerative Disorders, University of Nebraska Medical Center, Omaha, NE 68198 USA
| | - Aaron D. Schwab
- Department of Pharmacology and Experimental Neuroscience, Center for Neurodegenerative Disorders, University of Nebraska Medical Center, Omaha, NE 68198 USA
| | - Mackenzie J. Thurston
- Department of Pharmacology and Experimental Neuroscience, Center for Neurodegenerative Disorders, University of Nebraska Medical Center, Omaha, NE 68198 USA
| | - Yaman Lu
- Department of Pharmacology and Experimental Neuroscience, Center for Neurodegenerative Disorders, University of Nebraska Medical Center, Omaha, NE 68198 USA
| | - Ashley Woods
- Calibr, a Division of Scripps Research, La Jolla, CA 92037 USA
| | - Lei Lei
- Calibr, a Division of Scripps Research, La Jolla, CA 92037 USA
| | - Weijun Shen
- Calibr, a Division of Scripps Research, La Jolla, CA 92037 USA
| | - Feng Wang
- Key Laboratory of Protein and Peptide Pharmaceuticals, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101 China
| | - Sean B. Joseph
- Calibr, a Division of Scripps Research, La Jolla, CA 92037 USA
| | - Howard E. Gendelman
- Department of Pharmacology and Experimental Neuroscience, Center for Neurodegenerative Disorders, University of Nebraska Medical Center, Omaha, NE 68198 USA
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 6898-5880 USA
| | - R. Lee Mosley
- Department of Pharmacology and Experimental Neuroscience, Center for Neurodegenerative Disorders, University of Nebraska Medical Center, Omaha, NE 68198 USA
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 6898-5880 USA
| |
Collapse
|
46
|
The MAGIC algorithm probability is a validated response biomarker of treatment of acute graft-versus-host disease. Blood Adv 2020; 3:4034-4042. [PMID: 31816061 DOI: 10.1182/bloodadvances.2019000791] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Accepted: 09/19/2019] [Indexed: 12/12/2022] Open
Abstract
The Mount Sinai Acute GVHD International Consortium (MAGIC) algorithm probability (MAP), derived from 2 serum biomarkers, measures damage to crypts in the gastrointestinal tract during graft-versus-host disease (GVHD). We hypothesized that changes in MAP after treatment could validate it as a response biomarker. We prospectively collected serum samples and clinical stages of acute GVHD from 615 patients receiving hematopoietic cell transplantation in 20 centers at initiation of first-line systemic treatment and 4 weeks later. We computed MAPs and clinical responses and compared their abilities to predict 6-month nonrelapse mortality (NRM) in the validation cohort (n = 367). After 4 weeks of treatment, MAPs predicted NRM better than the change in clinical symptoms in all patients and identified 2 groups with significantly different NRM in both clinical responders (40% vs 12%, P < .0001) and nonresponders (65% vs 25%, P < .0001). MAPs successfully reclassified patients for NRM risk within every clinical grade of acute GVHD after 4 weeks of treatment. At the beginning of treatment, patients with a low MAP that rose above the threshold of 0.290 after 4 weeks of treatment had a significant increase in NRM, whereas patients with a high MAP at onset that fell below that threshold after treatment had a striking decrease in NRM that translated into clear differences in overall survival. We conclude that a MAP measured before and after treatment of acute GVHD is a response biomarker that predicts long-term outcomes more accurately than change in clinical symptoms. MAPs have the potential to guide therapy for acute GVHD and may function as a useful end point in clinical trials.
Collapse
|
47
|
Sabapathy V, Venkatadri R, Dogan M, Sharma R. The Yin and Yang of Alarmins in Regulation of Acute Kidney Injury. Front Med (Lausanne) 2020; 7:441. [PMID: 32974364 PMCID: PMC7472534 DOI: 10.3389/fmed.2020.00441] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Accepted: 07/06/2020] [Indexed: 12/16/2022] Open
Abstract
Acute kidney injury (AKI) is a major clinical burden affecting 20 to 50% of hospitalized and intensive care patients. Irrespective of the initiating factors, the immune system plays a major role in amplifying the disease pathogenesis with certain immune cells contributing to renal damage, whereas others offer protection and facilitate recovery. Alarmins are small molecules and proteins that include granulysins, high-mobility group box 1 protein, interleukin (IL)-1α, IL-16, IL-33, heat shock proteins, the Ca++ binding S100 proteins, adenosine triphosphate, and uric acid. Alarmins are mostly intracellular molecules, and their release to the extracellular milieu signals cellular stress or damage, generally leading to the recruitment of the cells of the immune system. Early studies indicated a pro-inflammatory role for the alarmins by contributing to immune-system dysregulation and worsening of AKI. However, recent developments demonstrate anti-inflammatory mechanisms of certain alarmins or alarmin-sensing receptors, which may participate in the prevention, resolution, and repair of AKI. This dual function of alarmins is intriguing and has confounded the role of alarmins in AKI. In this study, we review the contribution of various alarmins to the pathogenesis of AKI in experimental and clinical studies. We also analyze the approaches for the therapeutic utilization of alarmins for AKI.
Collapse
Affiliation(s)
| | | | | | - Rahul Sharma
- Division of Nephrology, Department of Medicine, Center for Immunity, Inflammation, and Regenerative Medicine (CIIR), University of Virginia, Charlottesville, VA, United States
| |
Collapse
|
48
|
The Functional Roles of IL-33/ST2 Axis in Ocular Diseases. Mediators Inflamm 2020; 2020:5230716. [PMID: 32908451 PMCID: PMC7450335 DOI: 10.1155/2020/5230716] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 07/25/2020] [Accepted: 07/31/2020] [Indexed: 01/10/2023] Open
Abstract
Interleukin-33 (IL-33), an important member of the IL-1 family, plays a pivotal role in regulating immune responses via combining with its receptor suppression of tumorigenicity 2 (ST2). We have already known IL-33/ST2 axis participates in the pathogenesis of various diseases, including liver diseases, renal diseases, and neurological diseases. Recently, emerging studies are indicating that IL-33/ST2 is also involved in a wide range of ocular diseases, such as allergic eye disease, keratitis and corneal regeneration, dry eye disease, uveitis, vitreoretinal diseases, and neuromyelitis optica spectrum disorder. In this review, we will summarize and discuss the current understanding about the functional roles of IL-33/ST2 in eyes, with an attempt to explore the possible study perspectives and therapeutic alternatives in the future.
Collapse
|
49
|
Chen S, Zeiser R. Novel Biomarkers for Outcome After Allogeneic Hematopoietic Stem Cell Transplantation. Front Immunol 2020; 11:1854. [PMID: 33013836 PMCID: PMC7461883 DOI: 10.3389/fimmu.2020.01854] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2020] [Accepted: 07/09/2020] [Indexed: 12/29/2022] Open
Abstract
Allogeneic hematopoietic stem cell transplantation (allo-HSCT) is a well-established curative treatment for various malignant hematological diseases. However, its clinical success is substantially limited by major complications including graft-vs.-host disease (GVHD) and relapse of the underlying disease. Although these complications are known to lead to significant morbidity and mortality, standardized pathways for risk stratification of patients undergoing allo-HSCT are lacking. Recent advances in the development of diagnostic and prognostic tools have allowed the identification of biomarkers in order to predict outcome after allo-HSCT. This review will provide a summary of clinically relevant biomarkers that have been studied to predict the development of acute GVHD, the responsiveness of affected patients to immunosuppressive treatment and the risk of non-relapse mortality. Furthermore, biomarkers associated with increased risk of relapse and subsequent mortality will be discussed.
Collapse
Affiliation(s)
- Sophia Chen
- Department of Immunology, Memorial Sloan Kettering Cancer Center, Sloan Kettering Institute, New York, NY, United States.,Department of Medicine I, Faculty of Medicine, Medical Center, University of Freiburg, Freiburg, Germany
| | - Robert Zeiser
- Department of Medicine I, Faculty of Medicine, Medical Center, University of Freiburg, Freiburg, Germany.,German Cancer Consortium (DKTK), Partner Site Freiburg, Freiburg, Germany.,Signalling Research Centres BIOSS and CIBSS - Centre for Integrative Biological Signalling Studies, University of Freiburg, Freiburg, Germany
| |
Collapse
|
50
|
Hill GR, Koyama M. Cytokines and costimulation in acute graft-versus-host disease. Blood 2020; 136:418-428. [PMID: 32526028 PMCID: PMC7378458 DOI: 10.1182/blood.2019000952] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Accepted: 01/18/2020] [Indexed: 12/11/2022] Open
Abstract
Allogeneic hematopoietic stem cell transplantation (alloSCT) is an important curative therapy for high-risk hematological malignancies, but the development of severe and/or steroid-refractory acute graft-versus-host disease (aGVHD) remains a significant limitation to optimal outcomes. New approaches to prevent and treat aGVHD remain an unmet need that can be best addressed by understanding the complex disease pathophysiology. It is now clear that chemoradiotherapy used prior to alloSCT induces the release of endogenous alarmins (eg, HMGB-1, ATP, IL-1α, IL-33) from recipient tissue. Exogenous pathogen-derived molecules (eg, lipopolysaccharide, nucleic acids) also translocate from the gastrointestinal tract lumen. Together, these danger signals activate antigen-presenting cells (APCs) to efficiently present alloantigen to donor T cells while releasing cytokines (eg, interleukin-12 [IL-12], IL-23, IL-6, IL-27, IL-10, transforming growth factor-β) that expand and differentiate both pathogenic and regulatory donor T cells. Concurrent costimulatory signals at the APC-T-cell interface (eg, CD80/CD86-CD28, CD40-CD40L, OX40L-OX40, CD155/CD112-DNAM-1) and subsequent coinhibitory signals (eg, CD80/CD86-CTLA4, PDL1/2-PD1, CD155/CD112-TIGIT) are critical to the acquisition of effector T-cell function and ensuing secretion of pathogenic cytokines (eg, IL-17, interferon-γ, tissue necrosis factor, granulocyte-macrophage colony-stimulating factor) and cytolytic degranulation pathway effectors (eg, perforin/granzyme). This review focuses on the combination of cytokine and costimulatory networks at the T-cell surface that culminates in effector function and subsequent aGVHD in target tissue. Together, these pathways now represent robust and clinically tractable targets for preventing the initiation of deleterious immunity after alloSCT.
Collapse
Affiliation(s)
- Geoffrey R Hill
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA; and
- Division of Medical Oncology, Department of Medicine, University of Washington, Seattle, WA
| | - Motoko Koyama
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA; and
| |
Collapse
|