1
|
Ni M, Cui J, Yang X, Ding Y, Zhao P, Hu T, Zhan Y, Kang Q, Hu X, Zhao J, Xu Y, Chen L, Liu M, Zhao M, Zhang F, Huang S, Li Y, Yang X, Zhang L, Zhang T, Deng B, Yang B, Lu D, Wang J. Dual roles of CD11b +CD33 +HLA-DR -/lowCD14 - myeloid-derived suppressor cells with a granulocytic morphology following allogeneic hematopoietic stem cell transplantation: from inflammation promoters to immune suppressors within 90 days. Front Immunol 2024; 15:1403272. [PMID: 39040102 PMCID: PMC11260618 DOI: 10.3389/fimmu.2024.1403272] [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: 03/19/2024] [Accepted: 06/25/2024] [Indexed: 07/24/2024] Open
Abstract
Introduction Granulocytic myeloid-derived suppressor cells (G-MDSCs) show fast recovery following allogeneic hematopoietic stem cell transplantation (allo-HSCT) constituting the major part of peripheral blood in the early phase. Although G-MDSCs mediate immune suppression through multiple mechanisms, they may also promote inflammation under specific conditions. Methods G-MDSCs were isolated from 82 patients following allo-HSCT within 90 days after allo-HSCT, and their interactions with autologous CD3+ T-cells were examined. T-cell proliferation was assessed by flow cytometry following CFSE staining, while differentiation and interferon-γ secretion were characterized using chemokine receptor profiling and ELISpot assays, respectively. NK cell cytotoxicity was evaluated through co-culture with K562 cells. An aGVHD xenogeneic model in humanized mice was employed to study the in vivo effects of human leukocytes. Furthermore, transcriptional alterations in G-MDSCs were analyzed via RNA sequencing to investigate functional transitions. Results G-MDSCs promoted inflammation in the early-stage, by facilitating cytokine secretion and proliferation of T cells, as well as their differentiation into pro-inflammatory T helper subsets. At day 28, patients with a higher number of G-MDSCs exhibited an increased risk of developing grades II-IV aGvHD. Besides, adoptive transfer of G-MDSCs from patients at day 28 into humanized mice exacerbated aGvHD. However, at day 90, G-MDSCs led to immunosuppression, characterized by upregulated expression of indoleamine 2,3-dioxygenase gene and interleukin-10 secretion, coupled with the inhibition of T cell proliferation. Furthermore, transcriptional analysis of G-MDSCs at day 28 and day 90 revealed that 1445 genes were differentially expressed. These genes were associated with various pathways, revealing the molecular signatures of early post-transplant differentiation in G-MDSCs. In addition, genes linked to the endoplasmic reticulum stress were upregulated in patients without aGvHD. The acquisition of immunosuppressive function by G-MDSCs may depend on the activation of CXCL2 and DERL1 genes. Conclusion Our findings revealed the alteration in the immune characteristics of G-MDSCs within the first 90 days post-allo-HSCT. Moreover, the quantity of G-MDSCs at day 28 may serve as a predictive indicator for the development of aGvHD.
Collapse
Affiliation(s)
- Ming Ni
- Department of Hematology, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Jing Cui
- Department of Dermatology, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Xin Yang
- Department of Hematology, Affiliated Hospital of Guizhou Medical University, Guiyang, China
- Department of Hematology, The Second Affiliated Hospital of Guizhou Medical University, Kaili, China
| | - Yuntian Ding
- Department of Internal Medicine V, University Clinic Heidelberg, Heidelberg, Germany
| | - Peng Zhao
- Department of Hematology, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Tianzhen Hu
- Department of Hematology, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Yun Zhan
- Department of Hematology, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Qian Kang
- Department of Hematology, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Xiuying Hu
- Department of Hematology, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Jiangyuan Zhao
- Department of Hematology, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Yao Xu
- Department of Hematology, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Lu Chen
- Department of Hematology, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Min Liu
- Department of Hematology, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Mei Zhao
- Department of Hematology, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Fengqi Zhang
- Department of Hematology, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Shisi Huang
- Department of Hematology, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Ya Li
- Department of Hematology, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Xueying Yang
- Department of Hematology, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Luxin Zhang
- Department of Hematology, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Tianzhuo Zhang
- Department of Hematology, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Bo Deng
- Department of Hematology, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Bing Yang
- Department of Hematology, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Deqin Lu
- Department of Pathophysiology, Guizhou Medical University, Guiyang, China
| | - Jishi Wang
- Department of Hematology, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| |
Collapse
|
2
|
Lin F, Zuo Y, Zhang Y, Cheng Y, Han T, Mo X, Suo P, Sun Y, Tang F, Wang F, Yan C, Chen Y, Han W, Wang J, Wang Y, Zhang X, Liu K, Huang X, Xu L. The impact of pretransplant serum ferritin on haploidentical hematopoietic stem cell transplant for acquired severe aplastic anemia in children and adolescents. Pediatr Blood Cancer 2022; 69:e29845. [PMID: 35731841 DOI: 10.1002/pbc.29845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Revised: 05/08/2022] [Accepted: 05/31/2022] [Indexed: 01/19/2023]
Abstract
Haploidentical hematopoietic stem cell transplant (haplo-HSCT) provides an important alternative for children and adolescents with acquired severe aplastic anemia (SAA) lacking matched donors. To test whether pretransplant serum ferritin (SF) represents a candidate predictor for survival and a potential biomarker for graft-versus-host disease (GvHD) in pediatric haplo-HSCT, we retrospectively evaluated 147 eligible patients with SAA who underwent haplo-HSCT. The patients were divided into the low-SF group (< 1000 ng/mL) and the high-SF group (≥ 1000 ng/mL). We found that SF ≥1000 ng/mL independently increased the risk of grade II-IV aGvHD (HR = 2.596; 95% CI, 1.304-5.167, P = 0.007) and grade III-IV aGvHD (HR = 3.350; 95% CI, 1.162-9.658, P = 0.025). Similar probabilities of transplant-related mortality at 100 days were observed in the two groups (6.19 ± 2.45% vs 8.00 ± 3.84%, P = 0.168). The two-year overall survival (85.29 ± 3.89% vs 92.00% ± 3.84%, P = 0.746) and failure-free survival (83.23% ± 4.08% vs 83.37% ± 6.27%, P = 0.915) were comparable. GvHD-/failure-free survival were 60.06 ± 5.10% and 75.56 ± 6.87%, respectively (P = 0.056). In conclusion, elevated pretransplant SF level is associated with higher incidences of grade II-IV aGvHD and grade III-IV aGvHD. However, it is not associated with worse survival after haplo-HSCT for children and adolescent patients with SAA.
Collapse
Affiliation(s)
- Fan Lin
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Collaborative Innovation Center of Hematology, Beijing Key Laboratory of Haematopoietic Stem Cell Transplant, Beijing, China
| | - Yangyang Zuo
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Collaborative Innovation Center of Hematology, Beijing Key Laboratory of Haematopoietic Stem Cell Transplant, Beijing, China
| | - Yuanyuan Zhang
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Collaborative Innovation Center of Hematology, Beijing Key Laboratory of Haematopoietic Stem Cell Transplant, Beijing, China
| | - Yifei Cheng
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Collaborative Innovation Center of Hematology, Beijing Key Laboratory of Haematopoietic Stem Cell Transplant, Beijing, China
| | - Tingting Han
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Collaborative Innovation Center of Hematology, Beijing Key Laboratory of Haematopoietic Stem Cell Transplant, Beijing, China
| | - Xiaodong Mo
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Collaborative Innovation Center of Hematology, Beijing Key Laboratory of Haematopoietic Stem Cell Transplant, Beijing, China
| | - Pan Suo
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Collaborative Innovation Center of Hematology, Beijing Key Laboratory of Haematopoietic Stem Cell Transplant, Beijing, China
| | - Yuqian Sun
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Collaborative Innovation Center of Hematology, Beijing Key Laboratory of Haematopoietic Stem Cell Transplant, Beijing, China
| | - Feifei Tang
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Collaborative Innovation Center of Hematology, Beijing Key Laboratory of Haematopoietic Stem Cell Transplant, Beijing, China
| | - Fengrong Wang
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Collaborative Innovation Center of Hematology, Beijing Key Laboratory of Haematopoietic Stem Cell Transplant, Beijing, China
| | - Chenhua Yan
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Collaborative Innovation Center of Hematology, Beijing Key Laboratory of Haematopoietic Stem Cell Transplant, Beijing, China
| | - Yuhong Chen
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Collaborative Innovation Center of Hematology, Beijing Key Laboratory of Haematopoietic Stem Cell Transplant, Beijing, China
| | - Wei Han
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Collaborative Innovation Center of Hematology, Beijing Key Laboratory of Haematopoietic Stem Cell Transplant, Beijing, China
| | - Jingzhi Wang
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Collaborative Innovation Center of Hematology, Beijing Key Laboratory of Haematopoietic Stem Cell Transplant, Beijing, China
| | - Yu Wang
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Collaborative Innovation Center of Hematology, Beijing Key Laboratory of Haematopoietic Stem Cell Transplant, Beijing, China
| | - Xiaohui Zhang
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Collaborative Innovation Center of Hematology, Beijing Key Laboratory of Haematopoietic Stem Cell Transplant, Beijing, China
| | - Kaiyan Liu
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Collaborative Innovation Center of Hematology, Beijing Key Laboratory of Haematopoietic Stem Cell Transplant, Beijing, China
| | - Xiaojun Huang
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Collaborative Innovation Center of Hematology, Beijing Key Laboratory of Haematopoietic Stem Cell Transplant, Beijing, China.,Peking-Tsinghua Centre for Life Sciences, Beijing, China
| | - Lanping Xu
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Collaborative Innovation Center of Hematology, Beijing Key Laboratory of Haematopoietic Stem Cell Transplant, Beijing, China
| |
Collapse
|
3
|
Verheij M, Zeerleder S, Voermans C. Heme oxygenase-1: Equally important in allogeneic hematopoietic stem cell transplantation and organ transplantation? Transpl Immunol 2021; 68:101419. [PMID: 34089821 DOI: 10.1016/j.trim.2021.101419] [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: 03/03/2021] [Revised: 05/30/2021] [Accepted: 05/31/2021] [Indexed: 12/17/2022]
Abstract
The intracellular enzyme heme oxygenase-1 (HO-1) is responsible for the degradation of cell-free (cf) heme. Cfheme, released upon cell damage and cell death from hemoglobin, mitochondria and myoglobin, functions as a powerful damage-associated molecular pattern (DAMP). Indeed, cfheme plays a role in a myriad of diseases characterized by (systemic) inflammation, and its rapid degradation by HO-1 is pivotal to maintain homeostasis. In the past decade, HO-1 has been extensively studied for its potential protective role in different transplantation settings, including allogeneic hematopoietic stem cell transplantation (HSCT), solid organ transplantation and pancreatic islet transplantation. These studies have shown that HO-1 can be induced by a wide range of molecules, and that induction of HO-1 has the potential to significantly reduce the incidence and severity of transplantation-related complications such as graft-versus-host disease (GvHD) and ischemia/reperfusion injury (IRI). As such, further investigation into the use of HO-1-inducing agents in human transplantation settings to facilitate the potential use of these agents in the clinic is warranted. In this review, we summarize the literature of the past 10 years on the role of HO-1 in allogeneic HSCT, solid organ transplantation (focusing on kidney and liver) and pancreatic islet transplantation. Furthermore, we provide a hypothesis about the way that HO-1 is able to provide protection against acute GvHD after allogeneic HSCT. A total of 48 research articles and 17 review articles were included in this review.
Collapse
Affiliation(s)
- Myrddin Verheij
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory, Amsterdam, the Netherlands
| | - Sacha Zeerleder
- Department of Hematology and Central Hematology Laboratory, Inselspital, Bern University Hospital, University of Bern, Switzerland; Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, Amsterdam, the Netherlands; Department for Biomedical Research, University of Bern, Switzerland
| | - Carlijn Voermans
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory, Amsterdam, the Netherlands.
| |
Collapse
|
4
|
Spilleboudt C, De Wilde V, Lewalle P, Cabanne L, Leclerc M, Beckerich F, Bories D, Cardoso S, Soares MP, Vokaer B, Hougardy JM, Flamand V, Racapé J, Abramowicz M, Maury S, Le Moine A. Donor-Derived Myeloid Heme Oxygenase-1 Controls the Development of Graft-Versus-Host Disease. Front Immunol 2021; 11:579151. [PMID: 33537027 PMCID: PMC7849683 DOI: 10.3389/fimmu.2020.579151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 11/30/2020] [Indexed: 11/13/2022] Open
Abstract
Graft-versus-host disease (GVHD) remains a major clinical drawback of allogeneic hematopoietic stem cell transplantation (HSCT). Here, we investigated how the stress responsive heme catabolizing enzyme heme oxygenase-1 (HO-1, encoded by HMOX1) regulates GVHD in response to allogeneic hematopoietic stem cell transplantation in mice and humans. We found that deletion of the Hmox1 allele, specifically in the myeloid compartment of mouse donor bone marrow, promotes the development of aggressive GVHD after allogeneic transplantation. The mechanism driving GVHD in mice transplanted with allogeneic bone marrow lacking HO-1 expression in the myeloid compartment involves enhanced T cell alloreactivity. The clinical relevance of these observations was validated in two independent cohorts of HSCT patients. Individuals transplanted with hematopoietic stem cells from donors carrying a long homozygous (GT)n repeat polymorphism (L/L) in the HMOX1 promoter, which is associated with lower HO-1 expression, were at higher risk of developing severe acute GVHD as compared to donors carrying a short (GT)n repeat (S/L or S/S) polymorphism associated with higher HO-1 expression. In this study, we showed the unique importance of donor-derived myeloid HO-1 in the prevention of lethal experimental GVHD and we corroborated this observation by demonstrating the association between human HMOX1 (GT)n microsatellite polymorphisms and the incidence of severe acute GVHD in two independent HSCT patient cohorts. Donor-derived myeloid HO-1 constitutes a potential therapeutic target for HSCT patients and large-scale prospective studies in HSCT patients are necessary to validate the HO-1 L/L genotype as an independent risk factor for developing severe acute GVHD.
Collapse
Affiliation(s)
- Chloé Spilleboudt
- Institute for Medical Immunology, Université Libre de Bruxelles, Gosselies, Belgium
| | - Virginie De Wilde
- Institute for Medical Immunology, Université Libre de Bruxelles, Gosselies, Belgium
- Erasme Hospital, Hematology Department, Université libre de Bruxelles, Brussels, Belgium
| | - Philippe Lewalle
- Jules Bordet Institute, Hematology Department, Université libre de Bruxelles, Brussels, Belgium
| | - Ludovic Cabanne
- AP-HP, Hôpital Henri Mondor, Department of Hematology, Créteil, France
- University Paris Est Créteil (UPEC), Créteil, France
| | - Mathieu Leclerc
- AP-HP, Hôpital Henri Mondor, Department of Hematology, Créteil, France
- University Paris Est Créteil (UPEC), Créteil, France
| | - Florence Beckerich
- AP-HP, Hôpital Henri Mondor, Department of Hematology, Créteil, France
- University Paris Est Créteil (UPEC), Créteil, France
| | - Dominique Bories
- AP-HP, Hôpital Henri Mondor, Department of Hematology, Créteil, France
- University Paris Est Créteil (UPEC), Créteil, France
| | | | | | - Benoît Vokaer
- Institute for Medical Immunology, Université Libre de Bruxelles, Gosselies, Belgium
| | - Jean-Michel Hougardy
- Erasme Hospital, Nephrology and Internal Medicine Department, Université libre de Bruxelles, Brussels, Belgium
| | - Véronique Flamand
- Institute for Medical Immunology, Université Libre de Bruxelles, Gosselies, Belgium
| | - Judith Racapé
- Erasme Hospital, Nephrology and Internal Medicine Department, Université libre de Bruxelles, Brussels, Belgium
- Centre de Recherche Épidémiologie, Biostatistique et Recherche clinique, École de Santé Publique, Université libre de Bruxelles, Brussels, Belgium
| | - Marc Abramowicz
- Department of Genetic Medicine and Development, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Sébastien Maury
- AP-HP, Hôpital Henri Mondor, Department of Hematology, Créteil, France
- University Paris Est Créteil (UPEC), Créteil, France
| | - Alain Le Moine
- Institute for Medical Immunology, Université Libre de Bruxelles, Gosselies, Belgium
- Erasme Hospital, Nephrology and Internal Medicine Department, Université libre de Bruxelles, Brussels, Belgium
| |
Collapse
|
5
|
Donor Heme Oxygenase-1 Promoter Gene Polymorphism Predicts Survival after Unrelated Bone Marrow Transplantation for High-Risk Patients. Cancers (Basel) 2020; 12:cancers12020424. [PMID: 32059452 PMCID: PMC7072481 DOI: 10.3390/cancers12020424] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Revised: 01/31/2020] [Accepted: 02/10/2020] [Indexed: 12/14/2022] Open
Abstract
Heme oxygenase-1 (HO-1), an intracellular enzyme that catalyzes the degradation of heme into biliverdin, free iron, and carbon monoxide, exerts anti-inflammatory and cytoprotective effects against endothelial cell injury. The HO-1 promoter gene has one important single-nucleotide polymorphism (SNP) rs2071746 (-413A>T) that is functional, and the A allele has been reported to be associated with higher HO-1 expression levels than the T allele. We investigated the influence of the HO-1 rs2071746 SNP on the transplant outcomes in 593 patients with hematological malignancies undergoing unrelated, human leukocyte antigen (HLA)-matched, T-cell-replete bone marrow transplantation (BMT) through the Japan Donor Marrow Program. In patients with high-risk diseases, the donor A/A or A/T genotype was associated with better 5 year overall survival (35% vs. 25%; p = 0.03) and 5 year disease-free survival (35% vs. 22%; p = 0.0072), compared to the donor T/T genotype. These effects were not observed in patients with low-risk diseases. The current findings therefore indicate that HO-1 rs2071746 genotyping could be useful for selecting donors and tailoring transplant strategies for patients with high-risk hematologic malignancies.
Collapse
|
6
|
Li H, Kaiser TK, Borschiwer M, Bohnenberger H, Reichardt SD, Lühder F, Walter L, Dressel R, Meijsing SH, Reichardt HM. Glucocorticoid resistance of allogeneic T cells alters the gene expression profile in the inflamed small intestine of mice suffering from acute graft-versus-host disease. J Steroid Biochem Mol Biol 2019; 195:105485. [PMID: 31561002 DOI: 10.1016/j.jsbmb.2019.105485] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Revised: 08/20/2019] [Accepted: 09/21/2019] [Indexed: 01/20/2023]
Abstract
Glucocorticoids (GCs) play an important role in controlling acute graft-versus-host disease (aGvHD), a frequent complication of allogeneic hematopoietic stem cell transplantation. The anti-inflammatory activity of GCs is mainly ascribed to the modulation of T cells and macrophages, for which reason a genetically induced GC resistance of either of these cell types causes aggravated aGvHD. Since only a few genes are currently known that are differentially regulated under these conditions, we analyzed the expression of 54 candidate genes in the inflamed small intestine of mice suffering from aGvHD when either allogeneic T cells or host myeloid cells were GC resistant using a microfluidic dynamic array platform for high-throughput quantitative PCR. The majority of genes categorized as cytokines (e.g. Il2, Il6), chemokines (e.g. Ccl2, Cxcl1), cell surface receptors (e.g. Fasl, Ctla4) and intracellular molecules (e.g. Dusp1, Arg1) were upregulated in mice transplanted with GC resistant allogeneic T cells. Moreover, the expression of several genes linked to energy metabolism (e.g. Glut1) was altered. Surprisingly, mice harboring GC resistant myeloid cells showed almost no changes in gene expression despite their fatal disease course after aGvHD induction. To identify additional genes in the inflamed small intestine that were affected by a GC resistance of allogeneic T cells, we performed an RNAseq analysis, which uncovered more than 500 differentially expressed transcripts (e.g. Cxcr6, Glut3, Otc, Aoc1, Il1r1, Sphk1) that were enriched for biological processes associated with inflammation and tissue disassembly. The changes in gene expression could be confirmed during full-blown disease but hardly any of them in the preclinical phase using high-throughput quantitative PCR. Further analysis of some of these genes revealed a highly selective expression pattern in T cells, intestinal epithelial cells and macrophages, which correlated with their regulation during disease progression. Collectively, we identified an altered gene expression profile caused by GC resistance of transplanted allogeneic T cells, which could help to define new targets for aGvHD therapy.
Collapse
Affiliation(s)
- Hu Li
- University Medical Center Göttingen, Institute for Cellular and Molecular Immunology, Humboldtallee 34, 37073 Göttingen, Germany
| | - Tina K Kaiser
- University Medical Center Göttingen, Institute for Cellular and Molecular Immunology, Humboldtallee 34, 37073 Göttingen, Germany
| | - Marina Borschiwer
- Max Planck Institute for Molecular Genetics, Ihnestraße 63, 14195 Berlin, Germany
| | - Hanibal Bohnenberger
- University Medical Center Göttingen, Institute for Pathology, Robert-Koch-Straße 40, 37075 Göttingen, Germany
| | - Sybille D Reichardt
- University Medical Center Göttingen, Institute for Cellular and Molecular Immunology, Humboldtallee 34, 37073 Göttingen, Germany
| | - Fred Lühder
- University Medical Center Göttingen, Institute for Neuroimmunology and Multiple Sclerosis Research, von-Siebold-Straße 3a, 37075 Göttingen, Germany
| | - Lutz Walter
- German Primate Center, Leibniz Institute for Primate Research, Primate Genetics Laboratory, Kellnerweg 4, 37077 Göttingen, Germany
| | - Ralf Dressel
- University Medical Center Göttingen, Institute for Cellular and Molecular Immunology, Humboldtallee 34, 37073 Göttingen, Germany
| | | | - Holger M Reichardt
- University Medical Center Göttingen, Institute for Cellular and Molecular Immunology, Humboldtallee 34, 37073 Göttingen, Germany.
| |
Collapse
|
7
|
Salvianolic acid B ameliorates liver injury in a murine aGvHD model by decreasing inflammatory responses via upregulation of HO-1. Transpl Immunol 2019; 55:101203. [PMID: 30904623 DOI: 10.1016/j.trim.2019.03.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Revised: 03/14/2019] [Accepted: 03/19/2019] [Indexed: 01/10/2023]
Abstract
Acute graft-versus-host disease (aGvHD) remains lethal, even after allogeneic hematopoietic stem cell transplantation. Inflammatory responses play an important role in aGvHD. Salvianolic acid B (Sal B) has been widely reported to have a major effect on the anti-inflammatory response, but these effects in an aGvHD model have never been reported. B6 donor splenocytes were transplanted into unirradiated BDF1 recipients and liver and serum were collected on day 14 after transplantation with or without Sal B administration. We measured the expression of pro-inflammatory cytokines and chemokines and other manifestations in aGvHD mice after Sal B treatment. Sal B ameliorated liver injury in aGvHD and promoted survival in mice. Sal B treatment resulted in decreased expression of pro-inflammatory cytokines and chemokines whose expressions in liver are normally elevated by aGvHD. Furthermore, Sal B treatment also enhanced PGC-1α expression in liver tissue and HO-1 expression in nonparenchymal cells. In addition, HO-1 inhibitor abrogated the improvement of survival rate of mice with aGvHD. These results indicated that the protective effect of Sal B relies on suppressing the inflammatory response phase in the aGvHD model, presumably by inducing HO-1. Taken together our data showed that Sal B ameliorates liver injury in aGvHD by decreasing inflammatory responses via upregulation of HO-1. It may provide a novel way to deal with this disease.
Collapse
|
8
|
Yu M, Wang J, Fang Q, Liu P, Chen S, Zhe N, Lin X, Zhang Y, Zhao J, Zhou Z. High expression of heme oxygenase-1 in target organs may attenuate acute graft-versus-host disease through regulation of immune balance of TH17/Treg. Transpl Immunol 2016; 37:10-17. [PMID: 27168057 DOI: 10.1016/j.trim.2016.05.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2016] [Revised: 05/02/2016] [Accepted: 05/03/2016] [Indexed: 12/31/2022]
Abstract
The high incidence of acute graft-versus-host disease (aGVHD) is a serious complication of allogeneic hematopoietic stem cell transplantation (allo-HSCT). Grades III and IV aGVHD are the leading causes of death in allo-HSCT recipients. Heme oxygenase-1(HO-1) has anti-inflammatory and immune-regulatory functions. In this study, we evaluated the none GVHD and grade I-IV patients samples which were collected at the first re-examination after successful allo-HSCT, we found that expressions of HO-1 mRNA in the bone marrow and peripheral blood mononuclear cells of allo-HSCT recipients who had subsequent non-GVHD and grade I aGVHD were significantly higher than those in patients with Grade III-IV aGVHD. We then demonstrated that enhanced expression of HO-1 in target organs by infusing HO-1-gene-modified Mesenchymal stem cells (MSCs) alleviated the clinical and histopathological severity of aGVHD in experimental mice. Flow cytometry revealed a higher expression of Treg cells and a lower expression of TH17 cells in splenic and lymph node tissues of mice with enhanced HO-1 expression, as compared to that in the aGVHD mice. This was further substantiated by lower expression levels of ROR-Υt and IL-17A mRNA, and higher levels of Foxp3 mRNA in the splenic tissue of mice with enhanced HO-1 expression. Our results indicate that high expression of HO-1 may reduce the severity of aGVHD by regulation of the TH17/Treg balance.
Collapse
Affiliation(s)
- Meisheng Yu
- Graduate School, Guizhou Medical University, PR China
| | - Jishi Wang
- Guizhou Province Laboratory of Hematopoietic Stem Cell Transplantation Centre, PR China; Department of Hematology, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou 550004, PR China.
| | - Qin Fang
- Department of Pharmacy, Affiliated Baiyun Hospital of Guizhou Medical University, Guiyang, Guizhou 550058, PR China
| | - Ping Liu
- Graduate School, Guizhou Medical University, PR China
| | - Shuya Chen
- Graduate School, Guizhou Medical University, PR China
| | - Nana Zhe
- Graduate School, Guizhou Medical University, PR China
| | - Xiaojing Lin
- Graduate School, Guizhou Medical University, PR China
| | - Yaming Zhang
- Guizhou Province Laboratory of Hematopoietic Stem Cell Transplantation Centre, PR China
| | - Jiangyuan Zhao
- Guizhou Province Laboratory of Hematopoietic Stem Cell Transplantation Centre, PR China
| | - Zhen Zhou
- Department of Pharmacy, Affiliated Baiyun Hospital of Guizhou Medical University, Guiyang, Guizhou 550058, PR China
| |
Collapse
|
9
|
Kartha RV, Zhou J, Basso L, Schröder H, Orchard PJ, Cloyd J. Mechanisms of Antioxidant Induction with High-Dose N-Acetylcysteine in Childhood Cerebral Adrenoleukodystrophy. CNS Drugs 2015; 29:1041-7. [PMID: 26670322 DOI: 10.1007/s40263-015-0300-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
BACKGROUND Childhood cerebral adrenoleukodystrophy (CCALD), a progressive demyelinating disease affecting school-aged boys, causes death within a few years. Oxidative stress is an important contributing factor. N-acetylcysteine (NAC; 280 mg/kg/day) added as adjunctive therapy to reduced-intensity hematopoietic cell transplantation (HCT) improves survival in advanced cases. However, the mechanisms underlying the benefits of NAC are unclear. OBJECTIVE The aim of this study was to understand the mechanism of action of NAC in the setting of HCT in CCALD. METHODS Immunoassays were carried out to determine changes in heme oxygenase-1 (HO-1) and ferritin expression in plasma samples collected from boys with CCALD at three different timepoints during the course of transplantation. In addition, the induction of HO-1 was also confirmed in normal fibroblasts following incubation with 10-100 µmol/L NAC for 4 h. RESULTS Following NAC therapy we observed an increase in expression of the antioxidants HO-1 (~4-fold) and its effector ferritin (~160-fold) in patient samples as compared with baseline. We also observed that NAC exposure significantly increased HO-1 expression in fibroblasts. CONCLUSION Our data suggest that HO-1 is a possible target protein of NAC and a mediator of its cytoprotective effects in these patients.
Collapse
Affiliation(s)
- Reena V Kartha
- Department of Experimental and Clinical Pharmacology, Center for Orphan Drug Research, University of Minnesota, Minneapolis, MN, USA. .,University of Minnesota, Room 4-214, McGuire Translational Research Facility, 2001-6th Street SE, Minneapolis, MN, 55455, USA.
| | - Jie Zhou
- Department of Experimental and Clinical Pharmacology, Center for Orphan Drug Research, University of Minnesota, Minneapolis, MN, USA
| | - Lisa Basso
- Division of Pediatric Blood and Marrow Transplantation, University of Minnesota, Minneapolis, MN, USA
| | - Henning Schröder
- Department of Pharmaceutics, University of Minnesota, Minneapolis, MN, USA
| | - Paul J Orchard
- Division of Pediatric Blood and Marrow Transplantation, University of Minnesota, Minneapolis, MN, USA
| | - James Cloyd
- Department of Experimental and Clinical Pharmacology, Center for Orphan Drug Research, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
10
|
Patel SR, Copland IB, Garcia MA, Metz R, Galipeau J. Human mesenchymal stromal cells suppress T-cell proliferation independent of heme oxygenase-1. Cytotherapy 2015; 17:382-91. [PMID: 25595329 DOI: 10.1016/j.jcyt.2014.11.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2014] [Revised: 11/17/2014] [Accepted: 11/27/2014] [Indexed: 01/14/2023]
Abstract
Mesenchymal stromal cells deploy immune suppressive properties amenable for use as cell therapy for inflammatory disorders. It is now recognized that mesenchymal stromal cells necessitate priming with an inflammatory milieu, in particular interferon-γ, to exert augmented immunosuppressive effects. It has been recently suggested that the heme-catabolizing enzyme heme oxygenase-1 is an essential component of the mesenchymal stromal cell-driven immune suppressive response. Because mesenchymal stromal cells upregulate indoleamine 2,3-dioxygenase expression on interferon-γ priming and indoleamine 2,3-dioxygenase requires heme as a cofactor for optimal catabolic function, we investigated the potential antagonism of heme oxygenase-1 activity on indoleamine 2, 3-dioxygenase and the impact on mesenchymal stromal cell immune plasticity. We herein sought to evaluate the molecular genetic effect of cytokine priming on human mesenchymal stromal cell heme oxygenase-1 expression and its functional role in differentially primed mesenchymal stromal cells. Contrary to previous reports, messenger RNA and protein analyses demonstrated that mesenchymal stromal cells derived from normal subjects (n = 6) do not express heme oxygenase-1 at steady state or after interferon-γ, tumor necrosis factor-α, and/or transforming growth factor-β priming. Pharmacological inhibition of heme oxygenase-1 with the use of tin protoporphyrin did not significantly abrogate the ability of mesenchymal stromal cells to suppress T-cell proliferation in vitro. Overall, these results unequivocally demonstrate that under steady state and after cytokine priming, human mesenchymal stromal cells immunoregulate T-cell proliferation independent of heme oxygenase-1.
Collapse
Affiliation(s)
- Seema R Patel
- Departments of Hematology & Medical Oncology, Emory University Winship Cancer Institute, Atlanta, Georgia, USA
| | - Ian B Copland
- Departments of Hematology & Medical Oncology, Emory University Winship Cancer Institute, Atlanta, Georgia, USA
| | - Marco A Garcia
- Departments of Hematology & Medical Oncology, Emory University Winship Cancer Institute, Atlanta, Georgia, USA
| | - Richard Metz
- NewLink Genetics Inc, Plymouth Meeting, Pennsylvania, USA
| | - Jacques Galipeau
- Departments of Hematology & Medical Oncology, Emory University Winship Cancer Institute, Atlanta, Georgia, USA; Department of Pediatrics, Emory University, Atlanta, Georgia, USA.
| |
Collapse
|
11
|
Tryptophan metabolite analog, N-(3,4-dimethoxycinnamonyl) anthranilic acid, ameliorates acute graft-versus-host disease through regulating T cell proliferation and polarization. Int Immunopharmacol 2013; 17:601-7. [DOI: 10.1016/j.intimp.2013.08.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2013] [Revised: 08/08/2013] [Accepted: 08/08/2013] [Indexed: 10/26/2022]
|
12
|
Li X, Xu F, Karoopongse E, Marcondes AM, Lee K, Kowdley KV, Miao CH, Trobridge GD, Campbell JS, Deeg HJ. Allogeneic transplantation, Fas signaling, and dysregulation of hepcidin. Biol Blood Marrow Transplant 2013; 19:1210-9. [PMID: 23707854 DOI: 10.1016/j.bbmt.2013.05.012] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2013] [Accepted: 05/14/2013] [Indexed: 12/21/2022]
Abstract
Hepatic iron overload is common in patients undergoing hematopoietic cell transplantation. We showed previously in a murine model that transplantation of allogeneic T cells induced iron deposition and down-regulation of hepcidin (Hamp) in hepatocytes. We hypothesized that hepatic injury was related to disrupted iron homeostasis triggered by the interaction of Fas-ligand, expressed on activated T cells, with Fas on hepatocytes. In the current study, we determined the effects of modified expression of the Flice inhibitory protein (FLIP long [FLIPL]), which interferes with Fas signaling, on the impact of Fas-initiated signals on the expression of IL-6 and Stat3 and their downstream target, Hamp. To exclude a possible contribution by other pathways, we used agonistic anti-Fas antibodies (rather than allogeneic T cells) to trigger Fas signaling. Inhibition of FLIPL by RNA interference resulted, as expected, not only in enhanced hepatocyte apoptosis in response to Fas signals, but also in decreased levels of IL-6, Stat3, and Hamp. In contrast, overexpression of FLIPL protected hepatocytes against agonistic anti-Fas antibody-mediated apoptosis and increased the levels of IL-6 and Stat3, thereby maintaining the expression of Hamp in an NF-κB-dependent fashion. In vivo overexpression of FLIPL in the liver via hydrodynamic transfection, similarly, interfered with Fas-initiated apoptosis and prevented down-regulation of IL-6, Stat3, and Hamp. These data indicate that Fas-dependent signals alter the regulation of iron homeostasis and suggest that signals initiated by Fas may contribute to peritransplantation iron accumulation.
Collapse
Affiliation(s)
- Xiang Li
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Donor-derived bone marrow transfusion produces mixed chimerism and promotes a Th2 shift in Th1/Th2 balance in rat heterotopic small bowel transplantation. Dig Liver Dis 2012; 44:988-94. [PMID: 22954489 DOI: 10.1016/j.dld.2012.08.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2012] [Revised: 07/13/2012] [Accepted: 08/05/2012] [Indexed: 12/11/2022]
Abstract
BACKGROUND AND AIM In this study, we investigated immunomodulatory effects of donor-derived bone marrow transfusion in rat heterotopic small bowel transplantation. METHODS Rat heterotopic segmental small bowel transplantation models (male Brown Norway to female Lewis) were established. The recipients were randomly divided into control group (pute small bowel transplantation), tacrolimus group (small bowel transplantation plus oral tacrolimus) and small bowel transplantation plus oral tacrolimus and intraportal infusion of donor-derived bone marrow cells group. We investigated the survival time, graft pathologic injuries and rejection grade by haematoxylin-eosin staining, serum IL-2 and IL-10 detection by enzyme labelled immunosorbent assay after small bowel transplantation. The recipients mixed chimerism were observed by detecting sex-determining region of Y chromosome gene in blood, liver, spleen and intestine by using real-time polymerase chain reaction and fluorescence in situ hybridization. RESULTS Bone marrow cells group showed a superior survival than the other groups, accompanied by milder pathologic injuries and lower rejection grade, decreasing serum IL-2 and increasing serum IL-10. The recipient chimerism rate in blood, liver, spleen and intestine in bone marrow cells group was significantly higher than the other groups. CONCLUSION Transfusion of donor-derived bone marrow cells via portal vein induces mixed chimerism in rats after small bowel transplantation, which may promote a Th2 shift in Th1/Th2 balance and facilitate the induction of immune tolerance.
Collapse
|
14
|
Li M, Vanella L, Zhang Y, Shi M, Takaki T, Shapiro JI, Ikehara S. Stem cell transplantation increases antioxidant effects in diabetic mice. Int J Biol Sci 2012; 8:1335-44. [PMID: 23139632 PMCID: PMC3492792 DOI: 10.7150/ijbs.4654] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2012] [Accepted: 09/13/2012] [Indexed: 12/14/2022] Open
Abstract
Intra bone marrow-bone marrow transplantation (IBM- BMT) + thymus transplantation (TT) has been shown to reduce the incidence of graft versus host disease (GVHD) and restore donor-derived T cell function. In addition, an increase in insulin sensitivity occurred in db/db mice after IBM-BMT+TT treatment. Heme oxygenase (HO)-1 is a stress inducible enzyme which exert antioxidant, antiapoptotic, and immune-modulating properties. We examined whether IBM-BMT+TT could modulate the expression of HO-1 in the kidneys of db/db mice. Six-week-old db/db mice with blood glucose levels higher than 250 mg/dl were treated with IBM-BMT+TT. Six weeks later, the db/db mice showed decreased body weight, blood glucose levels and insulin, and increased plasma adiponectin levels. The upregulation of HO-1 was associated with significantly (p<0.05) increased levels of peNOS and pAKT, but decreased levels of iNOS in the kidneys of db/db mice. Plasma creatinine levels also decreased (p<0.05), and the expression of type IV collagen was improved. Thus IBM-BMT+TT unregulated the expression of HO-1, peNOS and pAKT, while decreasing iNOS levels in the kidney of db/db mice. This was associated with an improvement in renal function.
Collapse
Affiliation(s)
- Ming Li
- Department of Stem Cell Disorders, Kansai Medical University, Moriguchi City, Osaka, Japan
| | | | | | | | | | | | | |
Collapse
|
15
|
The impact of inflammatory licensing on heme oxygenase-1-mediated induction of regulatory T cells by human mesenchymal stem cells. Blood 2011; 117:4826-35. [PMID: 21389316 DOI: 10.1182/blood-2010-12-324038] [Citation(s) in RCA: 173] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are characterized by their manifold immunomodulatory and regenerative properties. The stress-responsive, cytoprotective, and immunoregulatory molecule heme oxygenase-1 (HO-1) was recently identified as a key contributor for MSC-mediated suppression of alloactivated T cells. As HO-1 has also been implicated in the induction of regulatory T cells (Tregs), we sought to examine its impact on MSC-driven promotion of Tregs. Human MSCs were shown to induce, in a HO-1-dependent fashion, IL-10(+) Tr1 and transforming growth factor-β(+) Th3 Treg-subsets in allo- and T-cell receptor-activated lymphocytes. Because inflammatory stimuli modulate ("license") human MSCs, we were interested in whether an in vitro alloreactive micro-milieu within mixed lymphocyte reactions (MLRs) alters the HO-1 expression. We observed a substantial down-regulation of HO-1 facilitated by yet unidentified soluble factor(s) produced in an MLR, and most probably occurring at the level of its major transcription-factor NF-E2-related factor 2. Interestingly, HO-1 lost its impact regarding suppressiveness, Treg induction, and promotion of IL-10 production for MSCs, which were prelicensed in an MLR environment. Taken together, we show that HO-1 produced by human MSCs beyond its direct suppressive function promotes formation of Tr1 and Th3 Tregs and IL-10 production, functions, which are taken over by other molecules, among them COX-2, after an alloreactive priming.
Collapse
|
16
|
Sakai K, Kawata E, Ashihara E, Nakagawa Y, Yamauchi A, Yao H, Nagao R, Tanaka R, Yokota A, Takeuchi M, Hirai H, Kimura S, Hirashima M, Yoshimura N, Maekawa T. Galectin-9 ameliorates acute GVH disease through the induction of T-cell apoptosis. Eur J Immunol 2010; 41:67-75. [DOI: 10.1002/eji.200939931] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2010] [Revised: 09/10/2010] [Accepted: 10/29/2010] [Indexed: 01/27/2023]
|
17
|
Auletta JJ, Cooke KR, Solchaga LA, Deans RJ, van't Hof W. Regenerative stromal cell therapy in allogeneic hematopoietic stem cell transplantation: current impact and future directions. Biol Blood Marrow Transplant 2009; 16:891-906. [PMID: 20018250 DOI: 10.1016/j.bbmt.2009.12.005] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2009] [Accepted: 12/03/2009] [Indexed: 02/07/2023]
Abstract
Regenerative stromal cell therapy (RSCT) has the potential to become a novel therapy for preventing and treating acute graft-versus-host disease (GVHD) in the allogeneic hematopoietic stem cell transplant (HSCT) recipient. However, enthusiasm for using RSCT in allogeneic HSCT has been tempered by limited clinical data and poorly defined in vivo mechanisms of action. As a result, the full clinical potential of RSCT in supporting hematopoietic reconstitution and as treatment for GVHD remains to be determined. This manuscript reviews the immunomodulatory activity of regenerative stromal cells in preclinical models of allogeneic HSCT, and emphasizes an emerging literature suggesting that microenvironment influences RSC activation and function. Understanding this key finding may ultimately define the proper niche for RSCT in allogeneic HSCT. In particular, mechanistic studies are needed to delineate the in vivo effects of RSCT in response to inflammation and injury associated with allogeneic HSCT, and to define the relevant sites of RSC interaction with immune cells in the transplant recipient. Furthermore, development of in vivo imaging technology to correlate biodistribution patterns, desired RSC effect, and clinical outcome will be crucial to establishing dose-response effects and minimal biologic dose thresholds needed to advance translational treatment strategies for complications like GVHD.
Collapse
|
18
|
Deeg HJ, Spaulding E, Shulman HM. Iron overload, hematopoietic cell transplantation, and graft-versus-host disease. Leuk Lymphoma 2009; 50:1566-72. [PMID: 19863335 PMCID: PMC2887728 DOI: 10.1080/10428190903144659] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Many patients who undergo hematopoietic cell transplantation (HCT) present with anemia and have received red blood cell transfusions before HCT. As a result, iron overload is frequent and appears to be particularly prominent in patients with myelodysplastic syndromes. There is evidence that peritransplant events contribute to further iron accumulation, although the mechanism that disrupts normal iron homeostasis remains to be determined. Recent studies suggest that iron overload, as determined by ferritin levels, a surrogate marker for iron, is a risk factor for increased non-relapse mortality after HCT. Iron overload is associated with an increased rate of infections, in particular with fungal organisms. Furthermore anecdotal data suggest that increased hepatic iron may mimic the clinical picture of (chronic) graft-versus-host-disease (GVHD). Whether excess iron contributes to GVHD and whether iron depletion, be it by phlebotomy or chelation, reduces the post-transplantation complication rate and improves transplant outcome is yet to be determined.
Collapse
Affiliation(s)
- H Joachim Deeg
- Fred Hutchinson Cancer Research Center, Seattle, WA 98109-1024, USA.
| | | | | |
Collapse
|
19
|
Ewing P, Wilke A, Eissner G, Holler E, Andreesen R, Gerbitz A. Expression of Heme Oxygenase-1 Protects Endothelial Cells from Irradiation-Induced Apoptosis. ACTA ACUST UNITED AC 2009; 12:113-9. [PMID: 16291514 DOI: 10.1080/10623320500189814] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
A common side effect of therapeutic use of ionizing irradiation is endothelial cell damage resulting in a variety of clinical complications. Thus, preservation of endothelial function after irradiation could potentially limit toxicity. Using the murine endothelioma cell line bEnd2 we show here that induction of heme oxygenase-1 (HO-1) by cobalt protoporphyrine IX (CoPP) inhibits irradiation-induced apoptosis. In contrast, HO-1 induction by tin protoporphyrine IX (SnPP), a HO-1 inhibitor, does not affect survival after irradiation. The protective effects of CoPP could be partially reversed by blockade of HO-1 function with SnPP after induction by CoPP. Vice versa, blockade of HO-1 function by SnPP was reversible using CoPP. Treatment with CoPP inhibited cytochrome c release induced by irradiation. These results demonstrate that HO-1 induction and activation prior to irradiation inhibits endothelial apoptosis and might be used for possible cell protection in vivo.
Collapse
Affiliation(s)
- P Ewing
- Department of Hematology/Oncology, University of Regensburg, Regensburg, Germany
| | | | | | | | | | | |
Collapse
|
20
|
Gerbitz A, Hillemanns P, Schmid C, Wilke A, Jayaraman R, Kolb HJ, Eissner G, Holler E. Influence of polymorphism within the heme oxygenase-I promoter on overall survival and transplantation-related mortality after allogeneic stem cell transplantation. Biol Blood Marrow Transplant 2008; 14:1180-1189. [PMID: 18804049 DOI: 10.1016/j.bbmt.2008.08.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2008] [Accepted: 08/04/2008] [Indexed: 10/21/2022]
Abstract
Aside from major and minor histocompatibility antigens, genetic polymorphisms of various donor and host genes have been found to be risk factors for graft-versus-host disease and transplantation-related mortality (TRM). The heme oxygenase I (HO-I) protein has been implicated in regulating inflammatory response and has been described as a "protective gene" in solid organ transplantation. In humans, the promoter region displays length polymorphism due to a variable number of GT repeats. Individuals exhibiting 29 or fewer GT repeats express higher levels of HO-I on cellular stress compared with individuals with 30 or more GT repeats. We retrospectively analyzed length polymorphisms of 92 donor-host pairs undergoing allogeneic stem cell transplantation. Our findings demonstrate that mainly donor polymorphism leading to high expression of HO-1 (<30 GT repeats) on stress signals is associated with reduced overall survival, and that TRM is significantly increased in this group. This reduction in survival was most prominent when unrelated donors were used. Polymorphisms of the recipient HO-1 genes did not influence posttransplantation outcomes. We conclude that HO-1 polymorphism represents a new genetic risk factor for TRM and overall survival.
Collapse
Affiliation(s)
- Armin Gerbitz
- Department of Hematology and Oncology, Charité Berlin, Campus Benjamin Franklin, Berlin, Germany; Clinical Cooperation Group on Hematopoietic Stem Cell Transplantation, National Research Center for Environment and Health, Munich, Germany.
| | - Patrick Hillemanns
- Department of Hematology and Oncology, Ludwig Maximilian University of Munich, Munich, Germany
| | | | - Andrea Wilke
- Department of Hematology and Oncology, Ludwig Maximilian University of Munich, Munich, Germany; Clinical Cooperation Group on Hematopoietic Stem Cell Transplantation, National Research Center for Environment and Health, Munich, Germany
| | - Rajshri Jayaraman
- European School of Management and Technology ESMT, Schlossplatz, Berlin, Germany
| | - Hans-Jochem Kolb
- Department of Hematology and Oncology, Ludwig Maximilian University of Munich, Munich, Germany; Clinical Cooperation Group on Hematopoietic Stem Cell Transplantation, National Research Center for Environment and Health, Munich, Germany
| | - Gunther Eissner
- Department of Hematology and Oncology, University of Regensburg, Regensburg, Germany
| | - Ernst Holler
- Department of Hematology and Oncology, University of Regensburg, Regensburg, Germany
| |
Collapse
|
21
|
Lesnikov V, Gorden N, Fausto N, Spaulding E, Campbell J, Shulman H, Fleming RE, Deeg HJ. Transferrin fails to provide protection against Fas-induced hepatic injury in mice with deletion of functional transferrin-receptor type 2. Apoptosis 2008; 13:1005-12. [PMID: 18561026 DOI: 10.1007/s10495-008-0233-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
We reported previously that Fas-induced hepatic failure in normal mice was attenuated or prevented by exogenous transferrin (Tf), particularly apoTf. Here we show in C57BL6J/129 mice with genetic inactivation of transferrin receptor 2 (TfR2(Y245X)), that Fas-induced hepatotoxicity (apoptosis; rise in plasma aspartate aminotransferase (AST) levels) was comparable to that in wild-type mice, but was not modified by pretreatment with Tf. Rises in plasma AST were preceded by a decline in serum iron levels. AST elevations and iron declines were more profound in female than in male mice. Female mice also showed higher baseline levels of Bcl-xL in hepatocytes, which declined significantly upon treatment with agonistic anti-Fas antibody. These data confirm the cytoprotective function of Tf, and show a novel property of TfR2. Both apoptotic Fas responses and cytoprotective effects of Tf were associated with significant shifts in plasma iron levels, which quantitatively differed between male and female mice.
Collapse
Affiliation(s)
- Vladimir Lesnikov
- Fred Hutchinson Cancer Research Center, 1100 Fairview Avenue North, D1-100, P.O. Box 19024, Seattle, WA 98109-1024, USA
| | | | | | | | | | | | | | | |
Collapse
|
22
|
Mashreghi MF, Klemz R, Knosalla IS, Gerstmayer B, Janssen U, Buelow R, Jozkowicz A, Dulak J, Volk HD, Kotsch K. Inhibition of Dendritic Cell Maturation and Function Is Independent of Heme Oxygenase 1 but Requires the Activation of STAT3. THE JOURNAL OF IMMUNOLOGY 2008; 180:7919-30. [DOI: 10.4049/jimmunol.180.12.7919] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
23
|
Batzlsperger CA, Achatz S, Spreng J, Riegger GAJ, Griese DP. Evidence for a Possible Inhibitory Interaction between the HO-1/CO- and Akt/NO-Pathways in Human Endothelial Cells. Cardiovasc Drugs Ther 2007; 21:347-55. [PMID: 17896171 DOI: 10.1007/s10557-007-6051-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
OBJECTIVE The protective properties of heme oxygenase 1 (HO-1) give reason to study this mechanism as a potential therapeutic target for inflammatory and cardiovascular diseases. Recent evidence suggests a possible interaction between the HO-1/CO- and the protein kinase Akt/NO-pathway. This study was designed to examine the effects of continuous HO-1 overexpression in endothelial cells. METHODS Oncoretroviral vectors were constructed to achieve constitutive overexpression of HO-1, Akt, and green fluorescence protein in human umbilical vein endothelial cells. [(3)H]thymidine-incorporation and lipid-peroxidation were measured following exposure to heme and H(2)O(2). Expression of HO-1, Akt and its downstream-target endothelial NO-synthase were quantified by Western blot analysis. NO-synthase-activity was measured using the citrulline-conversion-assay. RESULTS HO-1-overexpression reduced proliferative rates and DNA-synthesis of HUVEC, but provided potent protection from oxidative stress induced by heme and H(2)O(2). Phosphorylated-Akt and eNOS was downregulated in HO-1-HUVEC. eNOS-activity was reduced in HO-1-HUVEC. Co-infection with the Akt-retrovirus restored proliferative rates and eNOS-expression and -activity. CONCLUSION Continuously elevated HO-1-activity protects EC from oxidative stress but inhibits Akt-mediated proliferation and eNOS-expression. This inhibitory feedback mechanism could be a limitation of HO-1 as a target for the treatment of vascular disease.
Collapse
Affiliation(s)
- Christian A Batzlsperger
- Department of Internal Medicine II, Regensburg University Medical Center, Franz Josef Strauss Allee 11, 93053 Regensburg, Germany
| | | | | | | | | |
Collapse
|
24
|
Trigona WL, Porter CM, Horvath-Arcidiacono JA, Majumdar AS, Bloom ET. Could heme-oxygenase-1 have a role in modulating the recipient immune response to embryonic stem cells? Antioxid Redox Signal 2007; 9:751-6. [PMID: 17511591 DOI: 10.1089/ars.2007.1602] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Pluripotent human embryonic stem cells (hESCs) may provide a potential source of cellular therapies, but as allogeneic cells may require evading the recipient's immune response. Using an NIH-registry hESC line, it was found that undifferentiated hESCs induce a reduced proliferative response compared to PBMC and demonstrate that this diminished response correlates with the activity of heme oxygenase-1 (HO-1). Inhibition of HO-1 significantly increases T cell proliferation against hESC, indicating the potential suppression of these cells during transplantation of allogeneic hESC. These data suggest the hypothesis that HO-1 provides a mechanism for protecting hESCs in vivo.
Collapse
Affiliation(s)
- Wendy L Trigona
- Gene Transfer and Immunogenicity Branch, Division of Cellular and Gene Therapies, Office of Cell, Gene, and Tissue Therapy, Center for Biologics Evaluation and Research, United States Food and Drug Administration, Bethesda, Maryland 20892, USA
| | | | | | | | | |
Collapse
|
25
|
Burman AC, Banovic T, Kuns RD, Clouston AD, Stanley AC, Morris ES, Rowe V, Bofinger H, Skoczylas R, Raffelt N, Fahy O, McColl SR, Engwerda CR, McDonald KPA, Hill GR. IFNgamma differentially controls the development of idiopathic pneumonia syndrome and GVHD of the gastrointestinal tract. Blood 2007; 110:1064-72. [PMID: 17449800 DOI: 10.1182/blood-2006-12-063982] [Citation(s) in RCA: 141] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Although proinflammatory cytokines are key mediators of tissue damage during graft-versus-host disease (GVHD), IFNgamma has previously been attributed with both protective and pathogenic effects. We have resolved this paradox by using wild-type (wt), IFNgamma(-/-), and IFNgammaR(-/-) mice as donors or recipients in well-described models of allogeneic stem cell transplantation (SCT). We show that donor-derived IFNgamma augments acute GVHD via direct effects on (1) the donor T cell to promote T helper 1 (Th1) differentiation and (2) the gastrointestinal (GI) tract to augment inflammatory cytokine generation. However, these detrimental effects are overwhelmed by a protective role of IFNgamma in preventing the development of idiopathic pneumonia syndrome (IPS). This is the result of direct effects on pulmonary parenchyma to prevent donor cell migration and expansion within the lung. Thus, IFNgamma is the key cytokine differentially controlling the development of IPS and gastrointestinal GVHD after allogeneic SCT.
Collapse
Affiliation(s)
- Angela C Burman
- Queensland Institute of Medical Research, 300 Herston Road, Herston, QLD 4006, Australia
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Kikuchi T, Naruse TK, Onizuka M, Li S, Kimura T, Oka A, Morishima Y, Kulski JK, Ichimiya S, Sato N, Inoko H. Mapping of susceptibility and protective loci for acute GVHD in unrelated HLA-matched bone marrow transplantation donors and recipients using 155 microsatellite markers on chromosome 22. Immunogenetics 2007; 59:99-108. [PMID: 17200870 DOI: 10.1007/s00251-006-0186-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2006] [Accepted: 11/21/2006] [Indexed: 02/02/2023]
Abstract
Despite matching donors and recipients for the human leukocyte antigens (HLAs) expressed by the major histocompatibility genomic region of the short arm of chromosome 6, several recipients still develop acute graft-versus-host disease (aGVHD) after bone marrow transplantation (BMT). This is possibly due to non-HLA gene polymorphisms, such as minor histocompatibility antigens (mHas) and genes coding for cytokines. However, a detailed genetic background for aGVHD has not yet been established. To find novel susceptibility and/or protective loci for aGVHD, a whole genome-wide association study of donors and recipients needs to be performed. As the first step to such a study, we retrospectively analyzed polymorphisms of 155 microsatellite markers spread across the long arm of chromosome 22 in 70 pairs of HLA-matched unrelated BMT donors and recipients. We performed individual typing and then compared the markers' allele frequencies (1) between all the aGVHD (grades III and IV GVHD) and GVHD-free (grade 0 GVHD) groups in donors and recipients and (2) between the aGVHD and aGVHD-free groups in donor/recipient pairs that were matched and mismatched for the microsatellite marker's allele. Screening of the microsatellite markers revealed five loci with a significant difference between the aGVHD and GVHD-free groups and revealed eight loci on chromosome 22, where the microsatellite allele mismatched markers were associated with aGVHD. This screening analysis suggests that several aGVHD-associated susceptible and protective loci exist on chromosome 22, which may encompass novel gene regions that need to be elucidated for their role in aGVHD.
Collapse
Affiliation(s)
- Tomoki Kikuchi
- Department of Pathology, Sapporo Medical University School of Medicine, Hokkaido, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Yachie A. Heme oxygenase and its role in defense system ; Paradigm shift of anti-inflammatory therapy. ACTA ACUST UNITED AC 2007; 30:11-21. [PMID: 17332700 DOI: 10.2177/jsci.30.11] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Heme oxygenase (HO) plays a central role in heme metabolism. At the same time, it protects cells from injury evoked by various oxidative stresses. A detailed analysis of the first human case of HO-1 deficiency revealed that HO-1 is involved in the protection of multiple tissues and organs. It is particularly important that in vivo HO-1 production is localized to selected cell types, e.g. renal tubular epithelium, reflecting the fact that HO-1 plays particularly important protective roles in these cells. In addition to renal epithelial cells and tissue macrophages, a minor subpopulation of circulating monocytes produced low, but significant levels of HO-1 and the number of these monocytes increased during episodes of acute inflammatory illnesses, indicating that monocytes play significant roles in controlling inflammation. On the other hand, excessive level of HO-1 induced by HO-1 gene transfection led to paradoxical susceptibility of the cells to oxidative injury. These results indicated that HO-1 expression is carefully controlled in vivo with regard to its location and the magnitude. Furthermore, it has been recently shown that HO-1 is involved in the immune regulation mediated by regulatory T cells. From these findings, it seems feasible to meticulously induce HO-1 protein in vivo as a novel therapeutic intervention to control various forms of inflammatory disorders.
Collapse
Affiliation(s)
- Akihiro Yachie
- Department of Clinical Laboratory Science, Division of Health Sciences, Kanazawa University Graduate School of Medical Science
| |
Collapse
|
28
|
Paul G, Bataille F, Obermeier F, Bock J, Klebl F, Strauch U, Lochbaum D, Rümmele P, Farkas S, Schölmerich J, Fleck M, Rogler G, Herfarth H. Analysis of intestinal haem-oxygenase-1 (HO-1) in clinical and experimental colitis. Clin Exp Immunol 2005; 140:547-55. [PMID: 15932518 PMCID: PMC1809385 DOI: 10.1111/j.1365-2249.2005.02775.x] [Citation(s) in RCA: 102] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/24/2005] [Indexed: 12/12/2022] Open
Abstract
Haem-oxygenase-1 (HO-1) has been shown to exert anti-inflammatory, anti-apoptotic and anti-proliferative effects. We investigated HO-1 expression in patients with inflammatory bowel disease (IBD) and could demonstrate a scattered expression of HO-1 in the intestinal epithelium of severely inflamed colonic mucosa of patients with IBD compared to control specimens such as diverticulitis, suggesting dysregulated expression in IBD. To further analyse potential mechanisms of HO-1 induction in the intestine we employed an in vitro epithelial cell apoptosis model and an experimental colitis model. In vitro induction of HO-1 by the HO-1 inducer cobalt protoporphyrin (CoPP) resulted in a dose-dependent down-regulation of caspase-3 activation in HT-29 cells, indicating an anti-apoptotic function of HO-1 in the intestine. In vivo, preventive HO-1 induction by CoPP in acute dextran sodium sulphate (DSS)-induced colitis led to a significant down-regulation of colonic inflammation (P < 0.01) with a concomitant reduction in interferon (IFN)-gamma - but unaffected interleukin (IL)-10-secretion by isolated mesenteric lymph nodes (P < 0.01). Additionally, TUNEL staining of colonic sections demonstrated fewer apoptotic epithelial cells in the colon of CoPP treated animals. No beneficial effects were observed if HO-1 was induced by CoPP after the onset of acute colitis or in chronic DSS-induced colitis. In conclusion, the data suggest a protective role of HO-1 if it is induced before the onset of inflammation. However, as shown by the lack of effects in established acute or in chronic colitis, the induction of HO-1 may not be a promising approach for the treatment of IBD.
Collapse
Affiliation(s)
- G Paul
- Department of Internal Medicine I, University of Regensburg, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|