1
|
Anders HJ, Kitching AR, Leung N, Romagnani P. Glomerulonephritis: immunopathogenesis and immunotherapy. Nat Rev Immunol 2023; 23:453-471. [PMID: 36635359 PMCID: PMC9838307 DOI: 10.1038/s41577-022-00816-y] [Citation(s) in RCA: 23] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/23/2022] [Indexed: 01/14/2023]
Abstract
'Glomerulonephritis' (GN) is a term used to describe a group of heterogeneous immune-mediated disorders characterized by inflammation of the filtration units of the kidney (the glomeruli). These disorders are currently classified largely on the basis of histopathological lesion patterns, but these patterns do not align well with their diverse pathological mechanisms and hence do not inform optimal therapy. Instead, we propose grouping GN disorders into five categories according to their immunopathogenesis: infection-related GN, autoimmune GN, alloimmune GN, autoinflammatory GN and monoclonal gammopathy-related GN. This categorization can inform the appropriate treatment; for example, infection control for infection-related GN, suppression of adaptive immunity for autoimmune GN and alloimmune GN, inhibition of single cytokines or complement factors for autoinflammatory GN arising from inborn errors in innate immunity, and plasma cell clone-directed or B cell clone-directed therapy for monoclonal gammopathies. Here we present the immunopathogenesis of GN and immunotherapies in use and in development and discuss how an immunopathogenesis-based GN classification can focus research, and improve patient management and teaching.
Collapse
Affiliation(s)
- Hans-Joachim Anders
- Division of Nephrology, Department of Medicine IV, University Hospital, Ludwig Maximilian University Munich, Munich, Germany.
| | - A Richard Kitching
- Centre for Inflammatory Diseases, Monash University Department of Medicine, Monash Medical Centre, Clayton, VIC, Australia
- Department of Nephrology, Monash Health, Clayton, VIC, Australia
- Department of Paediatric Nephrology, Monash Health, Clayton, VIC, Australia
| | - Nelson Leung
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, USA
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
| | - Paola Romagnani
- Department of Experimental and Biomedical Sciences "Mario Serio", University of Florence, Florence, Italy
- Nephrology and Dialysis Unit, Meyer Children's Hospital IRCCS, Florence, Italy
| |
Collapse
|
2
|
Yang L, Wei S, Zhang J, Hu Q, Hu W, Cao M, Zhang L, Wang Y, Wang P, Wang K. Construction of a predictive model for immunotherapy efficacy in lung squamous cell carcinoma based on the degree of tumor-infiltrating immune cells and molecular typing. Lab Invest 2022; 20:364. [PMID: 35962453 PMCID: PMC9373274 DOI: 10.1186/s12967-022-03565-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Accepted: 08/02/2022] [Indexed: 12/20/2022]
Abstract
Background To construct a predictive model of immunotherapy efficacy for patients with lung squamous cell carcinoma (LUSC) based on the degree of tumor-infiltrating immune cells (TIIC) in the tumor microenvironment (TME). Methods The data of 501 patients with LUSC in the TCGA database were used as a training set, and grouped using non-negative matrix factorization (NMF) based on the degree of TIIC assessed by single-sample gene set enrichment analysis (GSEA). Two data sets (GSE126044 and GSE135222) were used as validation sets. Genes screened for modeling by least absolute shrinkage and selection operator (LASSO) regression and used to construct a model based on immunophenotyping score (IPTS). RNA extraction and qPCR were performed to validate the prognostic value of IPTS in our independent LUSC cohort. The receiver operating characteristic (ROC) curve was constructed to determine the predictive value of the immune efficacy. Kaplan–Meier survival curve analysis was performed to evaluate the prognostic predictive ability. Correlation analysis and enrichment analysis were used to explore the potential mechanism of IPTS molecular typing involved in predicting the immunotherapy efficacy for patients with LUSC. Results The training set was divided into a low immune cell infiltration type (C1) and a high immune cell infiltration type (C2) by NMF typing, and the IPTS molecular typing based on the 17-gene model could replace the results of the NMF typing. The area under the ROC curve (AUC) was 0.82. In both validation sets, the IPTS of patients who responded to immunotherapy were significantly higher than those who did not respond to immunotherapy (P = 0.0032 and P = 0.0451), whereas the AUC was 0.95 (95% CI = 1.00–0.84) and 0.77 (95% CI = 0.58–0.96), respectively. In our independent cohort, we validated its ability to predict the response to cancer immunotherapy, for the AUC was 0.88 (95% CI = 1.00–0.66). GSEA suggested that the high IPTS group was mainly involved in immune-related signaling pathways. Conclusions IPTS molecular typing based on the degree of TIIC in the TME could well predict the efficacy of immunotherapy in patients with LUSC with a certain prognostic value. Supplementary Information The online version contains supplementary material available at 10.1186/s12967-022-03565-7.
Collapse
Affiliation(s)
- Lingge Yang
- Department of Respiratory and Critical Care Medicine, The Fourth Affiliated Hospital, International Institutes of Medicine, Zhejiang University School of Medicine, Yiwu, China
| | - Shuli Wei
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Jingnan Zhang
- Department of Respiratory and Critical Care Medicine, The Fourth Affiliated Hospital, International Institutes of Medicine, Zhejiang University School of Medicine, Yiwu, China
| | - Qiongjie Hu
- Department of Respiratory and Critical Care Medicine, The Fourth Affiliated Hospital, International Institutes of Medicine, Zhejiang University School of Medicine, Yiwu, China
| | - Wansong Hu
- Department of Heart Center, The Fourth Affiliated Hospital, International Institutes of Medicine, Zhejiang University School of Medicine, Yiwu, China
| | - Mengqing Cao
- Department of Respiratory and Critical Care Medicine, The Fourth Affiliated Hospital, International Institutes of Medicine, Zhejiang University School of Medicine, Yiwu, China
| | - Long Zhang
- Department of Respiratory and Critical Care Medicine, The Fourth Affiliated Hospital, International Institutes of Medicine, Zhejiang University School of Medicine, Yiwu, China
| | - Yongfang Wang
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Pingli Wang
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China.
| | - Kai Wang
- Department of Respiratory and Critical Care Medicine, The Fourth Affiliated Hospital, International Institutes of Medicine, Zhejiang University School of Medicine, Yiwu, China.
| |
Collapse
|
3
|
Miyata M, Ichikawa K, Matsuki E, Watanabe M, Peltier D, Toubai T. Recent Advances of Acute Kidney Injury in Hematopoietic Cell Transplantation. Front Immunol 2022; 12:779881. [PMID: 35058924 PMCID: PMC8763685 DOI: 10.3389/fimmu.2021.779881] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Accepted: 12/02/2021] [Indexed: 12/30/2022] Open
Abstract
Acute kidney injury (AKI) is a common complication of allogeneic hematopoietic cell transplantation (allo-HCT) and is associated with non-relapse mortality (NRM) and quality of life (QOL). Multiple factors may contribute to AKI during allo-HCT and are often present at the same time making it difficult to determine the cause of AKI in each patient. Nephrotoxic drugs, infections, thrombotic microangiopathy (TMA), and sinusoidal obstruction syndrome (SOS) are well described causes of AKI during allo-HCT. Acute graft-versus-host disease (aGVHD) is a major complication of allo-HCT that mainly targets the intestines, liver, and skin. However, recent studies suggest aGVHD may also attack the kidney and contribute to AKI following allo-HCT. For example, severe aGVHD is associated with AKI, suggesting a link between the two. In addition, animal models have shown donor immune cell infiltration and increased expression of inflammatory cytokines in recipient kidneys after allo-HCT. Therefore, aGVHD may also target the kidney and contribute to AKI following allo-HCT. Herein, we describe the etiology, diagnosis, risk factors, pathophysiology, prevention, and treatment of renal injury after allo-HCT. In addition, we highlight emerging evidence that aGVHD may contribute to the development of AKI after allo-HCT.
Collapse
Affiliation(s)
- Masahiro Miyata
- Department of Cardiology, Pulmonology, and Nephrology, Faculty of Medicine, Yamagata University, Yamagata, Japan
| | - Kazunobu Ichikawa
- Department of Cardiology, Pulmonology, and Nephrology, Faculty of Medicine, Yamagata University, Yamagata, Japan
| | - Eri Matsuki
- Department of Cardiology, Pulmonology, and Nephrology, Faculty of Medicine, Yamagata University, Yamagata, Japan
| | - Masafumi Watanabe
- Department of Cardiology, Pulmonology, and Nephrology, Faculty of Medicine, Yamagata University, Yamagata, Japan
| | - Daniel Peltier
- Department of Pediatric Hematology/Oncology, University Michigan Medical School, Ann Arbor, MI, United States
| | - Tomomi Toubai
- Department of Internal Medicine III, Division of Hematology and Cell Therapy, Faculty of Medicine, Yamagata University, Yamagata, Japan
| |
Collapse
|
4
|
Andronesi A, Sorohan B, Burcea A, Lipan L, Stanescu C, Craciun O, Stefan L, Ranete A, Varady Z, Ungureanu O, Lupusoru G, Agrigoroaei G, Andronesi D, Iliuta L, Obrisca B, Tanase A. Incidence and Risk Factors for Acute Kidney Injury after Allogeneic Stem Cell Transplantation: A Prospective Study. Biomedicines 2022; 10:biomedicines10020262. [PMID: 35203472 PMCID: PMC8869231 DOI: 10.3390/biomedicines10020262] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2021] [Revised: 01/14/2022] [Accepted: 01/24/2022] [Indexed: 01/11/2023] Open
Abstract
(1) Background: Acute kidney injury (AKI) is a serious complication of hematopoietic stem cell transplantation (HSCT). (2) Methods: The aim was to identify the incidence, severity, and risk factors for AKI during the first 100 days after allo-HSCT; we performed a prospective observational study on 135 consecutive patients. (3) Results: The mean age was 38.3 ± 11.9 years (50.6% females), AKI developed in 93 patients (68.9%), the median time of appearance was 28 days, and the mean serum creatinine at the time of AKI was 1.8 ± 0.8 mg/dL. A total of 36 (38.7%) patients developed stage 1 AKI, 33 (35.5%) patients developed stage 2, and 24 (25.8%) patients developed stage 3; eight (8.6%) patients required temporary hemodialysis, and the mortality rate in these patients was 87.5%. Death was twice as frequent in the AKI subgroup, without statistical significance. Cyclosporine overdose (HR = 2.36, 95% CI: 1.45-3.85, p = 0.001), tacrolimus overdose (HR = 4.72, 95% CI: 2.22-10.01, p < 0.001), acute graft-versus-host disease (aGVHD) (HR = 1.96, 95% CI: 1.13-3.40, p = 0.01), and CRP level (HR = 1.009, 95% CI: 1.007-1.10, p < 0.001) were independent risk factors for AKI. Sepsis (HR = 5.37, 95% CI: 1.75-16.48, p = 0.003) and sinusoidal obstruction syndrome (HR = 5.10, 95% CI: 2.02-12.85, p = 0.001) were found as independent risk factors for AKI stage 3. (4) Conclusions: AKI occurs with high incidence and increased severity after allo-HSCT. Careful monitoring of calcineurin inhibitors and proper management of sepsis may reduce this risk.
Collapse
Affiliation(s)
- Andreea Andronesi
- Department of Nephrology, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania; (B.S.); (G.L.); (G.A.); (B.O.)
- Nephrology Department, Fundeni Clinical Institute, 022328 Bucharest, Romania; (A.B.); (C.S.); (O.U.)
- Correspondence: ; Tel.: +40-723-361-457
| | - Bogdan Sorohan
- Department of Nephrology, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania; (B.S.); (G.L.); (G.A.); (B.O.)
- Nephrology Department, Fundeni Clinical Institute, 022328 Bucharest, Romania; (A.B.); (C.S.); (O.U.)
| | - Andreea Burcea
- Nephrology Department, Fundeni Clinical Institute, 022328 Bucharest, Romania; (A.B.); (C.S.); (O.U.)
| | - Lavinia Lipan
- Bone Marrow Transplant Department, Fundeni Clinical Institute, 022328 Bucharest, Romania; (L.L.); (O.C.); (L.S.); (A.R.); (Z.V.); (A.T.)
| | - Cristina Stanescu
- Nephrology Department, Fundeni Clinical Institute, 022328 Bucharest, Romania; (A.B.); (C.S.); (O.U.)
| | - Oana Craciun
- Bone Marrow Transplant Department, Fundeni Clinical Institute, 022328 Bucharest, Romania; (L.L.); (O.C.); (L.S.); (A.R.); (Z.V.); (A.T.)
| | - Laura Stefan
- Bone Marrow Transplant Department, Fundeni Clinical Institute, 022328 Bucharest, Romania; (L.L.); (O.C.); (L.S.); (A.R.); (Z.V.); (A.T.)
| | - Adela Ranete
- Bone Marrow Transplant Department, Fundeni Clinical Institute, 022328 Bucharest, Romania; (L.L.); (O.C.); (L.S.); (A.R.); (Z.V.); (A.T.)
| | - Zsofia Varady
- Bone Marrow Transplant Department, Fundeni Clinical Institute, 022328 Bucharest, Romania; (L.L.); (O.C.); (L.S.); (A.R.); (Z.V.); (A.T.)
| | - Oana Ungureanu
- Nephrology Department, Fundeni Clinical Institute, 022328 Bucharest, Romania; (A.B.); (C.S.); (O.U.)
| | - Gabriela Lupusoru
- Department of Nephrology, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania; (B.S.); (G.L.); (G.A.); (B.O.)
- Nephrology Department, Fundeni Clinical Institute, 022328 Bucharest, Romania; (A.B.); (C.S.); (O.U.)
| | - Gabriela Agrigoroaei
- Department of Nephrology, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania; (B.S.); (G.L.); (G.A.); (B.O.)
| | - Danut Andronesi
- Department of General Surgery and Liver Transplant, Fundeni Clinical Institute, 022328 Bucharest, Romania;
| | - Luminita Iliuta
- Department of Biostatistics, Marketing and Medical Technology, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania;
| | - Bogdan Obrisca
- Department of Nephrology, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania; (B.S.); (G.L.); (G.A.); (B.O.)
- Nephrology Department, Fundeni Clinical Institute, 022328 Bucharest, Romania; (A.B.); (C.S.); (O.U.)
| | - Alina Tanase
- Bone Marrow Transplant Department, Fundeni Clinical Institute, 022328 Bucharest, Romania; (L.L.); (O.C.); (L.S.); (A.R.); (Z.V.); (A.T.)
| |
Collapse
|
5
|
Imaging Tolerance Induction in Neonatal Mice: Hierarchical Interplay Between Allogeneic Adult and Neonatal Immune Cells. Transplantation 2021; 105:1730-1746. [PMID: 33273316 DOI: 10.1097/tp.0000000000003566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND In Medawar's murine neonatal tolerance model, injection of adult semiallogeneic lymphohematopoietic cells (spleen cells [SC] and bone marrow cells [BMC]) tolerizes the neonatal immune system. An eventual clinical application would require fully allogeneic (allo) cells, yet little is known about the complex in vivo/in situ interplay between those cells and the nonconditioned neonatal immune system. METHODS To this end, labeled adult SC and BMC were injected into allogeneic neonates; interactions between donor and host cells were analyzed and modulated by systematic depletion/inactivation of specific donor and host immune effector cell types. RESULTS Consistent with effector cell compositions, allo-SC and allo-SC/BMC each induced lethal acute graft-versus-host disease, whereas allo-BMC alone did so infrequently. CD8 T cells from SC inoculum appeared naïve, while those of BMC were more memory-like. Age-dependent, cell-type dominance defined the interplay between adult donor cells and the neonatal host immune system such that if the dominant adult effector type was removed, then the equivalent neonatal one became dominant. Depletion of donor/host peripheral T cells protected against acute graft-versus-host disease and prolonged heart allograft survival; peripheral CD8 T-cell depletion together with CD4 T cell-costimulation blockade induced more robust tolerance. CONCLUSIONS This comprehensive study provides direct observation of the cellular interplay between allogeneic donor and host immune systems, adds to our previous work with semiallogeneic donor cells, and provides important insights for robust tolerance induction. Induction of transplant tolerance in neonates will likely require "crowd sourcing" of multiple tolerizing cell types and involve depletion of immune effector cells with costimulation blockade.
Collapse
|
6
|
Amin R, He R, Gupta D, Zheng W, Burmakin M, Mohammad DK, DePierre JW, Sadeghi B, Olauson H, Wernerson A, El-Andaloussi S, Hassan M, Abedi-Valugerdi M. The kidney injury caused by the onset of acute graft-versus-host disease is associated with down-regulation of αKlotho. Int Immunopharmacol 2020; 78:106042. [DOI: 10.1016/j.intimp.2019.106042] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Revised: 10/24/2019] [Accepted: 11/08/2019] [Indexed: 12/11/2022]
|
7
|
Jaguś D, Lis K, Niemczyk L, Basak GW. Kidney dysfunction after hematopoietic cell transplantation-Etiology, management, and perspectives. Hematol Oncol Stem Cell Ther 2018; 11:195-205. [PMID: 30076790 DOI: 10.1016/j.hemonc.2018.07.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2018] [Revised: 04/26/2018] [Accepted: 07/12/2018] [Indexed: 12/18/2022] Open
Abstract
Kidney dysfunction is a common complication of hematopoietic cell transplantation (HCT) with proven negative impact on early and long-term mortality. Causes of this complication are diverse, usually overlapping, and poorly understood. Therefore, management implicates multidirectional investigations and simultaneous treatment of suspected causes. The etiology is frequently unconfirmed due to a lack of specific markers and prevalence of contraindications to renal biopsy among HCT recipients. Herein, we provide a summary of etiology and propose an algorithm for evaluation of kidney injury after HCT. We also map out the most urgent areas for research that aim to identify patients at risk of severe renal injury and develop nephroprotective strategies.
Collapse
Affiliation(s)
- Dorota Jaguś
- Department of Nephrology, Dialysis and Internal Medicine, Medical University of Warsaw, Warsaw, Poland
| | - Karol Lis
- Department of Hematology, Oncology and Internal Medicine, Medical University of Warsaw, Warsaw, Poland
| | - Longin Niemczyk
- Department of Nephrology, Dialysis and Internal Medicine, Medical University of Warsaw, Warsaw, Poland
| | - Grzegorz W Basak
- Department of Hematology, Oncology and Internal Medicine, Medical University of Warsaw, Warsaw, Poland.
| |
Collapse
|
8
|
Gam R, Shah P, Crossland RE, Norden J, Dickinson AM, Dressel R. Genetic Association of Hematopoietic Stem Cell Transplantation Outcome beyond Histocompatibility Genes. Front Immunol 2017; 8:380. [PMID: 28421078 PMCID: PMC5377073 DOI: 10.3389/fimmu.2017.00380] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Accepted: 03/16/2017] [Indexed: 12/18/2022] Open
Abstract
The outcome of hematopoietic stem cell transplantation (HSCT) is controlled by genetic factors among which the leukocyte antigen human leukocyte antigen (HLA) matching is most important. In addition, minor histocompatibility antigens and non-HLA gene polymorphisms in genes controlling immune responses are known to contribute to the risks associated with HSCT. Besides single-nucleotide polymorphisms (SNPs) in protein coding genes, SNPs in regulatory elements such as microRNAs (miRNAs) contribute to these genetic risks. However, genetic risks require for their realization the expression of the respective gene or miRNA. Thus, gene and miRNA expression studies may help to identify genes and SNPs that indeed affect the outcome of HSCT. In this review, we summarize gene expression profiling studies that were performed in recent years in both patients and animal models to identify genes regulated during HSCT. We discuss SNP–mRNA–miRNA regulatory networks and their contribution to the risks associated with HSCT in specific examples, including forkheadbox protein 3 and regulatory T cells, the role of the miR-155 and miR-146a regulatory network for graft-versus-host disease, and the function of MICA and its receptor NKG2D for the outcome of HSCT. These examples demonstrate how SNPs affect expression or function of proteins that modulate the alloimmune response and influence the outcome of HSCT. Specific miRNAs targeting these genes and directly affecting expression of mRNAs are identified. It might be valuable in the future to determine SNPs and to analyze miRNA and mRNA expression in parallel in cohorts of HSCT patients to further elucidate genetic risks of HSCT.
Collapse
Affiliation(s)
- Rihab Gam
- Hematological Sciences, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, UK
| | - Pranali Shah
- Institute of Cellular and Molecular Immunology, University Medical Centre Göttingen, Göttingen, Germany
| | - Rachel E Crossland
- Hematological Sciences, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, UK
| | - Jean Norden
- Hematological Sciences, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, UK
| | - Anne M Dickinson
- Hematological Sciences, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, UK
| | - Ralf Dressel
- Institute of Cellular and Molecular Immunology, University Medical Centre Göttingen, Göttingen, Germany
| |
Collapse
|
9
|
Boieri M, Shah P, Jalapothu D, Zaitseva O, Walter L, Rolstad B, Naper C, Dressel R, Inngjerdingen M. Rat acute GvHD is Th1 driven and characterized by predominant donor CD4 + T-cell infiltration of skin and gut. Exp Hematol 2017; 50:33-45.e3. [PMID: 28238806 DOI: 10.1016/j.exphem.2017.02.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Revised: 01/05/2017] [Accepted: 02/13/2017] [Indexed: 01/07/2023]
Abstract
Acute graft-versus-host disease (aGvHD) remains a significant hurdle to successful treatment of many hematological disorders. The disease is caused by infiltration of alloactivated donor T cells primarily into the gastrointestinal tract and skin. Although cytotoxic T cells mediate direct cellular damage, T helper (Th) cells differentially secrete immunoregulatory cytokines. aGvHD is thought to be initiated primarily by Th1 cells but a consensus is still lacking regarding the role of Th2 and Th17 cells. The aim of this study was to determine the contribution of distinct T-cell subsets to aGvHD in the rat. aGvHD was induced by transplanting irradiated rats with T-cell-depleted major histocompatibility complex-mismatched bone marrow, followed 2 weeks later by donor lymphocyte infusion. Near complete donor T-cell chimerism was achieved in the blood and lymphatic tissues, in contrast to mixed chimerism in the skin and gut. Skin and gut donor T cells were predominantly CD4+, in contrast to T cells in the blood and lymphatic tissues. Genes associated with Th1 cells were upregulated in gut, liver, lung, and skin tissues affected by aGvHD. Increased serum levels of CXCL10 and IL-18 preceded symptoms of aGvHD, accompanied by increased responsiveness to CXCL10 by blood CD4+ T cells. No changes in the expression of Th2- or Th17-associated genes were observed, indicating that aGvHD in this rat model is mainly Th1 driven. The rat model of aGvHD could be instrumental for further investigations of donor T-cell subsets in the skin and gut and for exploring therapeutic options to ameliorate symptoms of aGvHD.
Collapse
Affiliation(s)
- Margherita Boieri
- Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway; Department of Immunology, Oslo University Hospital - Rikshospitalet, Oslo, Norway
| | - Pranali Shah
- Institute of Cellular and Molecular Immunology, University Medical Center Göttingen, Göttingen, Germany
| | - Dasaradha Jalapothu
- Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Olena Zaitseva
- Primate Genetics Laboratory, German Primate Center, Göttingen, Germany
| | - Lutz Walter
- Primate Genetics Laboratory, German Primate Center, Göttingen, Germany
| | - Bent Rolstad
- Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Christian Naper
- Department of Immunology, Oslo University Hospital - Rikshospitalet, Oslo, Norway
| | - Ralf Dressel
- Institute of Cellular and Molecular Immunology, University Medical Center Göttingen, Göttingen, Germany
| | - Marit Inngjerdingen
- Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway; Department of Immunology, Oslo University Hospital - Rikshospitalet, Oslo, Norway.
| |
Collapse
|
10
|
Conditioning with Fludarabine-Busulfan versus Busulfan-Cyclophosphamide Is Associated with Lower aGVHD and Higher Survival but More Extensive and Long Standing Bone Marrow Damage. BIOMED RESEARCH INTERNATIONAL 2016; 2016:3071214. [PMID: 27843940 PMCID: PMC5098055 DOI: 10.1155/2016/3071214] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Revised: 08/15/2016] [Accepted: 09/18/2016] [Indexed: 11/18/2022]
Abstract
Acute graft-versus-host disease (aGVHD) is a major complication of allogeneic hematopoietic stem cell transplantation (allo-HSCT) and a major cause of nonrelapse mortality after allo-HSCT. A conditioning regimen plays a pivotal role in the development of aGVHD. To provide a platform for studying aGVHD and evaluating the impact of different conditioning regimens, we established a murine aGVHD model that simulates the clinical situation and can be conditioned with Busulfan-Cyclophosphamide (Bu-Cy) and Fludarabine-Busulfan (Flu-Bu). In our study, BALB/c mice were conditioned with Bu-Cy or Flu-Bu and transplanted with 2 × 107 bone marrow cells and 2 × 107 splenocytes from either allogeneic (C57BL/6) or syngeneic (BALB/c) donors. The allogeneic recipients conditioned with Bu-Cy had shorter survivals (P < 0.05), more severe clinical manifestations, and higher hepatic and intestinal pathology scores, associated with increased INF-γ expression and diminished IL-4 expression in serum, compared to allogeneic recipients conditioned with Flu-Bu. Moreover, higher donor-derived T-cell infiltration and severely impaired B-cell development were seen in the bone marrow of mice, exhibiting aGVHD and conditioned with Flu-Bu. Our study showed that the conditioning regimen with Bu-Cy resulted in more severe aGVHD while the Flu-Bu regimen was associated with more extensive and long standing bone marrow damage.
Collapse
|
11
|
Affiliation(s)
- Sangeeta Hingorani
- From the Department of Pediatrics, Division of Nephrology, University of Washington School of Medicine, and the Clinical Research Division, Fred Hutchinson Cancer Research Center - both in Seattle
| |
Collapse
|
12
|
Lopes JA, Jorge S, Neves M. Acute kidney injury in HCT: an update. Bone Marrow Transplant 2016; 51:755-62. [DOI: 10.1038/bmt.2015.357] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Revised: 11/24/2015] [Accepted: 12/11/2015] [Indexed: 01/02/2023]
|
13
|
Jodele S, Laskin BL, Dandoy CE, Myers KC, El-Bietar J, Davies SM, Goebel J, Dixon BP. A new paradigm: Diagnosis and management of HSCT-associated thrombotic microangiopathy as multi-system endothelial injury. Blood Rev 2014; 29:191-204. [PMID: 25483393 DOI: 10.1016/j.blre.2014.11.001] [Citation(s) in RCA: 206] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2014] [Revised: 10/18/2014] [Accepted: 11/20/2014] [Indexed: 01/03/2023]
Abstract
Hematopoietic stem cell transplantation (HSCT)-associated thrombotic microangiopathy (TA-TMA) is now a well-recognized and potentially severe complication of HSCT that carries a high risk of death. In those who survive, TA-TMA may be associated with long-term morbidity and chronic organ injury. Recently, there have been new insights into the incidence, pathophysiology, and management of TA-TMA. Specifically, TA-TMA can manifest as a multi-system disease occurring after various triggers of small vessel endothelial injury, leading to subsequent tissue damage in different organs. While the kidney is most commonly affected, TA-TMA involving organs such as the lung, bowel, heart, and brain is now known to have specific clinical presentations. We now review the most up-to-date research on TA-TMA, focusing on the pathogenesis of endothelial injury, the diagnosis of TA-TMA affecting the kidney and other organs, and new clinical approaches to the management of this complication after HSCT.
Collapse
Affiliation(s)
- Sonata Jodele
- Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children's Hospital Medical Center, USA.
| | - Benjamin L Laskin
- Division of Nephrology, The Children's Hospital of Philadelphia, USA
| | - Christopher E Dandoy
- Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children's Hospital Medical Center, USA
| | - Kasiani C Myers
- Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children's Hospital Medical Center, USA
| | - Javier El-Bietar
- Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children's Hospital Medical Center, USA
| | - Stella M Davies
- Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children's Hospital Medical Center, USA
| | - Jens Goebel
- Division of Nephrology and Hypertension, Cincinnati Children's Hospital Medical Center, USA
| | - Bradley P Dixon
- Division of Nephrology and Hypertension, Cincinnati Children's Hospital Medical Center, USA
| |
Collapse
|
14
|
Hingorani S, Finn LS, Pao E, Lawler R, Schoch G, McDonald GB, Najafian B, Sandmaier B, Gooley T. Urinary elafin and kidney injury in hematopoietic cell transplant recipients. Clin J Am Soc Nephrol 2014; 10:12-20. [PMID: 25388519 DOI: 10.2215/cjn.01840214] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
BACKGROUND AND OBJECTIVES Graft-versus-host disease (GVHD) is associated with kidney injury after hematopoietic cell transplantation (HCT). Because plasma elafin levels correlate with skin GVHD, this study examined urinary elafin as a potential marker of renal inflammation and injury. DESIGN, SETTING, PARTICIPANTS, & MEASUREMENTS Urine was collected prospectively on 205 patients undergoing their first HCT from 2003 to 2010. Collections were done at baseline, weekly through day 100, and monthly through year 1 to measure elafin and urine albumin-to-creatinine ratio (ACR). Associations between urinary elafin levels and microalbuminuria, macroalbuminuria, AKI and CKD, and mortality were examined using Cox proportional hazards or linear regression models. Available kidney biopsy specimens were processed for immunohistochemistry. RESULTS Mean urinary elafin levels to day 100 were higher in patients with micro- or macroalbuminuria (adjusted mean difference, 529 pg/ml; P=0.03) at day 100 than in those with a normal ACR (adjusted mean difference, 1295 pg/ml; P<0.001). Mean urinary elafin levels were higher in patients with AKI compared with patients without AKI (adjusted mean difference, 558 pg/ml; P<0.01). The average urinary elafin levels within the first 100 days after HCT were higher in patients who developed CKD at 1 year than in patients without CKD (adjusted mean difference, 894 pg/ml; P=0.002). Among allogeneic recipients, a higher proportion of patients with micro- or macroalbuminuria at day 100 also had grade II-IV acute GVHD (80% and 86%, respectively) compared with patients with a normal ACR (58%; global P<0.01). Each increase in elafin of 500 pg/ml resulted in a 10% increase in risk of persistent macroalbuminuria (hazard ratio, 1.10; 95% confidence interval [95% CI], 1.06 to 1.13; P<0.001) and a 7% increase in the risk of overall mortality (95% CI, 1.02 to 1.13, P<0.01). Renal biopsy specimens from a separate cohort of HCT survivors demonstrated elafin staining in distal and collecting duct tubules. CONCLUSION Higher urinary elafin levels are associated with an increased risk of micro- and macroalbuminuria, AKI and CKD, and death after HCT.
Collapse
Affiliation(s)
- Sangeeta Hingorani
- Departments of Pediatrics, Gastroenterology, and Pathology, University of Washington, Seattle, Washington; Seattle Children's Hospital, Seattle, Washington; and Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Laura S Finn
- Departments of Pediatrics, Gastroenterology, and Pathology, University of Washington, Seattle, Washington; Seattle Children's Hospital, Seattle, Washington; and
| | - Emily Pao
- Seattle Children's Hospital, Seattle, Washington; and
| | - Rick Lawler
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Gary Schoch
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - George B McDonald
- Departments of Pediatrics, Gastroenterology, and Pathology, University of Washington, Seattle, Washington; Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Behzad Najafian
- Departments of Pediatrics, Gastroenterology, and Pathology, University of Washington, Seattle, Washington
| | - Brenda Sandmaier
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Ted Gooley
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| |
Collapse
|
15
|
Al-Chaqmaqchi H, Sadeghi B, Abedi-Valugerdi M, Al-Hashmi S, Fares M, Kuiper R, Lundahl J, Hassan M, Moshfegh A. The role of programmed cell death ligand-1 (PD-L1/CD274) in the development of graft versus host disease. PLoS One 2013; 8:e60367. [PMID: 23593203 PMCID: PMC3617218 DOI: 10.1371/journal.pone.0060367] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2012] [Accepted: 02/26/2013] [Indexed: 11/18/2022] Open
Abstract
Programmed cell death ligand-1 (PD-L1/CD274) is an immunomodulatory molecule involved in cancer and complications of bone marrow transplantation, such as graft rejection and graft-versus-host disease. The present study was designed to assess the dynamic expression of this molecule after hematopoietic stem cell transplantation in relation to acute graft-versus-host disease. Female BALB/c mice were conditioned with busulfan and cyclophosphamide and transplanted with either syngeneic or allogeneic (male C57BL/6 mice) bone marrow and splenic cells. The expression of PD-L1 was evaluated at different time points employing qPCR, western blot and immunohistochemistry. Allogeneic- but not syngeneic-transplanted animals exhibited a marked up-regulation of PD-L1 expression in the muscle and kidney, but not the liver, at days 5 and 7 post transplantation. In mice transplanted with allogeneic bone marrow cells, the enhanced expression of PD-L1 was associated with high serum levels of IFNγ and TNFα at corresponding intervals. Our findings demonstrate that PD-L1 is differently induced and expressed after allogeneic transplantation than it is after syngeneic transplantation, and that it is in favor of target rather than non-target organs at the early stages of acute graft-versus-host disease. This is the first study to correlate the dynamics of PD-L1 at the gene-, protein- and activity levels with the early development of acute graft-versus-host disease. Our results suggest that the higher expression of PD-L1 in the muscle and kidney (non-target tissues) plays a protective role in skeletal muscle during acute graft-versus-host disease.
Collapse
Affiliation(s)
- Heevy Al-Chaqmaqchi
- Department of Laboratory Medicine, Division of Clinical Research Center, Experimental Cancer Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Behnam Sadeghi
- Department of Laboratory Medicine, Division of Clinical Research Center, Experimental Cancer Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Manuchehr Abedi-Valugerdi
- Department of Laboratory Medicine, Division of Clinical Research Center, Experimental Cancer Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Sulaiman Al-Hashmi
- Department of Laboratory Medicine, Division of Clinical Research Center, Experimental Cancer Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Mona Fares
- Department of Laboratory Medicine, Division of Clinical Research Center, Experimental Cancer Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Raoul Kuiper
- Department of Laboratory Medicine, The Unite of Morphologic Phenotype Analysis, Karolinska Institutet and Karolinska University Hospital-Huddinge, Stockholm, Sweden
| | - Joachim Lundahl
- Department of Clinical Immunology and Transfusion Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Moustapha Hassan
- Department of Laboratory Medicine, Division of Clinical Research Center, Experimental Cancer Medicine, Karolinska Institutet, Stockholm, Sweden
- Clinical Research Center (ECM, KFC, Novum), Karolinska University Hospital-Huddinge, Stockholm, Sweden
- * E-mail:
| | - Ali Moshfegh
- Department of Oncology and Pathology, Cancer Center Karolinska (CCK), Karolinska University Hospital and Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|