1
|
Fu S, Du Y, Pan T, Ma F, He H, Li Y. Causal role of immune cells in aplastic anemia: Mendelian randomization (MR) study. Sci Rep 2024; 14:18010. [PMID: 39097629 PMCID: PMC11297992 DOI: 10.1038/s41598-024-69104-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 07/31/2024] [Indexed: 08/05/2024] Open
Abstract
Prior research has identified associations between immune cells and aplastic anaemia (AA); however, the causal relationships between them have not been conclusively established. A two-sample Mendelian randomisation analysis was conducted to investigate the causal link between 731 immune cell signatures and AA risk using publicly available genetic data. Four types of immune signatures, including relative cell, absolute cell (AC), median fluorescence intensities and morphological parameters, were considered sensitivity analyses were also performed to verify the robustness of the results and assess potential issues such as heterogeneity and horizontal pleiotropy. Following multiple test adjustments using the False Discovery Rate (FDR) method, no statistically significant impact of any immunophenotype on AA was observed. However, twelve immunophenotypes exhibited a significant correlation with AA without FDR correction (p of IVW < 0.01), of which eight were harmful to AA: CD127- CD8br %T cell (Treg panel), CD25 on IgD + CD38dim (B cell panel), CD38 on naive-mature B cell (B cell panel), CD39 + resting Treg % CD4 Treg (Treg panel), CD39 + secreting Treg AC (Treg panel), CD8 on CD28 + CD45RA- CD8br (Treg panel), HLA DR + NK AC (TBNK panel), Naive DN (CD4-CD8-) AC (Maturation stages of T cell panel); and four were protective to AA: CD86 on CD62L + myeloid DC (cDC panel), DC AC (cDC panel), DN (CD4-CD8-) NKT %T cell (TBNK panel), and TD CD4 + AC (Maturation stages of T cell panel). The results of this study demonstrate a close link between immune cells and AA by genetic means, thereby improving the current understanding of the interaction between immune cells and AA risk and providing guidance for future clinical research.
Collapse
Affiliation(s)
- Shaojie Fu
- Department of Nephrology, The First Hospital of Jilin University, Changchun, China
| | - Yazhe Du
- Department of Hematology, The First Hospital of Jilin University, Changchun, China
| | - Tingting Pan
- Teaching Department, The First Hospital of Jilin University, Changchun, China
| | - Fuzhe Ma
- Department of Nephrology, The First Hospital of Jilin University, Changchun, China
| | - Hua He
- Department of Oncology, The First Hospital of Jilin University, Changchun, China
| | - Yuying Li
- Department of Hematology, The First Hospital of Jilin University, Changchun, China.
| |
Collapse
|
2
|
Schnell A. Stem-like T cells in cancer and autoimmunity. Immunol Rev 2024; 325:9-22. [PMID: 38804499 DOI: 10.1111/imr.13356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Stem-like T cells are characterized by their ability to self-renew, survive long-term, and give rise to a heterogeneous pool of effector and memory T cells. Recent advances in single-cell RNA-sequencing (scRNA-seq) and lineage tracing technologies revealed an important role for stem-like T cells in both autoimmunity and cancer. In cancer, stem-like T cells constitute an important arm of the anti-tumor immune response by giving rise to effector T cells that mediate tumor control. In contrast, in autoimmunity stem-like T cells perform an unfavorable role by forming a reservoir of long-lived autoreactive cells that replenish the pathogenic, effector T-cell pool and thereby driving disease pathology. This review provides background on the discovery of stem-like T cells and their function in cancer and autoimmunity. Moreover, the influence of the microbiota and metabolism on the stem-like T-cell pool is summarized. Lastly, the implications of our knowledge about stem-like T cells for clinical treatment strategies for cancer and autoimmunity will be discussed.
Collapse
Affiliation(s)
- Alexandra Schnell
- Whitehead Institute for Biomedical Research, Cambridge, Massachusetts, USA
- Howard Hughes Medical Institute, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| |
Collapse
|
3
|
Zahran AM, El-Badawy OH, Mahran H, Gad E, Saad K, Morsy SG, Makboul A, Zahran ZAM, Elhoufey A, Dailah HG, Elsayh KI. Detection and characterization of autoreactive memory stem T-cells in children with acute immune thrombocytopenia. Clin Exp Med 2024; 24:158. [PMID: 39004660 PMCID: PMC11247050 DOI: 10.1007/s10238-024-01386-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Accepted: 05/28/2024] [Indexed: 07/16/2024]
Abstract
Primary immune thrombocytopenia (ITP) is an acquired autoimmune disorder characterized by an isolated decrease in platelets below 100 × 109/l after the exclusion of other conditions associated with thrombocytopenia. We investigated the role of different memory T-cell subsets, including T stem cell memory (TSCM), in children diagnosed with primary ITP and its association with therapeutic duration. This case-control study included 39 pediatric patients with acute ITP admitted to the Children's Hospital at Assiut University. Using a FACSCanto flow cytometer, CD8 + and CD4 + T-lymphocytes were gated. Five different subsets were characterized in each of these cells according to CD45RO and CD45RA expression. Afterward, gating was performed based on CCR7, CD95, and CD27. Examination of the CD8 + T cells subpopulation showed that Central memory T (TCM) and CD8+ Naïve T (TN) cells were significantly lower in ITP patients than in healthy children (p < 0.0001) and (p = 0.01), respectively. In addition, CD8 + TEMRA was significantly higher in ITP children than in controls (p = 0.001). CD4 + TCM cells were significantly lower in the ITP patient group (p = 0.04). However, CD4 + TEM was significantly higher in patients than controls (p = 0.04). Our research found that ITP patients had an imbalance in the ratio of CD4+ to CD8+ T cells in the peripheral blood and that TCM cells may be involved in the pathogenetic mechanism of ITP. TCMs could help in prediction of patients with higher risk of developing ITP.
Collapse
Affiliation(s)
- Asmaa M Zahran
- Department of Clinical Pathology, South Egypt Cancer Institute, Assiut University, Assiut, Egypt
| | - Omnia H El-Badawy
- Department of Medical Microbiology & Immunology, Faculty of Medicine, Assiut University, Assiut, Egypt.
| | - Hayam Mahran
- Department of Pediatrics, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Eman Gad
- Department of Pediatrics, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Khaled Saad
- Department of Pediatrics, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Salma G Morsy
- Department of Cancer Biology, South Egypt Cancer Institute, Assiut University, Assiut, Egypt
| | - Ahmed Makboul
- Department of Clinical Pathology, South Egypt Cancer Institute, Assiut University, Assiut, Egypt
| | | | - Amira Elhoufey
- Department of Community Health Nursing, Alddrab University College, Jazan University, 45142, Jazan, Saudi Arabia
| | - Hamad Ghaleb Dailah
- Research and Scientific Studies Unit, College of Nursing, Jazan University, Jazan, Saudi Arabia
| | - Khalid I Elsayh
- Department of Pediatrics, Faculty of Medicine, Assiut University, Assiut, Egypt
| |
Collapse
|
4
|
Long J, You X, Yang Q, Wang SR, Zhou M, Zhou W, Wang C, Xie H, Zhang Y, Wang S, Lian ZX, Li L. Bone marrow CD8 + Trm cells induced by IL-15 and CD16 + monocytes contribute to HSPC destruction in human severe aplastic anemia. Clin Immunol 2024; 263:110223. [PMID: 38636890 DOI: 10.1016/j.clim.2024.110223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Accepted: 04/15/2024] [Indexed: 04/20/2024]
Abstract
Idiopathic severe aplastic anemia (SAA) is a disease of bone marrow failure caused by T-cell-induced destruction of hematopoietic stem and progenitor cells (HSPCs), however the mechanism remains unclear. We performed single-cell RNA sequencing of PBMCs and BMMCs from SAA patients and healthy donors and identified a CD8+ T cell subset with a tissue residency phenotype (Trm) in bone marrow that exhibit high IFN-γ and FasL expression and have a higher ability to induce apoptosis in HSPCs in vitro through FasL expression. CD8+ Trm cells were induced by IL-15 presented by IL-15Rα on monocytes, especially CD16+ monocytes, which were increased in SAA patients. CD16+ monocytes contributed to IL-15-induced CD38+CXCR6+ pre-Trm differentiation into CD8+ Trm cells, which can be inhibited by the CD38 inhibitor 78c. Our results demonstrate that IL-15-induced CD8+ Trm cells are pathogenic cells that mediate HSPC destruction in SAA patients and are therapeutic targets for future treatments.
Collapse
Affiliation(s)
- Jie Long
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Xing You
- School of Medicine South China University of Technology, Guangzhou, China; Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Qiong Yang
- School of Medicine South China University of Technology, Guangzhou, China
| | - Song-Rong Wang
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Ming Zhou
- Department of Hematology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| | - Wei Zhou
- Department of Hematology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| | - Caixia Wang
- Department of Hematology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| | - Huafeng Xie
- Center for Medical Research on Innovation and Translation, Institute of Clinical Medicine, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| | - Yuping Zhang
- Department of Hematology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| | - Shunqing Wang
- Department of Hematology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China.
| | - Zhe-Xiong Lian
- Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China.
| | - Liang Li
- Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China.
| |
Collapse
|
5
|
DeZern AE. A way to "mimic" the pathophysiology of acquired SAA. Blood 2024; 143:1318-1320. [PMID: 38573610 DOI: 10.1182/blood.2024024037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/05/2024] Open
|
6
|
Ben Hamza A, Welters C, Stadler S, Brüggemann M, Dietze K, Brauns O, Brümmendorf TH, Winkler T, Bullinger L, Blankenstein T, Rosenberger L, Leisegang M, Kammertöns T, Herr W, Moosmann A, Strobel J, Hackstein H, Dornmair K, Beier F, Hansmann L. Virus-reactive T cells expanded in aplastic anemia eliminate hematopoietic progenitor cells by molecular mimicry. Blood 2024; 143:1365-1378. [PMID: 38277625 DOI: 10.1182/blood.2023023142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 01/17/2024] [Accepted: 01/18/2024] [Indexed: 01/28/2024] Open
Abstract
ABSTRACT Acquired aplastic anemia is a bone marrow failure syndrome characterized by hypocellular bone marrow and peripheral blood pancytopenia. Frequent clinical responses to calcineurin inhibition and antithymocyte globulin strongly suggest critical roles for hematopoietic stem/progenitor cell-reactive T-cell clones in disease pathophysiology; however, their exact contribution and antigen specificities remain unclear. We determined differentiation states and targets of dominant T-cell clones along with their potential to eliminate hematopoietic progenitor cells in the bone marrow of 15 patients with acquired aplastic anemia. Single-cell sequencing and immunophenotyping revealed oligoclonal expansion and effector differentiation of CD8+ T-cell compartments. We reexpressed 28 dominant T-cell receptors (TCRs) of 9 patients in reporter cell lines to determine reactivity with (1) in vitro-expanded CD34+ bone marrow, (2) CD34- bone marrow, or (3) peptide pools covering immunodominant epitopes of highly prevalent viruses. Besides 5 cytomegalovirus-reactive TCRs, we identified 3 TCRs that recognized antigen presented on hematopoietic progenitor cells. T cells transduced with these TCRs eliminated hematopoietic progenitor cells of the respective patients in vitro. One progenitor cell-reactive TCR (11A5) also recognized an epitope of the Epstein-Barr virus-derived latent membrane protein 1 (LMP1) presented on HLA-A∗02:01. We identified 2 LMP1-related mimotopes within the human proteome as activating targets of TCR 11A5, providing proof of concept that molecular mimicry of viral and self-epitopes can drive T cell-mediated elimination of hematopoietic progenitor cells in aplastic anemia.
Collapse
Affiliation(s)
- Amin Ben Hamza
- Department of Hematology, Oncology and Tumor Immunology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Carlotta Welters
- Department of Hematology, Oncology and Tumor Immunology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Serena Stadler
- Department of Hematology, Oncology and Tumor Immunology, Charité-Universitätsmedizin Berlin, Berlin, Germany
- German Cancer Consortium, Partner Site Berlin, and German Cancer Research Center, Heidelberg, Germany
| | - Monika Brüggemann
- Department of Medicine II, Hematology and Oncology, University Hospital Schleswig Holstein, Kiel, Germany
| | - Kerstin Dietze
- Department of Hematology, Oncology and Tumor Immunology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Olaf Brauns
- Miltenyi Biotec B.V. & Co. KG, Bergisch Gladbach, Germany
| | - Tim H Brümmendorf
- Department of Hematology, Oncology, Hemostaseology and Stem Cell Transplantation, Medical Faculty, RWTH Aachen University, Aachen, Germany
- Center for Integrated Oncology, Aachen Bonn Cologne Düsseldorf, Aachen, Germany
| | - Thomas Winkler
- Division of Genetics, Department of Biology, Friedrich Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Lars Bullinger
- Department of Hematology, Oncology and Tumor Immunology, Charité-Universitätsmedizin Berlin, Berlin, Germany
- German Cancer Consortium, Partner Site Berlin, and German Cancer Research Center, Heidelberg, Germany
| | - Thomas Blankenstein
- Molecular Immunology and Gene Therapy, Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Leonie Rosenberger
- Institute of Immunology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Matthias Leisegang
- German Cancer Consortium, Partner Site Berlin, and German Cancer Research Center, Heidelberg, Germany
- Institute of Immunology, Charité-Universitätsmedizin Berlin, Berlin, Germany
- David and Etta Jonas Center for Cellular Therapy, The University of Chicago, Chicago, IL
| | - Thomas Kammertöns
- Institute of Immunology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Wolfgang Herr
- Department of Internal Medicine III, University Hospital Regensburg, Regensburg, Germany
| | - Andreas Moosmann
- Department of Medicine III, Klinikum der Universität München, Munich, Germany
- German Center for Infection Research, Munich, Germany
- Helmholtz Munich, Munich, Germany
| | - Julian Strobel
- Department of Transfusion Medicine and Hemostaseology, University Hospital Erlangen, Friedrich Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Holger Hackstein
- Department of Transfusion Medicine and Hemostaseology, University Hospital Erlangen, Friedrich Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Klaus Dornmair
- Institute of Clinical Neuroimmunology, University Hospital, Ludwig Maximilian University Munich, Munich, Germany
- Biomedical Center, Faculty of Medicine, Ludwig Maximilian University Munich, Martinsried, Germany
| | - Fabian Beier
- Department of Hematology, Oncology, Hemostaseology and Stem Cell Transplantation, Medical Faculty, RWTH Aachen University, Aachen, Germany
- Center for Integrated Oncology, Aachen Bonn Cologne Düsseldorf, Aachen, Germany
| | - Leo Hansmann
- Department of Hematology, Oncology and Tumor Immunology, Charité-Universitätsmedizin Berlin, Berlin, Germany
- German Cancer Consortium, Partner Site Berlin, and German Cancer Research Center, Heidelberg, Germany
- Department of Internal Medicine III, University Hospital Regensburg, Regensburg, Germany
| |
Collapse
|
7
|
Xu S, Xiao Y, Liang X, Lu Y, Deng M. The long-term outcomes and safety of severe aplastic anemia treated with porcine antilymphocyte globulin plus cyclosporine, with or without thrombopoietin receptor agonists: a double-center retrospective study. Expert Rev Hematol 2024; 17:181-188. [PMID: 38687471 DOI: 10.1080/17474086.2024.2350527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Accepted: 02/19/2024] [Indexed: 05/02/2024]
Abstract
BACKGROUND Porcine antilymphocyte globulin (p-ALG) combined with cyclosporine (CsA) has been commonly used for severe aplastic anemia (SAA) patients, but few studies on the combination of p-ALG and thrombopoietin receptor agonist (TPO-RA). RESEARCH DESIGN AND METHODS We retrospectively analyzed the data of 85 people with diagnosed SAA who underwent p-ALG plus CsA, with or without TPO-RA from 2014 to 2023. RESULTS The overall response rates were 55.3% and 65.9% at 3 and 6 months, and the TPO-RA group were 66.7% and 72.3% at 3 and 6 months, without TPO-RA group were 27.8% and 55.6%. In multivariate analysis, baseline platelet count of > 10 × 109/L was a simple predictor of favorable response at 6 months (p = 0.015). The median follow-up time for all patients was 39 months (range 0.4 ~ 104), the 5-year overall survival (OS) rate was 90.6% [95% CI = 82.1-95.2%], and the failure-free survival (FFS) rate was 68.9% [95% CI = 56.6-78.4%]. Having hematologic responses in 6 months was an independent positive predictor for FFS (p = 0.000). Twelve patients (14.1%) suffered from serum sickness, and 9.5% of patients had mild hepatic impairment. CONCLUSIONS p-ALG along with CsA is an effective choice for patients with SAA. p-ALG combined with TPO-RA may contribute to the early restoration of hematopoiesis.
Collapse
Affiliation(s)
- Shan Xu
- The Second Xiangya Hospital, Central South University, Changsha, China
| | - Yue Xiao
- The Second Xiangya Hospital, Central South University, Changsha, China
| | - Xinquan Liang
- Chenzhou First People's Hospital, Nanhua University, Chenzhou, China
| | - Yan Lu
- The Second Xiangya Hospital, Central South University, Changsha, China
| | - Mingyang Deng
- The Second Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
8
|
Hughes EP, Syage AR, Tantin D. Durable CD4 + T cell immunity: cherchez la stem. Trends Immunol 2024; 45:158-166. [PMID: 38388231 PMCID: PMC10947858 DOI: 10.1016/j.it.2024.01.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 01/28/2024] [Accepted: 01/29/2024] [Indexed: 02/24/2024]
Abstract
Mammalian stem cells govern development, tissue homeostasis, and regeneration. Following years of study, their functions have been delineated with increasing precision. The past decade has witnessed heightened widespread use of stem cell terminology in association with durable T cell responses to infection, antitumor immunity, and autoimmunity. Interpreting this literature is complicated by the fact that descriptions are diverse and criteria for labeling 'stem-like' T cells are evolving. Working under the hypothesis that conceptual frameworks developed for actual stem cells can be used to better evaluate and organize T cells described to have stem-like features, we outline widely accepted properties of stem cells and compare these to different 'stem-like' CD4+ T cell populations.
Collapse
Affiliation(s)
- Erik P Hughes
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT 84112, USA; Huntsman Cancer Institute, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| | - Amber R Syage
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT 84112, USA; Huntsman Cancer Institute, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| | - Dean Tantin
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT 84112, USA; Huntsman Cancer Institute, University of Utah School of Medicine, Salt Lake City, UT 84112, USA.
| |
Collapse
|
9
|
Wang J, Zhou R, Zhong L, Chen Y, Wu X, Huang L, Tian Y, Mo W, Wang S, Liu Y. High-dimensional immune profiling using mass cytometry reveals IL-17A-producing γδ T cells as biomarkers in patients with T-cell-activated idiopathic severe aplastic anemia. Int Immunopharmacol 2023; 125:111163. [PMID: 37976596 DOI: 10.1016/j.intimp.2023.111163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 10/19/2023] [Accepted: 10/31/2023] [Indexed: 11/19/2023]
Abstract
Severe aplastic anemia (SAA) is a bone marrow failure syndrome characterized by activated T cells. Features of T-cell activation in the pathophysiology of SAA remain unknown. To understand T cell activation states, we investigated the atlas of peripheral immune cells and the secreted cytokine network with single cell mass cytometry analysis. We found decreased γδ T-cell frequencies in all patients with SAA, together with a significantly increased proportion of interleukin (IL)-17A-producing cell subsets. Cytokine network analysis of immune cells showed significant positive relationship between IL and 17A production from immune cells and disease severity of severe aplastic anemia. On separating SAA into two distinct subgroups based on T-cell activation stage, the proportion of γδ T cells tended to decrease in the T-cell-activated SAA group compared with non-T-cell-activated group. And the proportion of IL-17A-producing γδ T cells (γδT17) within γδ T cells was newly found to be significantly higher in the T-cell-activated SAA group, implying that IL-17A production by γδ T cells was associated with T-cell activation. Overall, our study revealed a role of γδT17 cells in mediating autoreactive T-cell activation in SAA and provided a novel diagnostic indicator for monitoring autoreactive T-cell activation status during the progression of aplastic anemia in the clinic.
Collapse
Affiliation(s)
- Jianwei Wang
- Department of Hematology, Guangzhou First People's Hospital, the Second Affiliated Hospital of South China University of Technology, Guangzhou, Guangdong 510180, China; Center for Medical Research on Innovation and Translation, Guangzhou First People's Hospital, the Second Affiliated Hospital of South China University of Technology, Guangzhou, Guangdong 510005, China
| | - Ruiqing Zhou
- Department of Hematology, Guangzhou First People's Hospital, the Second Affiliated Hospital of South China University of Technology, Guangzhou, Guangdong 510180, China
| | - Limei Zhong
- Department of Laboratory Medicine, Guangdong Second Provincial General Hospital, Guangzhou, Guangdong 510317, China
| | - Yinchun Chen
- Department of Hematology, the Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong 510006, China
| | - Xiaojun Wu
- Department of Hematology, the Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong 510006, China
| | - Liping Huang
- Department of Obstetrics and Gynecology, Southern Medical University Nanfang Hospital, Guangzhou, Guangdong 510515, China
| | - Yan Tian
- Department of Anesthesiology, Jiangxi Provincial Children's Hospital, Nanchang, Jiangxi 330000, China
| | - Wenjian Mo
- Department of Hematology, Guangzhou First People's Hospital, the Second Affiliated Hospital of South China University of Technology, Guangzhou, Guangdong 510180, China
| | - Shunqing Wang
- Department of Hematology, Guangzhou First People's Hospital, the Second Affiliated Hospital of South China University of Technology, Guangzhou, Guangdong 510180, China
| | - Yufeng Liu
- Department of Hematology, Guangzhou First People's Hospital, the Second Affiliated Hospital of South China University of Technology, Guangzhou, Guangdong 510180, China; Center for Medical Research on Innovation and Translation, Guangzhou First People's Hospital, the Second Affiliated Hospital of South China University of Technology, Guangzhou, Guangdong 510005, China.
| |
Collapse
|
10
|
Pool ES, Kooy-Winkelaar Y, van Unen V, Falkenburg JF, Koning F, Heemskerk MHM, Tjon JML. Mass cytometric analysis unveils a disease-specific immune cell network in the bone marrow in acquired aplastic anemia. Front Immunol 2023; 14:1274116. [PMID: 38094307 PMCID: PMC10716190 DOI: 10.3389/fimmu.2023.1274116] [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: 08/07/2023] [Accepted: 11/14/2023] [Indexed: 12/18/2023] Open
Abstract
Idiopathic acquired aplastic anemia (AA) is considered an immune-mediated syndrome of bone marrow failure since approximately 70% of patients respond to immunosuppressive therapy (IST) consisting of a course of anti-thymocyte globulin (ATG) followed by long-term use of ciclosporin. However, the immune response that underlies the pathogenesis of AA remains poorly understood. In this study, we applied high-dimensional mass cytometry on bone marrow aspirates of AA patients pre-ATG, AA patients post-ATG and healthy donors to decipher which immune cells may be implicated in the pathogenesis of AA. We show that the bone marrow of AA patients features an immune cell composition distinct from healthy donors, with significant differences in the myeloid, B-cell, CD4+ and CD8+ T-cells lineages. Specifically, we discovered that AA pre-ATG is characterized by a disease-specific immune cell network with high frequencies of CD16+ myeloid cells, CCR6++ B-cells, Th17-like CCR6+ memory CD4+ T-cells, CD45RA+CCR7+CD38+ CD8+ T-cells and KLRG1+ terminally differentiated effector memory (EMRA) CD8+ T-cells, compatible with a state of chronic inflammation. Successful treatment with IST strongly reduced the levels of CD16+ myeloid cells and showed a trend toward normalization of the frequencies of CCR6++ B-cells, CCR6+ memory CD4+ T-cells and KLRG1+EMRA CD8+ T-cells. Altogether, our study provides a unique overview of the immune landscape in bone marrow in AA at a single-cell level and proposes CCR6 as a potential new therapeutic target in AA.
Collapse
Affiliation(s)
- Emma S. Pool
- Department of Hematology, Leiden University Medical Center, Leiden, Netherlands
| | | | - Vincent van Unen
- Department of Immunology, Leiden University Medical Center, Leiden, Netherlands
- Institute for Immunity, Transplantation, and Infection, Stanford University School of Medicine, Stanford, CA, United States
| | | | - Frits Koning
- Department of Immunology, Leiden University Medical Center, Leiden, Netherlands
| | | | - Jennifer M-L. Tjon
- Department of Hematology, Leiden University Medical Center, Leiden, Netherlands
| |
Collapse
|
11
|
Zhang M, Long X, Xiao Y, Jin J, Chen C, Meng J, Liu W, Liu A, Chen L. Assessment and predictive ability of the absolute neutrophil count in peripheral blood for in vivo CAR T cells expansion and CRS. J Immunother Cancer 2023; 11:e007790. [PMID: 38016717 PMCID: PMC10685953 DOI: 10.1136/jitc-2023-007790] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/28/2023] [Indexed: 11/30/2023] Open
Abstract
BACKGROUND Chimeric antigen receptor (CAR) T cell therapy is an advanced and effective immunotherapy for relapsed or refractory B-cell malignancies. High expansion of CAR T cells in vivo and durable antitumor activity indicate a persistent therapeutic response. However, this treatment is linked to a high frequency of adverse events, such as cytokine release syndrome (CRS), which affects its efficacy and can even be life-threatening. At present, a variety of markers associated with clinical response and treatment toxicity after CAR T cells infusion have been reported. Although these biomarkers can act as effective indicators reflecting CAR T cells expansion as well as CRS, they fail to predict the expansion rate of CAR T cells. Hence, further investigation is urgent to find a new biomarker to fill this void. METHODS We analyzed the association between the absolute neutrophil count (ANC) and CAR expansion and CRS in 45 patients with B-cell malignancies from two clinical trials. We proposed that ANC could be a practical biomarker for CAR T cells expansion and CRS, and conducted a feasibility analysis on its predictive ability. RESULTS In this study, 17 B-cell hematological malignancy patients with anti-B-cell maturation antigen CAR-treated and 28 with CAR19/22 T-cell-treated were enrolled and divided into an ANC-absence group and an ANC-presence group. The results showed that ANC absence correlated positively with CAR expansion and the expansion rate. The ANC can be used as a predictive marker for CAR T cells expansion. Moreover, the patients with ANC absence experienced a more severe CRS, and ANC performed a predictive ability for CRS. In addition, the peak serum concentration of several cytokines involved in CRS was higher in patients with ANC absence. CONCLUSION Thus, we suggest ANC as an evaluative and predictive biomarker for CAR expansion and CRS during CAR T cell therapy, which can help to maximize clinical efficacy, reduce treatment-related toxicity and prolong survival.
Collapse
Affiliation(s)
- Man Zhang
- Department of Hematology, Harbin Medical University Cancer Hospital, Harbin Medical University, Harbin, Heilongjiang, China
| | - Xiaolu Long
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yi Xiao
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Jin Jin
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Department of Hematology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Caixia Chen
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Jiao Meng
- Department of Hematology, Harbin Medical University Cancer Hospital, Harbin Medical University, Harbin, Heilongjiang, China
| | - Wanying Liu
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Aichun Liu
- Department of Hematology, Harbin Medical University Cancer Hospital, Harbin Medical University, Harbin, Heilongjiang, China
| | - Liting Chen
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| |
Collapse
|
12
|
Wu Z, Young NS. Single-cell genomics in acquired bone marrow failure syndromes. Blood 2023; 142:1193-1207. [PMID: 37478398 PMCID: PMC10644099 DOI: 10.1182/blood.2022018581] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 06/20/2023] [Accepted: 06/20/2023] [Indexed: 07/23/2023] Open
Abstract
Mechanistic studies of immune bone marrow failure are difficult because of the scarcity of residual cells, the involvement of multiple cell types, and the inherent complexities of hematopoiesis and immunity. Single-cell genomic technologies and bioinformatics allow extensive, multidimensional analysis of a very limited number of cells. We review emerging applications of single-cell techniques, and early results related to disease pathogenesis: effector and target cell populations and relationships, cell-autonomous and nonautonomous phenotypes in clonal hematopoiesis, transcript splicing, chromosomal abnormalities, and T-cell receptor usage and clonality. Dense and complex data from single-cell techniques provide insights into pathophysiology, natural history, and therapeutic drug effects.
Collapse
Affiliation(s)
- Zhijie Wu
- Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Neal S. Young
- Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
| |
Collapse
|
13
|
Koldej RM, Prabahran A, Tan CW, Ludford-Menting M, Morgan H, Holzwart N, Davis MJ, Ritchie DS. Spatial proteomics identifies a spectrum of immune dysregulation in acquired bone marrow failure syndromes. Front Immunol 2023; 14:1213560. [PMID: 37818364 PMCID: PMC10560754 DOI: 10.3389/fimmu.2023.1213560] [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: 04/28/2023] [Accepted: 09/04/2023] [Indexed: 10/12/2023] Open
Abstract
Poor graft function (PGF), manifested by multilineage cytopenias and complete donor chimerism post-allogeneic stem cell transplantation (alloSCT), and acquired aplastic anaemia (AA) are immune-mediated acquired bone marrow (BM) failure syndromes with a similar clinical presentation. In this study, we used spatial proteomics to compare the immunobiology of the BM microenvironment and identify common mechanisms of immune dysregulation under these conditions. Archival BM trephines from patients exhibited downregulation of the immunoregulatory protein VISTA and the M2 macrophage marker and suppressor of T-cell activation ARG1 with increased expression of the immune checkpoint B7-H3 compared to normal controls. Increased CD163 and CD14 expression suggested monocyte/macrophage skewing, which, combined with dysregulation of STING and VISTA, is indicative of an environment of reduced immunoregulation resulting in the profound suppression of hematopoiesis in these two conditions. There were no changes in the immune microenvironment between paired diagnostic AA and secondary MDS/AML samples suggesting that leukaemic clones develop in the impaired immune microenvironment of AA without the need for further alterations. Of the eight proteins with dysregulated expression shared by diagnostic AA and PGF, the diagnostic AA samples had a greater fold change in expression than PGF, suggesting that these diseases represent a spectrum of immune dysregulation. Unexpectedly, analysis of samples from patients with good graft function post-alloSCT demonstrated significant changes in the immune microenvironment compared to normal controls, with downregulation of CD44, STING, VISTA, and ARG1, suggesting that recovery of multilineage haematopoiesis post-alloSCT does not reflect recovery of immune function and may prime patients for the development of PGF upon further inflammatory insult. The demonstrable similarities in the immunopathology of AA and PGF will allow the design of clinical interventions that include both patient cohorts to accelerate therapeutic discovery and translation.
Collapse
Affiliation(s)
- Rachel M. Koldej
- Australian Cancer Research Foundation (ACRF) Translational Research Laboratory, Royal Melbourne Hospital, Melbourne, VIC, Australia
- Department of Medicine, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Melbourne, VIC, Australia
| | - Ashvind Prabahran
- Australian Cancer Research Foundation (ACRF) Translational Research Laboratory, Royal Melbourne Hospital, Melbourne, VIC, Australia
- Department of Medicine, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Melbourne, VIC, Australia
- Clinical Haematology, Peter MacCallum Cancer Centre and Royal Melbourne Hospital, Melbourne, VIC, Australia
| | - Chin Wee Tan
- Bioinformatics Division, The Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia
- Department of Medical Biology, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Melbourne, VIC, Australia
| | - Mandy Ludford-Menting
- Australian Cancer Research Foundation (ACRF) Translational Research Laboratory, Royal Melbourne Hospital, Melbourne, VIC, Australia
- Department of Medicine, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Melbourne, VIC, Australia
| | - Huw Morgan
- Australian Cancer Research Foundation (ACRF) Translational Research Laboratory, Royal Melbourne Hospital, Melbourne, VIC, Australia
- Department of Medicine, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Melbourne, VIC, Australia
| | - Nicholas Holzwart
- Australian Cancer Research Foundation (ACRF) Translational Research Laboratory, Royal Melbourne Hospital, Melbourne, VIC, Australia
- Department of Medicine, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Melbourne, VIC, Australia
| | - Melissa J. Davis
- Bioinformatics Division, The Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia
- Department of Medical Biology, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Melbourne, VIC, Australia
- Department of Clinical Pathology, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Melbourne, VIC, Australia
| | - David S. Ritchie
- Australian Cancer Research Foundation (ACRF) Translational Research Laboratory, Royal Melbourne Hospital, Melbourne, VIC, Australia
- Department of Medicine, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Melbourne, VIC, Australia
- Clinical Haematology, Peter MacCallum Cancer Centre and Royal Melbourne Hospital, Melbourne, VIC, Australia
| |
Collapse
|
14
|
Sachdeva M, Taneja S, Sachdeva N. Stem cell-like memory T cells: Role in viral infections and autoimmunity. World J Immunol 2023; 13:11-22. [DOI: 10.5411/wji.v13.i2.11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 07/06/2023] [Accepted: 07/27/2023] [Indexed: 08/14/2023] Open
Abstract
Stem cell-like memory T (TSCM) cells possess stem cell properties including multipotency and self-renewal and are being recognized as emerging players in various human diseases. Advanced technologies such as multiparametric flowcytometry and single cell sequencing have enabled their identification and molecular characterization. In case of chronic viral diseases such as human immunodeficiency virus-1, CD4+ TSCM cells, serve as major reservoirs of the latent virus. However, during immune activation and functional exhaustion of effector T cells, these cells also possess the potential to replenish the pool of functional effector cells to curtail the infection. More recently, these cells are speculated to play important role in protective immunity following acute viral infections such as coronavirus disease 2019 and might be amenable for therapeutics by ex vivo expansion. Similarly, studies are also investigating their pathological role in driving autoimmune responses. However, there are several gaps in the understanding of the role of TSCM cells in viral and autoimmune diseases to make them potential therapeutic targets. In this minireview, we have attempted an updated compilation of the dyadic role of these complex TSCM cells during such human diseases along with their biology and transcriptional programs.
Collapse
Affiliation(s)
- Meenakshi Sachdeva
- Department of Pediatrics, Post Graduate Institute of Medical Education and Research (PGIMER), Chandigarh 160012, India
| | - Shivangi Taneja
- Department of Endocrinology, Post Graduate Institute of Medical Education and Research (PGIMER), Chandigarh 160012, India
| | - Naresh Sachdeva
- Department of Endocrinology, Post Graduate Institute of Medical Education and Research (PGIMER), Chandigarh 160012, India
| |
Collapse
|
15
|
Liu Z, Jiao Q, Xie H, Liu J. Role of stem cell-like memory T cells in autoimmune diseases. ZHONG NAN DA XUE XUE BAO. YI XUE BAN = JOURNAL OF CENTRAL SOUTH UNIVERSITY. MEDICAL SCIENCES 2023; 48:1098-1104. [PMID: 37724413 PMCID: PMC10930044 DOI: 10.11817/j.issn.1672-7347.2023.230051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Indexed: 09/20/2023]
Abstract
Stem cell-like memory T (TSCM) cell is a memory T cell subset with characteristics of long life span, consistent self-renewing, and the multipotent capacity to reconstitute the memory and effector T cell subsets. TSCM cell is the least differentiated cell in the memory T lymphocyte system, endowed with the stem cell-like ability, and it is essential for maintaining functional immunity. In addition, owing to its robust potential for immune reconstitution, it is central player in many physiological and pathological human processes. TSCM cell plays an important role in the occurrence and development of various autoimmune diseases. The specific role of TSCM cell in autoimmune diseases may make it a potential target for the treatment of multiple autoimmune diseases, driving effective immune reconstitution in therapy.
Collapse
Affiliation(s)
- Zhichen Liu
- Department of Otolaryngology, First Affiliated Hospital of Soochow University, Suzhou Jiangsu 215006.
| | - Qingqing Jiao
- Department of Dermatology and Venereology, First Affiliated Hospital of Soochow University, Suzhou Jiangsu 215006, China
| | - Huanxia Xie
- Department of Otolaryngology, First Affiliated Hospital of Soochow University, Suzhou Jiangsu 215006
| | - Jisheng Liu
- Department of Otolaryngology, First Affiliated Hospital of Soochow University, Suzhou Jiangsu 215006.
| |
Collapse
|
16
|
Fazeli P, Kalani M, Hosseini M. T memory stem cell characteristics in autoimmune diseases and their promising therapeutic values. Front Immunol 2023; 14:1204231. [PMID: 37497231 PMCID: PMC10366905 DOI: 10.3389/fimmu.2023.1204231] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 06/16/2023] [Indexed: 07/28/2023] Open
Abstract
Memory T cells are conventionally subdivided into T central memory (TCM) and T effector memory (TEM) cells. However, a new subset of memory T cells named T memory stem cell (TSCM) cells has been recognized that possesses capabilities of both TCM and TEM cells including lymphoid homing and performing effector roles through secretion of cytokines such as interleukin-2 (IL-2) and interferon-gamma (IFN-γ). The TSCM subset has some biological properties including stemness, antigen independency, high proliferative potential, signaling pathway and lipid metabolism. On the other hand, memory T cells are considered one of the principal culprits in the pathogenesis of autoimmune diseases. TSCM cells are responsible for developing long-term defensive immunity against different foreign antigens, alongside tumor-associated antigens, which mainly derive from self-antigens. Hence, antigen-specific TSCM cells can produce antitumor responses that are potentially able to trigger autoimmune activities. Therefore, we reviewed recent evidence on TSCM cell functions in autoimmune disorders including type 1 diabetes, systemic lupus erythematosus, rheumatoid arthritis, acquired aplastic anemia, immune thrombocytopenia, and autoimmune uveitis. We also introduced TSCM cell lineage as an innovative prognostic biomarker and a promising therapeutic target in autoimmune settings.
Collapse
Affiliation(s)
- Pooria Fazeli
- Trauma Research Center, Shahid Rajaee (Emtiaz) Trauma Hospital, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mehdi Kalani
- Department of Immunology, Prof. Alborzi Clinical Microbiology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Maryam Hosseini
- Trauma Research Center, Shahid Rajaee (Emtiaz) Trauma Hospital, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
17
|
Fanti S, Stephenson E, Rocha-Vieira E, Protonotarios A, Kanoni S, Shahaj E, Longhi MP, Vyas VS, Dyer C, Pontarini E, Asimaki A, Bueno-Beti C, De Gaspari M, Rizzo S, Basso C, Bombardieri M, Coe D, Wang G, Harding D, Gallagher I, Solito E, Elliott P, Heymans S, Sikking M, Savvatis K, Mohiddin SA, Marelli-Berg FM. Circulating c-Met-Expressing Memory T Cells Define Cardiac Autoimmunity. Circulation 2022; 146:1930-1945. [PMID: 36417924 PMCID: PMC9770129 DOI: 10.1161/circulationaha.121.055610] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Accepted: 09/20/2022] [Indexed: 11/25/2022]
Abstract
BACKGROUND Autoimmunity is increasingly recognized as a key contributing factor in heart muscle diseases. The functional features of cardiac autoimmunity in humans remain undefined because of the challenge of studying immune responses in situ. We previously described a subset of c-mesenchymal epithelial transition factor (c-Met)-expressing (c-Met+) memory T lymphocytes that preferentially migrate to cardiac tissue in mice and humans. METHODS In-depth phenotyping of peripheral blood T cells, including c-Met+ T cells, was undertaken in groups of patients with inflammatory and noninflammatory cardiomyopathies, patients with noncardiac autoimmunity, and healthy controls. Validation studies were carried out using human cardiac tissue and in an experimental model of cardiac inflammation. RESULTS We show that c-Met+ T cells are selectively increased in the circulation and in the myocardium of patients with inflammatory cardiomyopathies. The phenotype and function of c-Met+ T cells are distinct from those of c-Met-negative (c-Met-) T cells, including preferential proliferation to cardiac myosin and coproduction of multiple cytokines (interleukin-4, interleukin-17, and interleukin-22). Furthermore, circulating c-Met+ T cell subpopulations in different heart muscle diseases identify distinct and overlapping mechanisms of heart inflammation. In experimental autoimmune myocarditis, elevations in autoantigen-specific c-Met+ T cells in peripheral blood mark the loss of immune tolerance to the heart. Disease development can be halted by pharmacologic c-Met inhibition, indicating a causative role for c-Met+ T cells. CONCLUSIONS Our study demonstrates that the detection of circulating c-Met+ T cells may have use in the diagnosis and monitoring of adaptive cardiac inflammation and definition of new targets for therapeutic intervention when cardiac autoimmunity causes or contributes to progressive cardiac injury.
Collapse
Affiliation(s)
- Silvia Fanti
- William Harvey Research Institute, Barts and The London Faculty of Medicine and Dentistry (S.F., E. Stephenson, E.R.-V., S.K., E. Shahaj, M.P.L., V.S.V., C.D., E.P., M.B., D.C., G.W., D.H., E. Solito, K.S., S.A.M., F.M.M.-B.), Queen Mary University of London, UK
| | - Edward Stephenson
- William Harvey Research Institute, Barts and The London Faculty of Medicine and Dentistry (S.F., E. Stephenson, E.R.-V., S.K., E. Shahaj, M.P.L., V.S.V., C.D., E.P., M.B., D.C., G.W., D.H., E. Solito, K.S., S.A.M., F.M.M.-B.), Queen Mary University of London, UK
- Barts Heart Centre, Barts Health NHS Trust, St Bartholomew’s Hospital, West Smithfield, London (E. Stephenson, A.P., V.S.V., D.H., P.E., K.S., S.A.M.)
| | - Etel Rocha-Vieira
- William Harvey Research Institute, Barts and The London Faculty of Medicine and Dentistry (S.F., E. Stephenson, E.R.-V., S.K., E. Shahaj, M.P.L., V.S.V., C.D., E.P., M.B., D.C., G.W., D.H., E. Solito, K.S., S.A.M., F.M.M.-B.), Queen Mary University of London, UK
- Federal University of Vales do Jequitinhonha e Mucuri, Diamantina, Minas Gerais, Brazil (E.R.-V.)
| | - Alexandros Protonotarios
- Barts Heart Centre, Barts Health NHS Trust, St Bartholomew’s Hospital, West Smithfield, London (E. Stephenson, A.P., V.S.V., D.H., P.E., K.S., S.A.M.)
- Institute of Cardiovascular Science, University College London, UK (A.P., P.E.)
| | - Stavroula Kanoni
- William Harvey Research Institute, Barts and The London Faculty of Medicine and Dentistry (S.F., E. Stephenson, E.R.-V., S.K., E. Shahaj, M.P.L., V.S.V., C.D., E.P., M.B., D.C., G.W., D.H., E. Solito, K.S., S.A.M., F.M.M.-B.), Queen Mary University of London, UK
| | - Eriomina Shahaj
- William Harvey Research Institute, Barts and The London Faculty of Medicine and Dentistry (S.F., E. Stephenson, E.R.-V., S.K., E. Shahaj, M.P.L., V.S.V., C.D., E.P., M.B., D.C., G.W., D.H., E. Solito, K.S., S.A.M., F.M.M.-B.), Queen Mary University of London, UK
| | - M. Paula Longhi
- William Harvey Research Institute, Barts and The London Faculty of Medicine and Dentistry (S.F., E. Stephenson, E.R.-V., S.K., E. Shahaj, M.P.L., V.S.V., C.D., E.P., M.B., D.C., G.W., D.H., E. Solito, K.S., S.A.M., F.M.M.-B.), Queen Mary University of London, UK
| | - Vishal S. Vyas
- William Harvey Research Institute, Barts and The London Faculty of Medicine and Dentistry (S.F., E. Stephenson, E.R.-V., S.K., E. Shahaj, M.P.L., V.S.V., C.D., E.P., M.B., D.C., G.W., D.H., E. Solito, K.S., S.A.M., F.M.M.-B.), Queen Mary University of London, UK
- Barts Heart Centre, Barts Health NHS Trust, St Bartholomew’s Hospital, West Smithfield, London (E. Stephenson, A.P., V.S.V., D.H., P.E., K.S., S.A.M.)
| | - Carlene Dyer
- William Harvey Research Institute, Barts and The London Faculty of Medicine and Dentistry (S.F., E. Stephenson, E.R.-V., S.K., E. Shahaj, M.P.L., V.S.V., C.D., E.P., M.B., D.C., G.W., D.H., E. Solito, K.S., S.A.M., F.M.M.-B.), Queen Mary University of London, UK
| | - Elena Pontarini
- William Harvey Research Institute, Barts and The London Faculty of Medicine and Dentistry (S.F., E. Stephenson, E.R.-V., S.K., E. Shahaj, M.P.L., V.S.V., C.D., E.P., M.B., D.C., G.W., D.H., E. Solito, K.S., S.A.M., F.M.M.-B.), Queen Mary University of London, UK
| | - Angeliki Asimaki
- Molecular and Clinical Science Institute, St George’s, University of London, UK (A.A., C.B.-B.)
| | - Carlos Bueno-Beti
- Molecular and Clinical Science Institute, St George’s, University of London, UK (A.A., C.B.-B.)
| | - Monica De Gaspari
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padua Medical School, Italy (M.D.G., S.R., C.B.)
| | - Stefania Rizzo
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padua Medical School, Italy (M.D.G., S.R., C.B.)
| | - Cristina Basso
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padua Medical School, Italy (M.D.G., S.R., C.B.)
| | - Michele Bombardieri
- William Harvey Research Institute, Barts and The London Faculty of Medicine and Dentistry (S.F., E. Stephenson, E.R.-V., S.K., E. Shahaj, M.P.L., V.S.V., C.D., E.P., M.B., D.C., G.W., D.H., E. Solito, K.S., S.A.M., F.M.M.-B.), Queen Mary University of London, UK
| | - David Coe
- William Harvey Research Institute, Barts and The London Faculty of Medicine and Dentistry (S.F., E. Stephenson, E.R.-V., S.K., E. Shahaj, M.P.L., V.S.V., C.D., E.P., M.B., D.C., G.W., D.H., E. Solito, K.S., S.A.M., F.M.M.-B.), Queen Mary University of London, UK
| | - Guosu Wang
- William Harvey Research Institute, Barts and The London Faculty of Medicine and Dentistry (S.F., E. Stephenson, E.R.-V., S.K., E. Shahaj, M.P.L., V.S.V., C.D., E.P., M.B., D.C., G.W., D.H., E. Solito, K.S., S.A.M., F.M.M.-B.), Queen Mary University of London, UK
| | - Daniel Harding
- William Harvey Research Institute, Barts and The London Faculty of Medicine and Dentistry (S.F., E. Stephenson, E.R.-V., S.K., E. Shahaj, M.P.L., V.S.V., C.D., E.P., M.B., D.C., G.W., D.H., E. Solito, K.S., S.A.M., F.M.M.-B.), Queen Mary University of London, UK
- Barts Heart Centre, Barts Health NHS Trust, St Bartholomew’s Hospital, West Smithfield, London (E. Stephenson, A.P., V.S.V., D.H., P.E., K.S., S.A.M.)
| | - Iain Gallagher
- Faculty of Health Sciences & Sport, University of Stirling, UK (I.G.)
| | - Egle Solito
- William Harvey Research Institute, Barts and The London Faculty of Medicine and Dentistry (S.F., E. Stephenson, E.R.-V., S.K., E. Shahaj, M.P.L., V.S.V., C.D., E.P., M.B., D.C., G.W., D.H., E. Solito, K.S., S.A.M., F.M.M.-B.), Queen Mary University of London, UK
- Department of Medicina Molecolare e Biotecnologie Mediche, University of Naples “Federico II,” Italy (E. Solito)
| | - Perry Elliott
- Barts Heart Centre, Barts Health NHS Trust, St Bartholomew’s Hospital, West Smithfield, London (E. Stephenson, A.P., V.S.V., D.H., P.E., K.S., S.A.M.)
- Institute of Cardiovascular Science, University College London, UK (A.P., P.E.)
| | - Stephane Heymans
- Maastricht University Medical Centre, Cardiovascular Research Institute Maastricht, the Netherlands (S.H., M.S.)
- Department of Cardiovascular Sciences, Centre for Vascular and Molecular Biology, KU Leuven, Belgium (S.H.)
| | - Maurits Sikking
- Maastricht University Medical Centre, Cardiovascular Research Institute Maastricht, the Netherlands (S.H., M.S.)
| | - Konstantinos Savvatis
- William Harvey Research Institute, Barts and The London Faculty of Medicine and Dentistry (S.F., E. Stephenson, E.R.-V., S.K., E. Shahaj, M.P.L., V.S.V., C.D., E.P., M.B., D.C., G.W., D.H., E. Solito, K.S., S.A.M., F.M.M.-B.), Queen Mary University of London, UK
- Barts Heart Centre, Barts Health NHS Trust, St Bartholomew’s Hospital, West Smithfield, London (E. Stephenson, A.P., V.S.V., D.H., P.E., K.S., S.A.M.)
| | - Saidi A. Mohiddin
- William Harvey Research Institute, Barts and The London Faculty of Medicine and Dentistry (S.F., E. Stephenson, E.R.-V., S.K., E. Shahaj, M.P.L., V.S.V., C.D., E.P., M.B., D.C., G.W., D.H., E. Solito, K.S., S.A.M., F.M.M.-B.), Queen Mary University of London, UK
- Barts Heart Centre, Barts Health NHS Trust, St Bartholomew’s Hospital, West Smithfield, London (E. Stephenson, A.P., V.S.V., D.H., P.E., K.S., S.A.M.)
| | - Federica M. Marelli-Berg
- William Harvey Research Institute, Barts and The London Faculty of Medicine and Dentistry (S.F., E. Stephenson, E.R.-V., S.K., E. Shahaj, M.P.L., V.S.V., C.D., E.P., M.B., D.C., G.W., D.H., E. Solito, K.S., S.A.M., F.M.M.-B.), Queen Mary University of London, UK
- Centre for Inflammation and Therapeutic Innovation (F.M.M.-B.), Queen Mary University of London, UK
| |
Collapse
|
18
|
Mori K, Nishii K, Kitamura W, Urata T, Baba T, Omori H, Nakanishi M, Tamura T, Kuyama S. Case report a case of durvalumab induced aplastic anemia after chemoradiotherapy for NSCLC. CURRENT PROBLEMS IN CANCER: CASE REPORTS 2022. [DOI: 10.1016/j.cpccr.2022.100192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
|
19
|
Fazeli P, Talepoor AG, Faghih Z, Gholijani N, Ataollahi MR, Ali‐Hassanzadeh M, Moravej H, Kalantar K. The frequency of CD4+ and CD8+ circulating T stem cell memory in type 1 diabetes. Immun Inflamm Dis 2022; 10:e715. [PMID: 36169248 PMCID: PMC9500591 DOI: 10.1002/iid3.715] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Revised: 08/27/2022] [Accepted: 09/13/2022] [Indexed: 11/07/2022] Open
Abstract
INTRODUCTION The frequencies and functions of T stem cell memory (TSCM) subsets vary in autoimmune diseases. We evaluated the frequencies of CD4+ and CD8+ TSCM subsets as well as their PD-1 expression levels in patients with T1D. METHODS Blood samples were collected from new case (NC) (n = 15), and long-term (LT) (n = 15) groups and healthy controls (n = 15). Five subsets of T cells including TCM(CD4+ /CD8+ CCR7+ CD45RO+ CD95+ ), TCMhi (CD4+ /CD8+ CCR7+ CD45ROhi CD95+ ), TEM(CD4+ /CD8+ CCR7- CD45RO+ CD95+ ), TSCM(CD4+ /CD8+ CCR7+ CD45RO- CD95+ ), and T naive (CD4+ /CD8+ CCR7+ CD45RO- CD95- ) were detected by flow-cytometry. RESULTS The frequency of CD4+ TSCM was higher in NC patients than LT patients and controls (p < .0001 and p = .0086, respectively). A higher percentage of the CD8+ T naive cells was shown in NC patients as compared with LT and healthy individuals (p = .0003 and p = .0002, respectively). An increased level of PD-1 expression was observed on the CD4+ TCM and TCMhi cells in LT patients as compared with healthy controls (p = .0037 and p = .0145, respectively). Also, the higher PD-1 expression was observed on the CD8+ TCM and TCMhi in NC and LT patients as compared with controls (p = .0068 and p < .0001; p = .0012 and p = .0012, respectively). CONCLUSION Considering TSCMs' capacities to generate all memory and effector T cells, our results may suggest a potential association between the increased frequencies of TSCMs and T1D progression.
Collapse
Affiliation(s)
- Pooriya Fazeli
- Department of ImmunologySchool of MedicineShiraz University of Medical SciencesShirazIran
| | - Atefe Ghamar Talepoor
- Department of ImmunologySchool of MedicineShiraz University of Medical SciencesShirazIran
| | - Zahra Faghih
- Shiraz Institute for Cancer ResearchSchool of MedicineShiraz University of Medical SciencesShirazIran
| | - Nasser Gholijani
- Autoimmune Diseases Research CenterShiraz University of Medical SciencesShirazIran
| | | | | | - Hossein Moravej
- Department of PediatricsSchool of MedicineShiraz University of Medical SciencesShirazIran
| | - Kurosh Kalantar
- Department of ImmunologySchool of MedicineShiraz University of Medical SciencesShirazIran
- Autoimmune Diseases Research CenterShiraz University of Medical SciencesShirazIran
| |
Collapse
|
20
|
Xie Y, Shao F, Lei J, Huang N, Fan Z, Yu H. Case report: A STAT1 gain-of-function mutation causes a syndrome of combined immunodeficiency, autoimmunity and pure red cell aplasia. Front Immunol 2022; 13:928213. [PMID: 36105803 PMCID: PMC9464931 DOI: 10.3389/fimmu.2022.928213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 08/05/2022] [Indexed: 11/13/2022] Open
Abstract
Inherited autosomal dominant gain-of-function (GOF) mutations of signal transducer and activator of transcription 1 (STAT1) cause a wide range of symptoms affecting multiple systems, including chronic mucocutaneous candidiasis (CMC), infections, and autoimmune disorders. We describe a rare case of STAT1 mutation with recurrent CMC, lung infections, and anemia. According to the whole-exome sequencing (WES), the patient was genetically mutated in STAT1 GOF (c.854A>G, p.Q285R), and bone marrow biopsy suggested pure red cell aplasia (PRCA). As a functional verification, STAT1 levels and phosphorylation (p-STAT1) of peripheral blood mononuclear cells (PBMCs) following IFN-γ stimulation in STAT1 GOF patient was higher than in the healthy control. Combination therapy of blood transfusion, antimicrobials, intravenous immunoglobulin, methylprednisolone, and the Janus Kinase (JAK) specific inhibitor ruxolitinib was used during treatment of patients. The patient also received a hematopoietic stem cell transplant (HSCT) to help with infections and anemia. This is the first reported case of STAT1 GOF disease complicated with PRCA. This complication might be attributed to immune disorders caused by STAT1 GOF. Furthermore, ruxolitinib may be a viable therapeutic option before HSCT to improve disease management.
Collapse
Affiliation(s)
- Yifan Xie
- Department of Rheumatology and Immunology, Children’s Hospital of Nanjing Medical University, Nanjing, China
| | - Fenli Shao
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Biotechnology and Pharmaceutical Sciences, School of Life Sciences, Nanjing University, Nanjing, China
| | - Juan Lei
- Department of Pediatric Nephrology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Na Huang
- Department of Rheumatology and Immunology, Children’s Hospital of Nanjing Medical University, Nanjing, China
| | - Zhidan Fan
- Department of Rheumatology and Immunology, Children’s Hospital of Nanjing Medical University, Nanjing, China
- *Correspondence: Haiguo Yu, ; Zhidan Fan,
| | - Haiguo Yu
- Department of Rheumatology and Immunology, Children’s Hospital of Nanjing Medical University, Nanjing, China
- *Correspondence: Haiguo Yu, ; Zhidan Fan,
| |
Collapse
|
21
|
Yi L, Yang L. Stem-like T cells and niches: Implications in human health and disease. Front Immunol 2022; 13:907172. [PMID: 36059484 PMCID: PMC9428355 DOI: 10.3389/fimmu.2022.907172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 07/28/2022] [Indexed: 11/13/2022] Open
Abstract
Recently, accumulating evidence has elucidated the important role of T cells with stem-like characteristics in long-term maintenance of T cell responses and better patient outcomes after immunotherapy. The fate of TSL cells has been correlated with many physiological and pathological human processes. In this review, we described present advances demonstrating that stem-like T (TSL) cells are central players in human health and disease. We interpreted the evolutionary characteristics, mechanism and functions of TSL cells. Moreover, we discuss the import role of distinct niches and how they affect the stemness of TSL cells. Furthermore, we also outlined currently available strategies to generate TSL cells and associated affecting factors. Moreover, we summarized implication of TSL cells in therapies in two areas: stemness enhancement for vaccines, ICB, and adoptive T cell therapies, and stemness disruption for autoimmune disorders.
Collapse
|
22
|
Kelkka T, Tyster M, Lundgren S, Feng X, Kerr C, Hosokawa K, Huuhtanen J, Keränen M, Patel B, Kawakami T, Maeda Y, Nieminen O, Kasanen T, Aronen P, Yadav B, Rajala H, Nakazawa H, Jaatinen T, Hellström-Lindberg E, Ogawa S, Ishida F, Nishikawa H, Nakao S, Maciejewski J, Young NS, Mustjoki S. Anti-COX-2 autoantibody is a novel biomarker of immune aplastic anemia. Leukemia 2022; 36:2317-2327. [PMID: 35927326 PMCID: PMC9417997 DOI: 10.1038/s41375-022-01654-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 07/06/2022] [Accepted: 07/11/2022] [Indexed: 11/09/2022]
Abstract
In immune aplastic anemia (IAA), severe pancytopenia results from the immune-mediated destruction of hematopoietic stem cells. Several autoantibodies have been reported, but no clinically applicable autoantibody tests are available for IAA. We screened autoantibodies using a microarray containing >9000 proteins and validated the findings in a large international cohort of IAA patients (n = 405) and controls (n = 815). We identified a novel autoantibody that binds to the C-terminal end of cyclooxygenase 2 (COX-2, aCOX-2 Ab). In total, 37% of all adult IAA patients tested positive for aCOX-2 Ab, while only 1.7% of the controls were aCOX-2 Ab positive. Sporadic non-IAA aCOX-2 Ab positive cases were observed among patients with related bone marrow failure diseases, multiple sclerosis, and type I diabetes, whereas no aCOX-2 Ab seropositivity was detected in the healthy controls, in patients with non-autoinflammatory diseases or rheumatoid arthritis. In IAA, anti-COX-2 Ab positivity correlated with age and the HLA-DRB1*15:01 genotype. 83% of the >40 years old IAA patients with HLA-DRB1*15:01 were anti-COX-2 Ab positive, indicating an excellent sensitivity in this group. aCOX-2 Ab positive IAA patients also presented lower platelet counts. Our results suggest that aCOX-2 Ab defines a distinct subgroup of IAA and may serve as a valuable disease biomarker.
Collapse
Affiliation(s)
- Tiina Kelkka
- Hematology Research Unit Helsinki, University of Helsinki and Department of Hematology, Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland.,Translational Immunology Research Program and Department of Clinical Chemistry and Hematology, University of Helsinki, Helsinki, Finland
| | - Mikko Tyster
- Hematology Research Unit Helsinki, University of Helsinki and Department of Hematology, Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland.,Translational Immunology Research Program and Department of Clinical Chemistry and Hematology, University of Helsinki, Helsinki, Finland
| | - Sofie Lundgren
- Hematology Research Unit Helsinki, University of Helsinki and Department of Hematology, Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland.,Translational Immunology Research Program and Department of Clinical Chemistry and Hematology, University of Helsinki, Helsinki, Finland
| | - Xingmin Feng
- Hematology Branch, National Heart, Lung, and Blood Institute, NIH, Bethesda, MD, USA
| | - Cassandra Kerr
- Department of Translational Hematology and Oncology Research and Leukemia Program, Department of Hematology and Medical Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Kohei Hosokawa
- Department of Hematology, Faculty of Medicine, Institute of Medical Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan
| | - Jani Huuhtanen
- Hematology Research Unit Helsinki, University of Helsinki and Department of Hematology, Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland.,Translational Immunology Research Program and Department of Clinical Chemistry and Hematology, University of Helsinki, Helsinki, Finland
| | - Mikko Keränen
- Hematology Research Unit Helsinki, University of Helsinki and Department of Hematology, Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland.,Translational Immunology Research Program and Department of Clinical Chemistry and Hematology, University of Helsinki, Helsinki, Finland
| | - Bhavisha Patel
- Hematology Branch, National Heart, Lung, and Blood Institute, NIH, Bethesda, MD, USA
| | - Toru Kawakami
- Division of Hematology, Department of Internal Medicine, Shinshu University School of Medicine, Matsumoto, Japan
| | - Yuka Maeda
- Division of Cancer Immunology, Research Institute/Exploratory Oncology Research and Clinical Trial Center, National Cancer Center Japan, Tokyo, Japan
| | - Otso Nieminen
- Hematology Research Unit Helsinki, University of Helsinki and Department of Hematology, Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland.,Translational Immunology Research Program and Department of Clinical Chemistry and Hematology, University of Helsinki, Helsinki, Finland
| | - Tiina Kasanen
- Hematology Research Unit Helsinki, University of Helsinki and Department of Hematology, Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland.,Translational Immunology Research Program and Department of Clinical Chemistry and Hematology, University of Helsinki, Helsinki, Finland
| | - Pasi Aronen
- Biostatistics Unit, Faculty of Medicine, University of Helsinki and Helsinki-Uusimaa Hospital District, Helsinki, Finland
| | - Bhagwan Yadav
- Hematology Research Unit Helsinki, University of Helsinki and Department of Hematology, Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland.,Translational Immunology Research Program and Department of Clinical Chemistry and Hematology, University of Helsinki, Helsinki, Finland
| | - Hanna Rajala
- Hematology Research Unit Helsinki, University of Helsinki and Department of Hematology, Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland.,Translational Immunology Research Program and Department of Clinical Chemistry and Hematology, University of Helsinki, Helsinki, Finland
| | - Hideyuki Nakazawa
- Department of Hematology, Shinshu University School of Medicine, Matsumoto, Japan
| | - Taina Jaatinen
- Histocompatibility Testing Laboratory, Finnish Red Cross Blood Service, Helsinki, Finland
| | - Eva Hellström-Lindberg
- Division of Hematology, Department of Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Seishi Ogawa
- Department of Pathology and Tumor Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Fumihiro Ishida
- Department of Biomedical Laboratory Sciences, Shinshu University School of Medicine, Matsumoto, Japan
| | - Hiroyoshi Nishikawa
- Division of Cancer Immunology, Research Institute/Exploratory Oncology Research and Clinical Trial Center, National Cancer Center Japan, Tokyo, Japan
| | - Shinji Nakao
- Department of Hematology, Faculty of Medicine, Institute of Medical Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan
| | - Jaroslaw Maciejewski
- Department of Translational Hematology and Oncology Research and Leukemia Program, Department of Hematology and Medical Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Neal S Young
- Hematology Branch, National Heart, Lung, and Blood Institute, NIH, Bethesda, MD, USA
| | - Satu Mustjoki
- Hematology Research Unit Helsinki, University of Helsinki and Department of Hematology, Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland. .,Translational Immunology Research Program and Department of Clinical Chemistry and Hematology, University of Helsinki, Helsinki, Finland. .,iCAN Digital Precision Cancer Medicine Flagship, Helsinki, Finland.
| |
Collapse
|
23
|
Stem cell like memory T cells: A new paradigm in cancer immunotherapy. Clin Immunol 2022; 241:109078. [PMID: 35840054 DOI: 10.1016/j.clim.2022.109078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 07/04/2022] [Accepted: 07/09/2022] [Indexed: 11/03/2022]
Abstract
Stem cell like memory T (TSCM) cells have emerged as the apex of memory T cell differentiation for their properties of self-renewal and replenishing progenies. With potent long-term persistence, proliferative capacity and antitumor activity, TSCM cells were thought to be the ideal candidate for cancer immunotherapies. Several strategies have been proposed, such as manipulations of cytokines, metabolic factors, signal pathways, and T cell receptor signal intensity, to induce more TSCM cells in vitro, in the hope that they could reach a clinical order of magnitude to provide more long-lasting and effective anti-tumor effects in vivo. In this review, we summarized the differentiation characteristics of TSCM cells and strategies to generate more TSCM cells. We focused on their roles and application in the cancer immunotherapy especially in adoptive cell transfer therapy and cancer therapeutic vaccines, and hopefully provided clues for future understanding and researches.
Collapse
|
24
|
[The characterization analysis of pathogenic T cells in immune-mediated aplastic anemia mouse model]. ZHONGHUA XUE YE XUE ZA ZHI = ZHONGHUA XUEYEXUE ZAZHI 2022; 43:587-593. [PMID: 36709137 PMCID: PMC9395574 DOI: 10.3760/cma.j.issn.0253-2727.2022.07.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Objective: This study aims, in addition to characterizing pathogenic T cells trafficking to bone marrow (BM) and other organs, to establish immune-mediated AA C.B10 mouse model by DsRed mouse (B6 background) lymph nodes (LN) cells infusion after a total body irradiation (TBI) . Methods: The C.B10 mice received a 5 Gy TBI and then were infused with DsRed mouse (B6 background) LN cells at 5×10(6)/mouse via a tail vein injection. The severity of bone marrow failure (BMF) was observed by mononuclear cell count in bone marrow and peripheral blood cell count. On days 3, 6, 9, and 12, mice were sacrificed and collected BM, spleens, LN, or thymus to analyze the dynamic change and activation status of donor T cells in these organs by a flow cytometry. At day 12, the donor-derived T cells from BM, spleens, and LN were sorted to collect the DsRed(+)CD3(+)CD4(+) T cells and DsRed(+)CD3(+)CD8(+) T cells for RNA isolation and gene expression analyses by PCR array. Results: Relative to control animals that received 5 Gy TBI without LN cell infusion, AA mice developed severe BMF with dramatic decrease in total BM cells, hemoglobin, white blood cells, and platelets in peripheral blood on days 9 and 12 after the LN cell infusion. The frequencies of DsRed(+) T cells trafficking to BM, LN, and spleens increased with time. Surprisingly, although the DsRed(+) T cells in BM increased dramatically at a level much higher than those in the spleens and LN on day 12, there were very few DsRed(+) T cells in BM on days three and six, which was significantly lower than those in spleens or LN. The frequency of DsRed(+) T cells in thymus was the lowest during the whole process. On day 12, the DsRed(+)CD3(+)CD4(+) T cells of BM, LN, and spleens from AA mice were (91.38±2.10) %, (39.78±6.98) %, and (67.87±12.77) %, respectively. On the contrary, the DsRed(+)CD3(+)CD8(+)T cells of BM, LN, and spleens were (98.21±1.49) %, (94.06±4.20) %, and (96.29±1.23) %, respectively. We assessed the donor T cell phenotypes using the CD44 and CD62L markers and found that almost all of the DsRed(+)CD4(+) or DsRed(+)CD8(+) T cells in BM were effector memory T cell phenotype from day 9 to day 12. Meanwhile, transcriptome analyses showed higher expression in CD38, IFN-γ, LAG3, CSF1, SPP1, and TNFSF13B in BM DsRed(+)CD4(+) and DsRed(+)CD8(+) T cells. However, there was a lower expression in FOXP3 and CTLA4 in BM DsRed(+)CD4(+) T cells than those in spleens and LN. Conclusions: The DsRed LN cells infusion to induce BMF in CB10 mice enabled to track the donor-derived pathogenic T cells. Besides previously published findings in this model, we demonstrated that donor CD4(+) and CD8(+) T cells primarily homed to spleens and LN, expanded and differentiated, then infiltrated in BM with a terminal effector memory phenotype. The T cells infiltrated in BM showed more activation and less immunosuppression characteristics compared to those homing to spleens and LN during the BMF development.
Collapse
|
25
|
La Manna MP, Shekarkar Azgomi M, Tamburini B, Badami GD, Mohammadnezhad L, Dieli F, Caccamo N. Phenotypic and Immunometabolic Aspects on Stem Cell Memory and Resident Memory CD8+ T Cells. Front Immunol 2022; 13:884148. [PMID: 35784300 PMCID: PMC9247337 DOI: 10.3389/fimmu.2022.884148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 05/19/2022] [Indexed: 11/13/2022] Open
Abstract
The immune system, smartly and surprisingly, saves the exposure of a particular pathogen in its memory and reacts to the pathogen very rapidly, preventing serious diseases.Immunologists have long been fascinated by understanding the ability to recall and respond faster and more vigorously to a pathogen, known as “memory”.T-cell populations can be better described by using more sophisticated techniques to define phenotype, transcriptional and epigenetic signatures and metabolic pathways (single-cell resolution), which uncovered the heterogeneity of the memory T-compartment. Phenotype, effector functions, maintenance, and metabolic pathways help identify these different subsets. Here, we examine recent developments in the characterization of the heterogeneity of the memory T cell compartment. In particular, we focus on the emerging role of CD8+ TRM and TSCM cells, providing evidence on how their immunometabolism or modulation can play a vital role in their generation and maintenance in chronic conditions such as infections or autoimmune diseases.
Collapse
Affiliation(s)
- Marco Pio La Manna
- Central Laboratory of Advanced Diagnosis and Biomedical Research (CLADIBIOR) Azienda Ospedaliera Universitaria Policlinico (A.O.U.P.) Paolo Giaccone, University of Palermo, Palermo, Italy
- Department of Biomedicine, Neurosciences and Advanced Diagnostic (Bi.N.D.), University of Palermo, Palermo, Italy
| | - Mojtaba Shekarkar Azgomi
- Central Laboratory of Advanced Diagnosis and Biomedical Research (CLADIBIOR) Azienda Ospedaliera Universitaria Policlinico (A.O.U.P.) Paolo Giaccone, University of Palermo, Palermo, Italy
- Department of Biomedicine, Neurosciences and Advanced Diagnostic (Bi.N.D.), University of Palermo, Palermo, Italy
| | - Bartolo Tamburini
- Central Laboratory of Advanced Diagnosis and Biomedical Research (CLADIBIOR) Azienda Ospedaliera Universitaria Policlinico (A.O.U.P.) Paolo Giaccone, University of Palermo, Palermo, Italy
- Department of Biomedicine, Neurosciences and Advanced Diagnostic (Bi.N.D.), University of Palermo, Palermo, Italy
| | - Giusto Davide Badami
- Central Laboratory of Advanced Diagnosis and Biomedical Research (CLADIBIOR) Azienda Ospedaliera Universitaria Policlinico (A.O.U.P.) Paolo Giaccone, University of Palermo, Palermo, Italy
- Department of Biomedicine, Neurosciences and Advanced Diagnostic (Bi.N.D.), University of Palermo, Palermo, Italy
| | - Leila Mohammadnezhad
- Central Laboratory of Advanced Diagnosis and Biomedical Research (CLADIBIOR) Azienda Ospedaliera Universitaria Policlinico (A.O.U.P.) Paolo Giaccone, University of Palermo, Palermo, Italy
- Department of Biomedicine, Neurosciences and Advanced Diagnostic (Bi.N.D.), University of Palermo, Palermo, Italy
| | - Francesco Dieli
- Central Laboratory of Advanced Diagnosis and Biomedical Research (CLADIBIOR) Azienda Ospedaliera Universitaria Policlinico (A.O.U.P.) Paolo Giaccone, University of Palermo, Palermo, Italy
- Department of Biomedicine, Neurosciences and Advanced Diagnostic (Bi.N.D.), University of Palermo, Palermo, Italy
| | - Nadia Caccamo
- Central Laboratory of Advanced Diagnosis and Biomedical Research (CLADIBIOR) Azienda Ospedaliera Universitaria Policlinico (A.O.U.P.) Paolo Giaccone, University of Palermo, Palermo, Italy
- Department of Biomedicine, Neurosciences and Advanced Diagnostic (Bi.N.D.), University of Palermo, Palermo, Italy
- *Correspondence: Nadia Caccamo,
| |
Collapse
|
26
|
You X, Yang Q, Yan K, Wang SR, Huang RR, Wang SQ, Gao CY, Li L, Lian ZX. Multi-Omics Profiling Identifies Pathways Associated With CD8 + T-Cell Activation in Severe Aplastic Anemia. Front Genet 2022; 12:790990. [PMID: 35058969 PMCID: PMC8764265 DOI: 10.3389/fgene.2021.790990] [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: 10/07/2021] [Accepted: 11/29/2021] [Indexed: 12/28/2022] Open
Abstract
Severe aplastic anemia (SAA) is an autoimmune disease characterized by immune-mediated destruction of hematopoietic stem and progenitor cells. Autoreactive CD8+ T cells have been reported as the effector cells; however, the mechanisms regulating their cell activation in SAA remain largely unknown. Here, we performed proteomics and metabolomics analyses of plasma and bone marrow supernatant, together with transcriptional analysis of CD8+ T cells from SAA patients and healthy donors, to find key pathways that are involved in pathogenic CD8+ T-cell activation. We identified 21 differential proteins and 50 differential metabolites in SAA patients that were mainly involved in energy metabolism, complement and coagulation cascades, and HIF-1α signaling pathways. Interestingly, we found that these pathways are also enriched in T cells from SAA patients by analyzing available single-cell RNA sequencing data. Moreover, CD8+ T cells from SAA patients contain a highly activated CD38+ subset, which was increased in the bone marrow of SAA patients and a murine model of SAA. This subset presented enriched genes associated with the glycolysis or gluconeogenesis pathway, HIF-1α signaling pathway, and complement associated pathways, all of which were of importance in T-cell activation. In conclusion, our study reveals new pathways that may regulate CD8+ T-cell activation in SAA patients and provides potential therapeutic targets for SAA treatment.
Collapse
Affiliation(s)
- Xing You
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, China
| | - Qiong Yang
- Chronic Disease Laboratory, School of Medicine, South China University of Technology, Guangzhou, China
| | - Kai Yan
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Song-Rong Wang
- Chronic Disease Laboratory, School of Medicine, South China University of Technology, Guangzhou, China
| | - Rong-Rong Huang
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Shun-Qing Wang
- Department of Hematology, Guangzhou First People's Hospital, The Second Affiliated Hospital of South China University of Technology, Guangzhou, China
| | - Cai-Yue Gao
- Chronic Disease Laboratory, School of Medicine, South China University of Technology, Guangzhou, China
| | - Liang Li
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, China
| | - Zhe-Xiong Lian
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, China.,Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China.,National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, China.,Key Laboratory of Biomedical Engineering of Guangdong Province, South China University of Technology, Guangzhou, China.,Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou, China.,Innovation Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, China
| |
Collapse
|
27
|
Rosenberg JM, Peters JM, Hughes T, Lareau CA, Ludwig LS, Massoth LR, Austin-Tse C, Rehm HL, Bryson B, Chen YB, Regev A, Shalek AK, Fortune SM, Sykes DB. JAK inhibition in a patient with a STAT1 gain-of-function variant reveals STAT1 dysregulation as a common feature of aplastic anemia. MED (NEW YORK, N.Y.) 2022; 3:42-57.e5. [PMID: 35590143 PMCID: PMC9123284 DOI: 10.1016/j.medj.2021.12.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 11/17/2021] [Accepted: 12/13/2021] [Indexed: 01/16/2023]
Abstract
BACKGROUND Idiopathic aplastic anemia is a potentially lethal disease, characterized by T cell-mediated autoimmune attack of bone marrow hematopoietic stem cells. Standard of care therapies (stem cell transplantation or immunosuppression) are effective but associated with a risk of serious toxicities. METHODS An 18-year-old man presented with aplastic anemia in the context of a germline gain-of-function variant in STAT1. Treatment with the JAK1 inhibitor itacitinib resulted in a rapid resolution of aplastic anemia and a sustained recovery of hematopoiesis. Peripheral blood and bone marrow samples were compared before and after JAK1 inhibitor therapy. FINDINGS Following therapy, samples showed a decrease in the plasma concentration of interferon-γ, a decrease in PD1-positive exhausted CD8+ T cell population, and a decrease in an interferon responsive myeloid population. Single-cell analysis of chromatin accessibility showed decreased accessibility of STAT1 across CD4+ and CD8+ T cells, as well as CD14+ monocytes. To query whether other cases of aplastic anemia share a similar STAT1-mediated pathophysiology, we examined a cohort of 9 patients with idiopathic aplastic anemia. Bone marrow from six of nine patients also displayed abnormal STAT1 hyper-activation. CONCLUSIONS These findings raise the possibility that STAT1 hyperactivition defines a subset of idiopathic aplastic anemia patients for whom JAK inhibition may be an efficacious therapy. FUNDING Funding was provided by the Massachusetts General Hospital Department of Medicine Pathways Program and NIH T32 AI007387. A trial registration is at https://clinicaltrials.gov/ct2/show/NCT03906318.
Collapse
Affiliation(s)
- Jacob M. Rosenberg
- Department of Medicine, Massachusetts General Hospital; Boston, MA, 02114,Ragon Institute of MGH, MIT, and Harvard; Cambridge, MA,Harvard Medical School; Boston, MA,Harvard T.H. Chan School of Public Health; Boston, MA,Corresponding Author and Lead Contact: Jacob M. Rosenberg, 55 Fruit Street, Boston, MA, 02114, USA, , 860-930-5744
| | - Joshua M. Peters
- Ragon Institute of MGH, MIT, and Harvard; Cambridge, MA,Broad Institute of MIT and Harvard; Cambridge, MA
| | - Travis Hughes
- Ragon Institute of MGH, MIT, and Harvard; Cambridge, MA,Harvard Medical School; Boston, MA,Institute for Medical Engineering and Science, Department of Chemistry, and Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology; Cambridge, MA
| | - Caleb A. Lareau
- Harvard Medical School; Boston, MA,Broad Institute of MIT and Harvard; Cambridge, MA
| | - Leif S. Ludwig
- Harvard Medical School; Boston, MA,Broad Institute of MIT and Harvard; Cambridge, MA
| | - Lucas R. Massoth
- Harvard Medical School; Boston, MA,Department of Pathology, Massachusetts General Hospital; Boston, MA
| | - Christina Austin-Tse
- Department of Medicine, Massachusetts General Hospital; Boston, MA, 02114,Center for Genomic Medicine, Massachusetts General Hospital; Boston, MA,Laboratory for Molecular Medicine, Partners Personalized Medicine; Cambridge, MA,Department of Pathology, Massachusetts General Hospital; Boston, MA
| | - Heidi L. Rehm
- Department of Medicine, Massachusetts General Hospital; Boston, MA, 02114,Harvard Medical School; Boston, MA,Broad Institute of MIT and Harvard; Cambridge, MA,Center for Genomic Medicine, Massachusetts General Hospital; Boston, MA,Department of Pathology, Massachusetts General Hospital; Boston, MA
| | - Bryan Bryson
- Ragon Institute of MGH, MIT, and Harvard; Cambridge, MA,Institute for Medical Engineering and Science, Department of Chemistry, and Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology; Cambridge, MA
| | - Yi-Bin Chen
- Department of Medicine, Massachusetts General Hospital; Boston, MA, 02114,Massachusetts General Hospital Cancer Center; Boston, MA
| | - Aviv Regev
- Broad Institute of MIT and Harvard; Cambridge, MA,Genentech, South San Francisco, CA, USA
| | - Alex K. Shalek
- Ragon Institute of MGH, MIT, and Harvard; Cambridge, MA,Harvard Medical School; Boston, MA,Institute for Medical Engineering and Science, Department of Chemistry, and Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology; Cambridge, MA,Broad Institute of MIT and Harvard; Cambridge, MA
| | - Sarah M. Fortune
- Ragon Institute of MGH, MIT, and Harvard; Cambridge, MA,Harvard Medical School; Boston, MA,Harvard T.H. Chan School of Public Health; Boston, MA
| | - David B. Sykes
- Department of Medicine, Massachusetts General Hospital; Boston, MA, 02114,Center for Regenerative Medicine, Department of Medicine, Massachusetts General Hospital; Boston, MA,Harvard Stem Cell Institute; Cambridge, MA,Massachusetts General Hospital Cancer Center; Boston, MA
| |
Collapse
|
28
|
Lambert K, Moo KG, Arnett A, Goel G, Hu A, Flynn KJ, Speake C, Wiedeman AE, Gersuk VH, Linsley PS, Greenbaum CJ, Long SA, Partridge R, Buckner JH, Khor B. Deep immune phenotyping reveals similarities between aging, Down syndrome, and autoimmunity. Sci Transl Med 2022; 14:eabi4888. [PMID: 35020411 DOI: 10.1126/scitranslmed.abi4888] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Katharina Lambert
- Center for Translational Immunology, Benaroya Research Institute at Virginia Mason, 1201 Ninth Avenue, Seattle, WA 98101, USA
| | - Keagan G Moo
- Center for Translational Immunology, Benaroya Research Institute at Virginia Mason, 1201 Ninth Avenue, Seattle, WA 98101, USA
| | - Azlann Arnett
- Center for Translational Immunology, Benaroya Research Institute at Virginia Mason, 1201 Ninth Avenue, Seattle, WA 98101, USA
| | - Gautam Goel
- Center for Systems Immunology, Benaroya Research Institute at Virginia Mason, 1201 Ninth Avenue, Seattle, WA 98101, USA
| | - Alex Hu
- Center for Systems Immunology, Benaroya Research Institute at Virginia Mason, 1201 Ninth Avenue, Seattle, WA 98101, USA
| | - Kaitlin J Flynn
- Center for Systems Immunology, Benaroya Research Institute at Virginia Mason, 1201 Ninth Avenue, Seattle, WA 98101, USA
| | - Cate Speake
- Center for Interventional Immunology, Benaroya Research Institute at Virginia Mason, 1201 Ninth Avenue, Seattle, WA 98101, USA
| | - Alice E Wiedeman
- Center for Translational Immunology, Benaroya Research Institute at Virginia Mason, 1201 Ninth Avenue, Seattle, WA 98101, USA
| | - Vivian H Gersuk
- Center for Systems Immunology, Benaroya Research Institute at Virginia Mason, 1201 Ninth Avenue, Seattle, WA 98101, USA
| | - Peter S Linsley
- Center for Systems Immunology, Benaroya Research Institute at Virginia Mason, 1201 Ninth Avenue, Seattle, WA 98101, USA
| | - Carla J Greenbaum
- Center for Interventional Immunology, Benaroya Research Institute at Virginia Mason, 1201 Ninth Avenue, Seattle, WA 98101, USA
| | - S Alice Long
- Center for Translational Immunology, Benaroya Research Institute at Virginia Mason, 1201 Ninth Avenue, Seattle, WA 98101, USA
| | - Rebecca Partridge
- Center for Translational Immunology, Benaroya Research Institute at Virginia Mason, 1201 Ninth Avenue, Seattle, WA 98101, USA.,Department of Pediatrics, Virginia Mason Medical Center, 100 N.E. Gilman Blvd., Issaquah, WA 98027, USA
| | - Jane H Buckner
- Center for Translational Immunology, Benaroya Research Institute at Virginia Mason, 1201 Ninth Avenue, Seattle, WA 98101, USA
| | - Bernard Khor
- Center for Translational Immunology, Benaroya Research Institute at Virginia Mason, 1201 Ninth Avenue, Seattle, WA 98101, USA
| |
Collapse
|
29
|
Li Y, Wu D, Yang X, Zhou S. Immunotherapeutic Potential of T Memory Stem Cells. Front Oncol 2021; 11:723888. [PMID: 34604060 PMCID: PMC8485052 DOI: 10.3389/fonc.2021.723888] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Accepted: 08/23/2021] [Indexed: 11/13/2022] Open
Abstract
Memory T cells include T memory stem cells (TSCM) and central memory T cells (TCM). Compared with effector memory T cells (TEM) and effector T cells (TEFF), they have better durability and anti-tumor immunity. Recent studies have shown that although TSCM has excellent self-renewal ability and versatility, if it is often exposed to antigens and inflammatory signals, TSCM will behave as a variety of inhibitory receptors such as PD-1, TIM-3 and LAG-3 expression, and metabolic changes from oxidative phosphorylation to glycolysis. These changes can lead to the exhaustion of T cells. Cumulative evidence in animal experiments shows that it is the least differentiated cell in the memory T lymphocyte system and is a central participant in many physiological and pathological processes in humans. It has a good clinical application prospect, so it is more and more important to study the factors affecting the formation of TSCM. This article summarizes and prospects the phenotypic and functional characteristics of TSCM, the regulation mechanism of formation, and its application in treatment of clinical diseases.
Collapse
Affiliation(s)
- Yujie Li
- Department of Biochemistry and Molecular Biology, School of Pre-Clinical Science, Guangxi Medical University, Nanning, China
| | - Dengqiang Wu
- National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Bio-targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, China
| | - Xuejia Yang
- National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Bio-targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, China
| | - Sufang Zhou
- Department of Biochemistry and Molecular Biology, School of Pre-Clinical Science, Guangxi Medical University, Nanning, China.,National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Bio-targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, China
| |
Collapse
|
30
|
Wang S, Wang L, Liu Y, Zhu Y, Liu Y. Characteristics of T-cell receptor repertoire of stem cell-like memory CD4+ T cells. PeerJ 2021; 9:e11987. [PMID: 34527440 PMCID: PMC8401816 DOI: 10.7717/peerj.11987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Accepted: 07/26/2021] [Indexed: 11/20/2022] Open
Abstract
Stem cell-like memory T cells (Tscm) combine phenotypes of naïve and memory. However, it remains unclear how T cell receptor (TCR) characteristics contribute to heterogeneity in Tscm and other memory T cells. We compared the TCR-beta (TRB) repertoire characteristics of CD4+ Tscm with those of naïve and other CD4+ memory (Tm) in 16 human subjects. Compared with Tm, Tscm had an increased diversity across all stretches of TRB repertoire structure, a skewed gene usage, and a shorter length distribution of CDR3 region. These distinctions between Tscm and Tm were enlarged in top1000 abundant clonotypes. Furthermore, top1000 clonotypes in Tscm were more public than those in Tm and grouped in more clusters, implying more epitope types recognized by top1000 clonotypes in Tscm. Importantly, self-reactive clonotypes were public and enriched in Tscm rather than Tm, of type one diabetes patients. Therefore, this study highlights the unique features of Tscm different from those of other memory subsets and provides clues to understand the physiological and pathological functions of Tscm.
Collapse
Affiliation(s)
- Shiyu Wang
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China.,BGI-Shenzhen, Shenzhen, China.,China National GeneBank, BGI-Shenzhen, Shenzhen, China
| | - Longlong Wang
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China.,BGI-Shenzhen, Shenzhen, China.,China National GeneBank, BGI-Shenzhen, Shenzhen, China
| | - Yang Liu
- BGI-Shenzhen, Shenzhen, China.,China National GeneBank, BGI-Shenzhen, Shenzhen, China
| | - Yonggang Zhu
- School of Mechanical Engineering and Automation, Harbin Institute of Technology, Shenzhen, Shenzhen, China
| | - Ya Liu
- BGI-Shenzhen, Shenzhen, China.,China National GeneBank, BGI-Shenzhen, Shenzhen, China.,Shenzhen Key Laboratory of Single-Cell Omics, BGI-Shenzhen, Shenzhen, China
| |
Collapse
|
31
|
Javan MR, Saki N, Moghimian-Boroujeni B. Aplastic anemia, cellular and molecular aspects. Cell Biol Int 2021; 45:2395-2402. [PMID: 34405925 DOI: 10.1002/cbin.11689] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 08/07/2021] [Accepted: 08/14/2021] [Indexed: 11/11/2022]
Abstract
Aplastic anemia (AA) is an autoimmune disorder characterized by bone marrow and peripheral blood pancytopenia. Different environmental and genetical conditions could be effective in an outbreak of this disease. The exact pathogenesis of this disease, however, is still idiopathic. The present study is based on Pubmed database information (2002-2021) using the words "Aplastic Anemia," "Hematopoietic Stem Cells niche," "Signaling pathway," "Cytokines," and "Immuno cells." In this disease, both hematopoietic stem cells and mesenchymal stromal cells are impaired, which is associated with impaired hematopoiesis and decreased hematopoietic cells. Inflammatory cytokines increase, which changes the ratio of T lymphocytes and leads to disease progression. In addition, the most common mechanism of AA is damage by the immune system, which leads to increased apoptosis in progenitor cells. We have shown in this review that the disease involves quantitative defects in stem cell numbers and qualitative abnormalities in the function of these cells and the activity of many different cellular and molecular factors can damage hematopoietic cells and the protective substrate of these cells in this disease.
Collapse
Affiliation(s)
- Mohammad R Javan
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Iranian Blood Transfusion Organization (IBTO), Tehran, Iran
| | - Najmaldin Saki
- Thalassemia & Hemoglobinopathy Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Bahareh Moghimian-Boroujeni
- Thalassemia & Hemoglobinopathy Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|
32
|
Wang Y, Qiu F, Xu Y, Hou X, Zhang Z, Huang L, Wang H, Xing H, Wu S. Stem cell-like memory T cells: The generation and application. J Leukoc Biol 2021; 110:1209-1223. [PMID: 34402104 DOI: 10.1002/jlb.5mr0321-145r] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 05/30/2021] [Accepted: 06/15/2021] [Indexed: 12/12/2022] Open
Abstract
Stem cell-like memory T cells (Tscm), are a newly defined memory T cell subset with characteristics of long life span, consistent self-renewing, rapid differentiation into effector T cells, and apoptosis resistance. These features indicate that Tscm have great therapeutic or preventive purposes, including being applied in chimeric Ag receptor-engineered T cells, TCR gene-modified T cells, and vaccines. However, the little knowledge about Tscm development restrains their applications. Strength and duration of TCR signaling, cytokines and metabolism in the T cells during activation all influence the Tscm development via regulating transcriptional factors and cell signaling pathways. Here, we summarize the molecular and cellular pathways involving Tscm differentiation, and its clinical application for cancer immunotherapy and prevention.
Collapse
Affiliation(s)
- Yutong Wang
- Department of Laboratory Medicine, Nanhai Hospital, Southern Medical University, Foshan, Guangdong, China.,Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Feng Qiu
- Department of Laboratory Medicine, Nanhai Hospital, Southern Medical University, Foshan, Guangdong, China
| | - Yifan Xu
- Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Xiaorui Hou
- Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Zhili Zhang
- Clinical Laboratory Department, Guangdong Women and Children Hospital, Guangzhou, Guangdong, China
| | - Lei Huang
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Framlington Place, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Huijun Wang
- Department of Laboratory Medicine, Nanhai Hospital, Southern Medical University, Foshan, Guangdong, China
| | - Hui Xing
- Department of Obstetrics and Gynecology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei, China
| | - Sha Wu
- Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
33
|
Single-cell transcriptomics dissects hematopoietic cell destruction and T-cell engagement in aplastic anemia. Blood 2021; 138:23-33. [PMID: 33763704 DOI: 10.1182/blood.2020008966] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 03/11/2021] [Indexed: 02/06/2023] Open
Abstract
Aplastic anemia (AA) is a T cell-mediated autoimmune disorder of the hematopoietic system manifested by severe depletion of the hematopoietic stem and progenitor cells (HSPCs). Nonetheless, our understanding of the complex relationship between HSPCs and T cells is still obscure, mainly limited by techniques and the sparsity of HSPCs in the context of bone marrow failure. Here we performed single-cell transcriptome analysis of residual HSPCs and T cells to identify the molecular players from patients with AA. We observed that residual HSPCs in AA exhibited lineage-specific alterations in gene expression and transcriptional regulatory networks, indicating a selective disruption of distinct lineage-committed progenitor pools. In particular, HSPCs displayed frequently altered alternative splicing events and skewed patterns of polyadenylation in transcripts related to DNA damage and repair, suggesting a likely role in AA progression to myelodysplastic syndromes. We further identified cell type-specific ligand-receptor interactions as potential mediators for ongoing HSPCs destruction by T cells. By tracking patients after immunosuppressive therapy (IST), we showed that hematopoiesis remission was incomplete accompanied by IST insensitive interactions between HSPCs and T cells as well as sustained abnormal transcription state. These data collectively constitute the transcriptomic landscape of disrupted hematopoiesis in AA at single-cell resolution, providing new insights into the molecular interactions of engaged T cells with residual HSPCs and render novel therapeutic opportunities for AA.
Collapse
|
34
|
Piotrowska M, Spodzieja M, Kuncewicz K, Rodziewicz-Motowidło S, Orlikowska M. CD160 protein as a new therapeutic target in a battle against autoimmune, infectious and lifestyle diseases. Analysis of the structure, interactions and functions. Eur J Med Chem 2021; 224:113694. [PMID: 34273660 DOI: 10.1016/j.ejmech.2021.113694] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Accepted: 07/07/2021] [Indexed: 11/25/2022]
Abstract
The glycosylphosphatidylinositol-anchored transmembrane glycoprotein CD160 (cluster of differentiation 160) is a member of the immunoglobulin superfamily. Four isoforms, which differ by the presence or absence of an immunoglobulin-like domain and the mode of anchoring in the cell membrane, have been identified. CD160 has a significant impact on the proper functioning of the immune system by activating natural killer cells and inhibiting T cells. CD160 is a natural ligand for herpes virus entry mediator (HVEM), a member of the tumor necrosis factor superfamily. The CD160-HVEM complex is a rare example of direct interaction between the two different superfamilies. The interaction of these two proteins leads to the inhibition of CD4+ T cells which, in consequence, leads to the inhibition of the correct response of the immune system. Available research articles indicate that CD160 plays a role in various types of cancer, chronic viral diseases, malaria, paroxysmal nocturnal hemoglobinuria, atherosclerosis, autoimmune diseases, skin inflammation, acute liver damage and retinal vascular disease. We present here an overview of the CD160 protein, the general characteristics of the receptor and its isoforms, details of structural studies of CD160 and the CD160-HVEM complex, as well as a description of the role of this protein in selected human diseases.
Collapse
Affiliation(s)
- Marta Piotrowska
- Department of Biomedical Chemistry, Faculty of Chemistry, University of Gdańsk, Wita Stwosza 63, 80-308, Gdańsk, Poland
| | - Marta Spodzieja
- Department of Biomedical Chemistry, Faculty of Chemistry, University of Gdańsk, Wita Stwosza 63, 80-308, Gdańsk, Poland
| | - Katarzyna Kuncewicz
- Department of Biomedical Chemistry, Faculty of Chemistry, University of Gdańsk, Wita Stwosza 63, 80-308, Gdańsk, Poland
| | - Sylwia Rodziewicz-Motowidło
- Department of Biomedical Chemistry, Faculty of Chemistry, University of Gdańsk, Wita Stwosza 63, 80-308, Gdańsk, Poland
| | - Marta Orlikowska
- Department of Biomedical Chemistry, Faculty of Chemistry, University of Gdańsk, Wita Stwosza 63, 80-308, Gdańsk, Poland.
| |
Collapse
|
35
|
Collier JL, Weiss SA, Pauken KE, Sen DR, Sharpe AH. Not-so-opposite ends of the spectrum: CD8 + T cell dysfunction across chronic infection, cancer and autoimmunity. Nat Immunol 2021; 22:809-819. [PMID: 34140679 PMCID: PMC9197228 DOI: 10.1038/s41590-021-00949-7] [Citation(s) in RCA: 124] [Impact Index Per Article: 41.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Accepted: 04/29/2021] [Indexed: 02/05/2023]
Abstract
CD8+ T cells are critical mediators of cytotoxic effector function in infection, cancer and autoimmunity. In cancer and chronic viral infection, CD8+ T cells undergo a progressive loss of cytokine production and cytotoxicity, a state termed T cell exhaustion. In autoimmunity, autoreactive CD8+ T cells retain the capacity to effectively mediate the destruction of host tissues. Although the clinical outcome differs in each context, CD8+ T cells are chronically exposed to antigen in all three. These chronically stimulated CD8+ T cells share some common phenotypic features, as well as transcriptional and epigenetic programming, across disease contexts. A better understanding of these CD8+ T cell states may reveal novel strategies to augment clearance of chronic viral infection and cancer and to mitigate self-reactivity leading to tissue damage in autoimmunity.
Collapse
Affiliation(s)
- Jenna L Collier
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, Massachusetts 02115, USA and Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women’s Hospital
| | - Sarah A Weiss
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, Massachusetts 02115, USA and Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women’s Hospital.,Department of Medical Oncology, Dana-Farber Cancer Institute, Boston MA.,Broad Institute of MIT and Harvard, Cambridge MA
| | - Kristen E Pauken
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, Massachusetts 02115, USA and Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women’s Hospital
| | - Debattama R Sen
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, Massachusetts 02115, USA and Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women’s Hospital.,Center for Cancer Research, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA
| | - Arlene H Sharpe
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, Massachusetts 02115, USA and Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women’s Hospital.,Broad Institute of MIT and Harvard, Cambridge MA
| |
Collapse
|
36
|
Lee YJ, Park EH, Park JW, Jung KC, Lee EB. Proinflammatory Features of Stem Cell-like Memory T Cells from Human Patients with Rheumatoid Arthritis. THE JOURNAL OF IMMUNOLOGY 2021; 207:381-388. [PMID: 34162725 DOI: 10.4049/jimmunol.2000814] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 05/06/2021] [Indexed: 01/01/2023]
Abstract
Stem cell-like memory T (Tscm) cells are a subset of memory T cells that have characteristics of stem cells. The characteristics of Tscm cells in patients with rheumatoid arthritis (RA) are not well known. The percentage of CD4+ and CD8+ Tscm cells in PBMCs and synovial fluid mononuclear cells was measured. After confirming the stem cell nature of Tscm cells, we examined their pathogenicity in RA patients and healthy controls (HCs) by assessing T cell activation markers and cytokine secretion after stimulation with anti-CD3/CD28 beads and/or IL-6. Finally, RNA transcriptome patterns in Tscm cells from RA patients were compared with those in HCs. In this study, the percentage of CD4+ and CD8+ Tscm cells in total T cells was significantly higher in RA patients than in HCs. Tscm cells self-proliferated and differentiated into memory and effector T cell subsets when stimulated. Compared with Tscm cells from HCs, Tscm cells from RA patients were more easily activated by anti-CD3/CD28 beads augmented by IL-6. Transcriptome analyses revealed that Tscm cells from RA patients showed a pattern distinct from those in HCs; RA-specific transcriptome patterns were not completely resolved in RA patients in complete clinical remission. In conclusion, Tscm cells from RA patients show a transcriptionally distinct pattern and are easily activated to produce inflammatory cytokines when stimulated by TCRs in the presence of IL-6. Tscm cells can be a continuous source of pathogenicity in RA.
Collapse
Affiliation(s)
- Ye Ji Lee
- Graduate Course of Translational Medicine (Immunology), Seoul National University College of Medicine, Seoul, Korea
| | - Eun Hye Park
- Division of Rheumatology, Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Jun Won Park
- Division of Rheumatology, Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Kyeong Cheon Jung
- Department of Pathology, Seoul National University College of Medicine, Seoul, Korea; and
| | - Eun Bong Lee
- Division of Rheumatology, Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea; .,Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, Korea
| |
Collapse
|
37
|
Li Y, Wan D, Guo R, Wang F, Han L, Zhang D, Xing H, Cao W, Liu Y, Xie X, Yu J, Jiang Z. Decreased bone marrow regulatory innate lymphoid cells show a distinctive miRNA profiling in aplastic anemia. ACTA ACUST UNITED AC 2021; 26:37-42. [PMID: 33375909 DOI: 10.1080/16078454.2020.1866304] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
OBJECTIVES A new regulatory subpopulation of innate lymphoid cells (ILCs), regulatory innate lymphoid cells (ILCregs), has been identified with both innate lymphoid cells and regulatory cells characteristics. The purpose of this study is to explore ILCregs and its associated miRNAs in patients with aplastic anemia (AA) by evaluating ILCregs frequency, associated miRNA quantification, and their significance. METHODS Using 4 color combinations of surface and intracellular antibody staining, the CD45+Lin-CD127+IL-10+ ILCregs from 30 healthy donors and 30 patients newly diagnosed with AA were measured by flow cytometry. Bone marrow cells were studied by next-generation sequence miRNAs quantification. RESULTS Our results showed that the frequency of ILCregs in bone marrow cells from healthy donors (HD) and AA patients were 0.703 ± 0.941 and 0.171 ± 0.233%, respectively. The frequencies of ILCregs in AA patients were significantly lower than that in HD (p <0.05). miRNA detection results showed different expression patterns in the AA patient group comparing with HD. Comparing with HD, there were 52 miRNAs up-regulated and 130 miRNAs down-regulated from AA patients. Analysis of miRNAs from ILCregs associated genes demonstrated different miRNAs expression patterns between HD and AA patient. CONCLUSION The present study demonstrated the deficiency of ILCregs and differential expression pattern of ILCregs gene-related miRNA in patients with AA. Further studies need to be done to explore the clinical significance of ILCregs and related miRNAs in patients with AA with large samples size and clinical follow-up.
Collapse
Affiliation(s)
- Yingmei Li
- Department of Hematology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, People's Republic of China
| | - Dingming Wan
- Department of Hematology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, People's Republic of China
| | - Rong Guo
- Department of Hematology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, People's Republic of China
| | - Fang Wang
- Department of Hematology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, People's Republic of China
| | - Lijie Han
- Department of Hematology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, People's Republic of China
| | - Danfeng Zhang
- Department of Hematology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, People's Republic of China
| | - Haizhou Xing
- Department of Hematology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, People's Republic of China
| | - Weijie Cao
- Department of Hematology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, People's Republic of China
| | - Yu Liu
- Department of Hematology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, People's Republic of China
| | - Xinsheng Xie
- Department of Hematology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, People's Republic of China
| | - Jifeng Yu
- Department of Hematology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, People's Republic of China.,Department of Oncology, Academy of Medical and Pharmaceutical Sciences of Zhengzhou University, Zhengzhou, People's Republic of China
| | - Zhongxing Jiang
- Department of Hematology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, People's Republic of China
| |
Collapse
|
38
|
Solorzano S, Kim J, Chen J, Feng X, Young NS. Minimal role of interleukin 6 and toll-like receptor 2 and 4 in murine models of immune-mediated bone marrow failure. PLoS One 2021; 16:e0248343. [PMID: 33711076 PMCID: PMC7954294 DOI: 10.1371/journal.pone.0248343] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Accepted: 02/24/2021] [Indexed: 02/05/2023] Open
Abstract
Immune aplastic anemia (AA) results from T cell attack on hematopoietic cells, resulting in bone marrow hypocellularity and pancytopenia. Animal models have been successfully developed to study pathophysiological mechanisms in AA. While we have systemically defined the critical components of the adaptive immune response in the pathogenesis of immune marrow failure using this model, the role of innate immunity has not been fully investigated. Here, we demonstrate that lymph node (LN) cells from B6-based donor mice carrying IL-6, TLR2, or TLR4 gene deletions were fully functional in inducing severe pancytopenia and bone marrow failure (BMF) when infused into MHC-mismatched CByB6F1 recipients. Conversely, B6-based recipient mice with IL-6, TLR2, and TLR4 deletion backgrounds were all susceptible to immune-mediated BMF relative to wild-type B6 recipients following infusion of MHC-mismatched LN cells from FVB donors, but the disease appeared more severe in IL-6 deficient mice. We conclude that IL-6, TLR2, and TLR4, molecular elements important in maintenance of normal innate immunity, have limited roles in a murine model of immune-mediated BMF. Rather, adaptive immunity appears to be the major contributor to the animal disease.
Collapse
Affiliation(s)
- Sabrina Solorzano
- Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, United States of America
- Center for Cancer and Blood Disorders, Children’s National Medical Center, Washington DC, United States of America
| | - Jisoo Kim
- Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Jichun Chen
- Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Xingmin Feng
- Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, United States of America
- * E-mail:
| | - Neal S. Young
- Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| |
Collapse
|
39
|
Abstract
Immunologic memory is the ability of adaptive immune system to quickly and specifically recognize previously encountered antigens and initiate an effector response. Alloreactive memory cells can mount rapid and robust responses to the transplanted organ resulting in allograft injury. Thus preexisting humoral or cellular memory alloresponses are typically associated with poor graft outcomes in experimental and clinical transplantation. While both B and T lymphocytes exhibit memory responses, this review discusses recent updates on the biology of memory T cells and their relevance to the field of transplantation. Three major areas of focus are the emergence and characterization of tissue resident memory T cells, manipulation of T cell metabolic pathways, and the latest promising approaches to targeting detrimental T cell memory in the settings of organ transplantation.
Collapse
|
40
|
Stem cell-like memory T cells: A perspective from the dark side. Cell Immunol 2021; 361:104273. [PMID: 33422699 DOI: 10.1016/j.cellimm.2020.104273] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2020] [Revised: 12/10/2020] [Accepted: 12/17/2020] [Indexed: 02/06/2023]
Abstract
Much attention has been paid to a newly discovered subset of memory T (TM) cells-stem cell-like memory T (TSCM) cells for their high self-renewal ability, multi-differentiation potential and long-term effector function in adoptive therapy against tumors. Despite their application in cancer therapy, an excess of TSCM cells also contributes to the persistence of autoimmune diseases for their immune memory and HIV infection as a long-lived HIV reservoir. Signaling pathways Wnt, AMPK/mTOR and NF-κB are key determinants for TM cell generation, maintenance and proinflammatory effect. In this review, we focus on the phenotypic and functional characteristics of TSCM cells and discuss their role in autoimmune diseases and HIV-1 chronic infection. Also, we explore the potential mechanism and signaling pathways involved in immune memory and look into the future therapy strategies of targeting long-lived TM cells to suppress pathogenic immune memory.
Collapse
|
41
|
Padgett LE, Dinh HQ, Wu R, Gaddis DE, Araujo DJ, Winkels H, Nguyen A, McNamara CA, Hedrick CC. Naive CD8 + T Cells Expressing CD95 Increase Human Cardiovascular Disease Severity. Arterioscler Thromb Vasc Biol 2020; 40:2845-2859. [PMID: 33054398 DOI: 10.1161/atvbaha.120.315106] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
OBJECTIVE Cardiovascular disease (CVD) remains a significant global health concern with a high degree of mortality. While CD4+ T cells have been extensively studied in CVD, the importance of CD8+ T cells in this disease, despite their abundance and increased activation in human atherosclerotic plaques, remains largely unknown. Thus, the objective of this study was to compare peripheral T-cell signatures between humans with a high (severe) risk of CVD (including myocardial infarction or stroke) and those with a low risk of CVD. Approach and Results: Using mass cytometry, we uncovered a naive CD8+ T (TN) cell population expressing CD95 (termed CD95+CD8+ stem cell memory T [CD8 TSCM] cells) that was enriched in patients with high compared with low CVD. This T-cell subset enrichment within individuals with high CVD was a relative increase and resulted from the loss of CD95lo cells within the TN compartment. We found that CD8 TSCM cells positively correlated with CVD risk in humans, while CD8+ TN cells were inversely correlated. Atherosclerotic apolipoprotein E-deficient (ApoE-/-) mice also displayed respective 7- and 2-fold increases in CD8+ TSCM frequencies within the peripheral blood and aorta-draining paraaortic lymph nodes compared with C57BL/6J mice. CD8+ TSCM cells were 1.7-fold increased in aortas from western diet fed ApoE-/- mice compared with normal laboratory diet-fed ApoE-/- mice. Importantly, transfer of TSCM cells into immune-deficient Rag.Ldlr recipient mice that lacked T cells increased atherosclerosis, illustrating the importance of these cells in atherogenesis. CONCLUSIONS CD8+ TSCM cells are increased in humans with high CVD. As these TSCM cells promote atherosclerosis, targeting them may attenuate atherosclerotic plaque progression.
Collapse
Affiliation(s)
- Lindsey E Padgett
- Division of Inflammation Biology, La Jolla Institute for Immunology, CA (L.E.P., H.Q.D., R.W., D.E.G., D.J.A., H.W., C.C.H.)
| | - Huy Q Dinh
- Division of Inflammation Biology, La Jolla Institute for Immunology, CA (L.E.P., H.Q.D., R.W., D.E.G., D.J.A., H.W., C.C.H.)
| | - Runpei Wu
- Division of Inflammation Biology, La Jolla Institute for Immunology, CA (L.E.P., H.Q.D., R.W., D.E.G., D.J.A., H.W., C.C.H.)
| | - Dalia E Gaddis
- Division of Inflammation Biology, La Jolla Institute for Immunology, CA (L.E.P., H.Q.D., R.W., D.E.G., D.J.A., H.W., C.C.H.)
| | - Daniel J Araujo
- Division of Inflammation Biology, La Jolla Institute for Immunology, CA (L.E.P., H.Q.D., R.W., D.E.G., D.J.A., H.W., C.C.H.)
| | - Holger Winkels
- Division of Inflammation Biology, La Jolla Institute for Immunology, CA (L.E.P., H.Q.D., R.W., D.E.G., D.J.A., H.W., C.C.H.)
| | - Anh Nguyen
- Cardiovascular Research Center and Division of Cardiovascular Medicine, University of Virginia, Charlottesville (A.N., C.A.M.)
| | - Coleen A McNamara
- Cardiovascular Research Center and Division of Cardiovascular Medicine, University of Virginia, Charlottesville (A.N., C.A.M.)
| | - Catherine C Hedrick
- Division of Inflammation Biology, La Jolla Institute for Immunology, CA (L.E.P., H.Q.D., R.W., D.E.G., D.J.A., H.W., C.C.H.)
| |
Collapse
|
42
|
Liu ZF, Zhang F, Guo DD, Pan XM, Bi HS. Cytotoxic effect of specific T cells from mice with experimental autoimmune uveitis on murine photoreceptor cells. Int J Ophthalmol 2020; 13:1180-1186. [PMID: 32821670 DOI: 10.18240/ijo.2020.08.02] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2019] [Accepted: 03/10/2020] [Indexed: 12/29/2022] Open
Abstract
AIM To investigate the cytotoxic effect of specific T cells from mice with experimental autoimmune uveitis (EAU) as well as their secreted interferon (IFN)-γ and interleukin (IL)-17A on murine photoreceptor (661W) cells. METHODS An EAU model was established in female mice by injection of interphotoreceptor retinoid binding protein (IRBP) emulsion supplemented with complete Freund's adjuvant (CFA) and Mycobacterium tuberculosis (TB). On day 12 after induction of EAU, specific T cells from spleen and lymph node tissues were isolated and cultured for 4d and the levels of IFN-γ and IL-17A in the supernatants were determined by enzyme-linked immunosorbent assays (ELISAs). T cells and their supernatants were added to 661W cells to observe the alteration of cell morphology; IFN-γ and IL-17A were separately added to 661W cells to observe the effect of IFN-γ and IL-17A on cell proliferation. RESULTS The levels of IFN-γ and IL-17A in the T cell supernatants were 1568.64±38.79 pg/mL and 1456.57±46.98 pg/mL, respectively. The supernatants apparently inhibited 661W cell proliferation (P<0.05). T cells could also attach to the surface of 661W cells, and IFN-γ showed a more serious cytotoxic effect on 661W cells than IL-17A, inhibiting cell proliferation (P<0.01). CONCLUSION IFN-γ and IL-17A from T cells of EAU mice model can exert cytotoxic effects on murine photoreceptor cell proliferation, and IFN-γ shows more serious cytotoxic effects on murine photoreceptor cells than IL-17A.
Collapse
Affiliation(s)
- Zheng-Feng Liu
- Medical School of Ophthalmology & Optometry, Shandong University of Traditional Chinese Medicine, Jinan 250355, Shandong Province, China
| | - Feng Zhang
- Shandong Provincial Hospital Affiliated to Shandong University, Jinan 250021, Shandong Province, China.,Shandong Maternity & Child Health Care Hospital, Jinan 250014, Shandong Province, China
| | - Da-Dong Guo
- Affiliated Eye Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250002, Shandong Province, China.,Shandong Provincial Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Therapy of Ocular Diseases; Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Therapy of Ocular Diseases in Universities of Shandong; Eye Institute of Shandong University of Traditional Chinese Medicine, Jinan 250002, Shandong Province, China
| | - Xue-Mei Pan
- Affiliated Eye Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250002, Shandong Province, China
| | - Hong-Sheng Bi
- Affiliated Eye Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250002, Shandong Province, China.,Shandong Provincial Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Therapy of Ocular Diseases; Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Therapy of Ocular Diseases in Universities of Shandong; Eye Institute of Shandong University of Traditional Chinese Medicine, Jinan 250002, Shandong Province, China
| |
Collapse
|
43
|
Brzeźniakiewicz-Janus K, Rupa-Matysek J, Gil L. Acquired Aplastic Anemia as a Clonal Disorder of Hematopoietic Stem Cells. Stem Cell Rev Rep 2020; 16:472-481. [PMID: 32270433 PMCID: PMC7253510 DOI: 10.1007/s12015-020-09971-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Aplastic anemia is rare disorder presenting with bone marrow failure syndrome due to autoimmune destruction of early hematopoietic stem cells (HSCs) and stem cell progenitors. Recent advances in newer genomic sequencing and other molecular techniques have contributed to a better understanding of the pathogenesis of aplastic anemia with respect to the inflammaging, somatic mutations, cytogenetic abnormalities and defective telomerase functions of HSCs. These have been summarized in this review and may be helpful in differentiating aplastic anemia from hypocellular myelodysplastic syndrome. Furthermore, responses to immunosuppressive therapy and outcomes may be determined by molecular pathogenesis of HSCs autoimmune destruction, as well as treatment personalization in the future.
Collapse
Affiliation(s)
- Katarzyna Brzeźniakiewicz-Janus
- Department of Hematology, Multi-Specialist Hospital Gorzów Wielkopolski, Faculty of Medicine and Health Science, University of Zielona Góra, Gorzów Wielkopolski, Poland.
| | - Joanna Rupa-Matysek
- Department of Hematology and Bone Marrow Transplantation, Poznań University of Medical Sciences, Poznań, Poland
| | - Lidia Gil
- Department of Hematology and Bone Marrow Transplantation, Poznań University of Medical Sciences, Poznań, Poland
| |
Collapse
|
44
|
Lu S, Yadav AK, Qiao X. Identification of potential miRNA–mRNA interaction network in bone marrow T cells of acquired aplastic anemia. Hematology 2020; 25:168-175. [PMID: 32338587 DOI: 10.1080/16078454.2020.1757332] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Affiliation(s)
- Shuanglong Lu
- Department of Pediatrics, Tongji Hospital, Tongji University School of Medicine, Shanghai, People’s Republic of China
| | - Anil Kumar Yadav
- Department of Pediatrics, Tongji Hospital, Tongji University School of Medicine, Shanghai, People’s Republic of China
| | - Xiaohong Qiao
- Department of Pediatrics, Tongji Hospital, Tongji University School of Medicine, Shanghai, People’s Republic of China
| |
Collapse
|
45
|
Zaimoku Y, Patel BA, Kajigaya S, Feng X, Alemu L, Quinones Raffo D, Groarke EM, Young NS. Deficit of circulating CD19 + CD24 hi CD38 hi regulatory B cells in severe aplastic anaemia. Br J Haematol 2020; 190:610-617. [PMID: 32311088 PMCID: PMC7496711 DOI: 10.1111/bjh.16651] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 03/10/2020] [Accepted: 03/18/2020] [Indexed: 12/12/2022]
Abstract
Immune aplastic anaemia (AA) is caused by cytotoxic T lymphocytes (CTLs) that destroy haematopoietic stem and progenitor cells. Enhanced type 1 T helper (Th1) responses and reduced regulatory T cells (Tregs) are involved in the immune pathophysiology. CD24hiCD38hi regulatory B cells (Bregs) suppress CTLs and Th1 responses, and induce Tregs via interleukin 10 (IL‐10). We investigated circulating B‐cell subpopulations, including CD24hiCD38hi Bregs, as well as total B cells, CD4+ T cells, CD8+ T cells and natural killer cells in 104 untreated patients with severe and very severe AA, aged ≥18 years. All patients were treated with standard immunosuppressive therapy (IST) plus eltrombopag. CD24hiCD38hi Bregs were markedly reduced in patients with AA compared to healthy individuals, especially in very severe AA, but residual Bregs remained functional, capable of producing IL‐10; total B‐cell counts and the other B‐cell subpopulations were similar to those of healthy individuals. CD24hiCD38hi Bregs did not correlate with responses to IST, and they recovered to levels present in healthy individuals after therapy. Mature naïve B‐cell counts were unexpectedly associated with IST response. Markedly reduced CD24hiCD38hi Bregs, especially in very severe AA, with recovery after IST suggest Breg deficits may contribute to the pathophysiology of immune AA.
Collapse
Affiliation(s)
- Yoshitaka Zaimoku
- Hematology Branch, National Heart, Lung, and Blood Institute, NIH, Bethesda, MD, USA
| | - Bhavisha A Patel
- Hematology Branch, National Heart, Lung, and Blood Institute, NIH, Bethesda, MD, USA
| | - Sachiko Kajigaya
- Hematology Branch, National Heart, Lung, and Blood Institute, NIH, Bethesda, MD, USA
| | - Xingmin Feng
- Hematology Branch, National Heart, Lung, and Blood Institute, NIH, Bethesda, MD, USA
| | - Lemlem Alemu
- Hematology Branch, National Heart, Lung, and Blood Institute, NIH, Bethesda, MD, USA
| | - Diego Quinones Raffo
- Hematology Branch, National Heart, Lung, and Blood Institute, NIH, Bethesda, MD, USA
| | - Emma M Groarke
- Hematology Branch, National Heart, Lung, and Blood Institute, NIH, Bethesda, MD, USA
| | - Neal S Young
- Hematology Branch, National Heart, Lung, and Blood Institute, NIH, Bethesda, MD, USA
| |
Collapse
|
46
|
Cianciotti BC, Ruggiero E, Campochiaro C, Oliveira G, Magnani ZI, Baldini M, Doglio M, Tassara M, Manfredi AA, Baldissera E, Ciceri F, Cieri N, Bonini C. CD4+ Memory Stem T Cells Recognizing Citrullinated Epitopes Are Expanded in Patients With Rheumatoid Arthritis and Sensitive to Tumor Necrosis Factor Blockade. Arthritis Rheumatol 2020; 72:565-575. [PMID: 31682074 DOI: 10.1002/art.41157] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Accepted: 10/31/2019] [Indexed: 12/17/2022]
Abstract
OBJECTIVE Memory stem T (Tscm) cells are long-lived, self-renewing T cells that play a relevant role in immunologic memory. This study was undertaken to investigate whether Tscm cells accumulate in rheumatoid arthritis (RA). METHODS The polarization and differentiation profiles of circulating T cells were assessed by flow cytometry. Antigen-specific T cells were characterized by staining with major histocompatibility complex class II tetramers. The T cell receptor (TCR) repertoire was analyzed by high-throughput sequencing using an unbiased RNA-based approach in CD4+ T cell subpopulations sorted by fluorescence-activated cell sorting. RESULTS We analyzed the dynamics of circulating Tscm cells (identified as CD45RA+CD62L+CD95+ T cells) by flow cytometry in 27 RA patients, 16 of whom were also studied during treatment with the anti-tumor necrosis factor (anti-TNF) agent etanercept. Age-matched healthy donors were used as controls. CD4+ Tscm cells were selectively and significantly expanded in RA patients in terms of frequency and absolute numbers, and significantly contracted upon anti-TNF treatment. Expanded CD4+ Tscm cells displayed a prevalent Th17 phenotype and a skewed TCR repertoire in RA patients, with the 10 most abundant clones representing up to 53.7% of the detected sequences. CD4+ lymphocytes specific for a citrullinated vimentin (Cit-vimentin) epitope were expanded in RA patients with active disease. Tscm cells accounted for a large fraction of Cit-vimentin-specific CD4+ cells. CONCLUSION Our results indicate that Tscm cells, including expanded clones specific for relevant autoantigens, accumulate in RA patients not exposed to biologic agents, and might be involved in the natural history of the disease. Further analysis of Tscm cell dynamics in autoimmune disorders may have implications for the design and efficacy assessment of innovative therapies.
Collapse
Affiliation(s)
| | | | - Corrado Campochiaro
- IRCCS Ospedale San Raffaele and Vita-Salute San Raffaele University, Milan, Italy
| | | | | | | | | | | | - Angelo A Manfredi
- IRCCS Ospedale San Raffaele and Vita-Salute San Raffaele University, Milan, Italy
| | | | - Fabio Ciceri
- IRCCS Ospedale San Raffaele and Vita-Salute San Raffaele University, Milan, Italy
| | | | - Chiara Bonini
- IRCCS Ospedale San Raffaele and Vita-Salute San Raffaele University, Milan, Italy
| |
Collapse
|
47
|
Munusamy Ponnan S, Hayes P, Fernandez N, Thiruvengadam K, Pattabiram S, Nesakumar M, Srinivasan A, Kathirvel S, Shankar J, Goyal R, Singla N, Mukherjee J, Chatrath S, Gilmour J, Subramanyam S, Prasad Tripathy S, Swaminathan S, Hanna LE. Evaluation of antiviral T cell responses and TSCM cells in volunteers enrolled in a phase I HIV-1 subtype C prophylactic vaccine trial in India. PLoS One 2020; 15:e0229461. [PMID: 32097435 PMCID: PMC7041807 DOI: 10.1371/journal.pone.0229461] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Accepted: 02/06/2020] [Indexed: 02/07/2023] Open
Abstract
T cells play an important role in controlling viral replication during HIV infection. An effective vaccine should, therefore, lead to the induction of a strong and early viral-specific CD8+ T cell response. While polyfunctional T cell responses are thought to be important contributors to the antiviral response, there is evidence to show that polyfunctional HIV- specific CD8+ T cells are just a small fraction of the total HIV-specific CD8+ T cells and may be absent in many individuals who control HIV replication, suggesting that other HIV-1 specific CD8+ effector T cell subsets may be key players in HIV control. Stem cell-like memory T cells (TSCM) are a subset of T cells with a long half-life and self-renewal capacity. They serve as key reservoirs for HIV and contribute a significant barrier to HIV eradication. The present study evaluated vaccine-induced antiviral responses and TSCM cells in volunteers vaccinated with a subtype C prophylactic HIV-1 vaccine candidate administered in a prime-boost regimen. We found that ADVAX DNA prime followed by MVA boost induced significantly more peripheral CD8+ TSCM cells and higher levels of CD8+ T cell-mediated inhibition of replication of different HIV-1 clades as compared to MVA alone and placebo. These findings are novel and provide encouraging evidence to demonstrate the induction of TSCM and cytotoxic immune responses by a subtype C HIV-1 prophylactic vaccine administered using a prime-boost strategy.
Collapse
Affiliation(s)
| | - Peter Hayes
- IAVI Human Immunology Laboratory, Imperial College, London, England, United Kingdom
| | - Natalia Fernandez
- IAVI Human Immunology Laboratory, Imperial College, London, England, United Kingdom
| | - Kannan Thiruvengadam
- National Institute for Research in Tuberculosis (Indian Council of Medical Research), Chennai, India
| | - Sathyamurthi Pattabiram
- National Institute for Research in Tuberculosis (Indian Council of Medical Research), Chennai, India
| | - Manohar Nesakumar
- National Institute for Research in Tuberculosis (Indian Council of Medical Research), Chennai, India
| | - Ashokkumar Srinivasan
- National Institute for Research in Tuberculosis (Indian Council of Medical Research), Chennai, India
| | - Sujitha Kathirvel
- National Institute for Research in Tuberculosis (Indian Council of Medical Research), Chennai, India
| | - Janani Shankar
- National Institute for Research in Tuberculosis (Indian Council of Medical Research), Chennai, India
| | - Rajat Goyal
- International AIDS Vaccine Initiative, New Delhi, India
| | - Nikhil Singla
- International AIDS Vaccine Initiative, New Delhi, India
| | | | | | - Jill Gilmour
- IAVI Human Immunology Laboratory, Imperial College, London, England, United Kingdom
| | - Sudha Subramanyam
- National Institute for Research in Tuberculosis (Indian Council of Medical Research), Chennai, India
| | - Srikanth Prasad Tripathy
- National Institute for Research in Tuberculosis (Indian Council of Medical Research), Chennai, India
| | - Soumya Swaminathan
- National Institute for Research in Tuberculosis (Indian Council of Medical Research), Chennai, India
| | - Luke Elizabeth Hanna
- National Institute for Research in Tuberculosis (Indian Council of Medical Research), Chennai, India
- * E-mail:
| |
Collapse
|
48
|
Zhao XC, Zhao L, Sun XY, Xu ZS, Ju B, Meng FJ, Zhao HG. Excellent response of severe aplastic anemia to treatment of gut inflammation: A case report and review of the literature. World J Clin Cases 2020; 8:425-435. [PMID: 32047795 PMCID: PMC7000934 DOI: 10.12998/wjcc.v8.i2.425] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Revised: 12/02/2019] [Accepted: 12/22/2019] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Cumulative evidence suggests that the aberrant immune responses in acquired aplastic anemia (AA) are sustained by active chronic infections in genetically susceptible individuals. Recently, the constant source to trigger and sustain the pathophysiology has been proposed to come from the altered gut microbiota and chronic intestinal inflammation. In this case, our serendipitous finding provides convincing evidence that the persistently dysregulated autoimmunity may be generated, at least in a significant proposition of AA patients, by the altered gut microbiota and compromised intestinal epithelium.
CASE SUMMARY A 30-year-old Chinese male patient with refractory severe AA experienced a 3-month-long febrile episode, and his fever was refractory to many kinds of injected broad-spectrum antibiotics. When presenting with abdominal cramps, he was prescribed oral mannitol and gentamycin to get rid of the gut infection. This treatment resulted in a quick resolution of the fever. Unanticipatedly, it also produced an excellent hematological response. He had undergone three episodes of recurrence within the one-year treatment, with each recurrence occurring 7-8 wk from the gastrointestinal inflammation eliminating preparations. However, subsequent treatments were able to produce subsequent remissions and consecutive treatments were successful in achieving durative hematological improvements, strongly indicating an etiological association between chronic gut inflammation and the development of AA. Interestingly, comorbid diseases superimposed on this patient (namely, psychiatric disorders, hypertension, insulin resistance, and renal dysfunction) were ameliorated together with the hematological improvements.
CONCLUSION Chronic gut inflammation may be responsible for AA pathogenesis. The comorbidities and AA may share a common etiological association.
Collapse
Affiliation(s)
- Xi-Chen Zhao
- Department of Hematology, The Central Hospital of Qingdao West Coast New Area, Qingdao 266555, Shandong Province, China
| | - Li Zhao
- Department of Hematology, The Central Hospital of Qingdao West Coast New Area, Qingdao 266555, Shandong Province, China
| | - Xiao-Yun Sun
- Department of Hematology, The Central Hospital of Qingdao West Coast New Area, Qingdao 266555, Shandong Province, China
| | - Zeng-Shan Xu
- Department of Hematology, The Central Hospital of Qingdao West Coast New Area, Qingdao 266555, Shandong Province, China
| | - Bo Ju
- Department of Hematology, The Central Hospital of Qingdao West Coast New Area, Qingdao 266555, Shandong Province, China
| | - Fan-Jun Meng
- Department of Hematology, The Affiliated Hospital of Qingdao University, Qingdao 266000, Shandong Province, China
| | - Hong-Guo Zhao
- Department of Hematology, The Affiliated Hospital of Qingdao University, Qingdao 266000, Shandong Province, China
| |
Collapse
|
49
|
Enose-Akahata Y, Oh U, Ohayon J, Billioux BJ, Massoud R, Bryant BR, Vellucci A, Ngouth N, Cortese I, Waldmann TA, Jacobson S. Clinical trial of a humanized anti-IL-2/IL-15 receptor β chain in HAM/TSP. Ann Clin Transl Neurol 2019; 6:1383-1394. [PMID: 31402625 PMCID: PMC6689682 DOI: 10.1002/acn3.50820] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Accepted: 05/19/2019] [Indexed: 12/17/2022] Open
Abstract
Objective Human T cell lymphotropic virus 1 (HTLV‐1)‐associated myelopathy/tropical spastic paraparesis (HAM/TSP) is a chronic, progressive, neurological disease. Chronic activation of CD8+ T cells, as evidenced by increased spontaneous lymphoproliferation and HTLV‐1‐specific cytotoxic T cells, has been demonstrated in HAM/TSP patients. Since IL‐2 and IL‐15 stimulate memory CD8+ T cell activity, these cytokines have been implicated in the immunopathogenesis of HAM/TSP. In this phase I trial, we evaluated the safety, pharmacokinetics, and ability of Hu‐Mikβ1, a humanized monoclonal antibody directed toward the IL‐2/IL‐15 receptor β‐chain (IL‐2/IL‐15Rβ: CD122), to saturate CD122 and regulate abnormal immune responses in patients with HAM/TSP by inhibition of IL‐15 action. Methods Hu‐Mikβ1 was administered intravenously at doses of 0.5 mg/kg, 1.0 mg/kg, or 1.5 mg/kg in a total of nine HAM/TSP patients. Five doses of Hu‐Mikβ1 were administered at 3‐week intervals. The clinical response was evaluated using standardized scales. Viral and immunologic outcome measures were examined including HTLV‐1 proviral load, T cell phenotypic analysis and spontaneous lymphoproliferation in HAM/TSP patients. Results There was no significant toxicity associated with Hu‐Mikβ1 administration in HAM/TSP patients. Saturation of CD122 by Hu‐Mikβ1 was achieved in five out of nine HAM/TSP patients. Administration of Hu‐Mikβ1 was associated with inhibition of aberrant CD8+ T cell function including spontaneous lymphoproliferation and degranulation and IFN‐γ expression, especially in HAM/TSP patients that achieved CD122 saturation. Interpretation The treatment with Hu‐Mikβ1 had a number of immunological effects on HAM/TSP patients although no clinical efficacy was observed. We also did not see any dose‐related toxicity.
Collapse
Affiliation(s)
- Yoshimi Enose-Akahata
- Viral Immunology Section, Division of Neuroimmunology and Neurovirology, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, 20892
| | - Unsong Oh
- Viral Immunology Section, Division of Neuroimmunology and Neurovirology, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, 20892
| | - Joan Ohayon
- Neuroimmunology Clinic, Division of Neuroimmunology and Neurovirology, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, 20892
| | - Bridgette Jeanne Billioux
- Viral Immunology Section, Division of Neuroimmunology and Neurovirology, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, 20892
| | - Raya Massoud
- Viral Immunology Section, Division of Neuroimmunology and Neurovirology, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, 20892
| | - Bonita R Bryant
- Lymphoid Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, 20892
| | - Ashley Vellucci
- Viral Immunology Section, Division of Neuroimmunology and Neurovirology, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, 20892
| | - Nyater Ngouth
- Viral Immunology Section, Division of Neuroimmunology and Neurovirology, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, 20892
| | - Irene Cortese
- Neuroimmunology Clinic, Division of Neuroimmunology and Neurovirology, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, 20892
| | - Thomas A Waldmann
- Lymphoid Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, 20892
| | - Steven Jacobson
- Viral Immunology Section, Division of Neuroimmunology and Neurovirology, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, 20892
| |
Collapse
|
50
|
Jimbo K, Konuma T, Watanabe E, Kohara C, Mizukami M, Nagai E, Oiwa-Monna M, Mizusawa M, Isobe M, Kato S, Takahashi S, Tojo A. T memory stem cells after allogeneic haematopoietic cell transplantation: unique long-term kinetics and influence of chronic graft-versus-host disease. Br J Haematol 2019; 186:866-878. [PMID: 31135974 DOI: 10.1111/bjh.15995] [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: 02/10/2019] [Accepted: 04/11/2019] [Indexed: 12/21/2022]
Abstract
T memory stem cells (TSCMs) are a subset of primitive T cells capable of both self-renewal and differentiation into all subsets of memory and effector T cells. Therefore, TSCMs may play a role in immune reconstitution and graft-versus-host disease (GVHD) in patients receiving allogeneic haematopoietic cell transplantation (HCT). We conducted a cross-sectional study to evaluate the proportions, absolute counts, phenotypes and functions of TSCMs in 152 adult patients without disease recurrence at least 12 months after undergoing HCT. CD4+ TSCMs were negatively correlated with number of months after transplantation in HCT patients that received cord blood transplantation, but not in patients that received bone marrow transplantation or peripheral blood stem cell transplantation. The proportions and absolute counts of CD4+ TSCMs and expression levels of inducible co-stimulator (ICOS) in CD8+ TSCMs were significantly higher in patients with mild and moderate/severe cGVHD compared to patients without cGVHD. These data suggested that, more than 12 months after allogeneic HCT, the kinetics of CD4+ TSCMs were dependent on the type of donor source, and further that CD4+ TSCMs and ICOS levels in CD8+ TSCMs were associated with cGVHD.
Collapse
Affiliation(s)
- Koji Jimbo
- Department of Haematology/Oncology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Takaaki Konuma
- Department of Haematology/Oncology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Eri Watanabe
- Department of IMSUT Clinical Flow Cytometry Laboratory, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Chisato Kohara
- Department of Haematology/Oncology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Motoko Mizukami
- Department of Laboratory Medicine, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Etsuko Nagai
- Department of Laboratory Medicine, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Maki Oiwa-Monna
- Department of Haematology/Oncology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Mai Mizusawa
- Department of Haematology/Oncology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Masamichi Isobe
- Department of Haematology/Oncology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Seiko Kato
- Department of Haematology/Oncology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Satoshi Takahashi
- Department of Haematology/Oncology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Arinobu Tojo
- Department of Haematology/Oncology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| |
Collapse
|