1
|
Erice PA, Huang X, Seasock MJ, Robertson MJ, Tung HY, Perez-Negron MA, Lotlikar SL, Corry DB, Kheradmand F, Rodriguez A. Downregulation of Mirlet7 miRNA family promotes Tc17 differentiation and emphysema via de-repression of RORγt. eLife 2024; 13:RP92879. [PMID: 38722677 PMCID: PMC11081633 DOI: 10.7554/elife.92879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/12/2024] Open
Abstract
Environmental air irritants including nanosized carbon black (nCB) can drive systemic inflammation, promoting chronic obstructive pulmonary disease (COPD) and emphysema development. The let-7 microRNA (Mirlet7 miRNA) family is associated with IL-17-driven T cell inflammation, a canonical signature of lung inflammation. Recent evidence suggests the Mirlet7 family is downregulated in patients with COPD, however, whether this repression conveys a functional consequence on emphysema pathology has not been elucidated. Here, we show that overall expression of the Mirlet7 clusters, Mirlet7b/Mirlet7c2 and Mirlet7a1/Mirlet7f1/Mirlet7d, are reduced in the lungs and T cells of smokers with emphysema as well as in mice with cigarette smoke (CS)- or nCB-elicited emphysema. We demonstrate that loss of the Mirlet7b/Mirlet7c2 cluster in T cells predisposed mice to exaggerated CS- or nCB-elicited emphysema. Furthermore, ablation of the Mirlet7b/Mirlet7c2 cluster enhanced CD8+IL17a+ T cells (Tc17) formation in emphysema development in mice. Additionally, transgenic mice overexpressing Mirlet7g in T cells are resistant to Tc17 and CD4+IL17a+ T cells (Th17) development when exposed to nCB. Mechanistically, our findings reveal the master regulator of Tc17/Th17 differentiation, RAR-related orphan receptor gamma t (RORγt), as a direct target of Mirlet7 in T cells. Overall, our findings shed light on the Mirlet7/RORγt axis with Mirlet7 acting as a molecular brake in the generation of Tc17 cells and suggest a novel therapeutic approach for tempering the augmented IL-17-mediated response in emphysema.
Collapse
Affiliation(s)
- Phillip A Erice
- Immunology Graduate Program, Baylor College of MedicineHoustonUnited States
- Department of Medicine, Immunology & Allergy Rheumatology, Baylor College of MedicineHoustonUnited States
| | - Xinyan Huang
- Department of Medicine, Immunology & Allergy Rheumatology, Baylor College of MedicineHoustonUnited States
| | - Matthew J Seasock
- Immunology Graduate Program, Baylor College of MedicineHoustonUnited States
- Department of Medicine, Immunology & Allergy Rheumatology, Baylor College of MedicineHoustonUnited States
| | - Matthew J Robertson
- Dan Duncan Comprehensive Cancer Center, Baylor College of MedicineHoustonUnited States
| | - Hui-Ying Tung
- Department of Pathology and Immunology, Baylor College of MedicineHoustonUnited States
| | - Melissa A Perez-Negron
- Department of Medicine, Immunology & Allergy Rheumatology, Baylor College of MedicineHoustonUnited States
| | - Shivani L Lotlikar
- Department of Medicine, Immunology & Allergy Rheumatology, Baylor College of MedicineHoustonUnited States
| | - David B Corry
- Department of Medicine, Immunology & Allergy Rheumatology, Baylor College of MedicineHoustonUnited States
- Department of Pathology and Immunology, Baylor College of MedicineHoustonUnited States
- Center for Translational Research on Inflammatory Diseases, Michael E Debakey, Baylor College of MedicineHoustonUnited States
| | - Farrah Kheradmand
- Department of Pathology and Immunology, Baylor College of MedicineHoustonUnited States
- Center for Translational Research on Inflammatory Diseases, Michael E Debakey, Baylor College of MedicineHoustonUnited States
- Department of Medicine, Section of Pulmonary and Critical Care, Baylor College of MedicineHoustonUnited States
| | - Antony Rodriguez
- Department of Medicine, Immunology & Allergy Rheumatology, Baylor College of MedicineHoustonUnited States
- Center for Translational Research on Inflammatory Diseases, Michael E Debakey, Baylor College of MedicineHoustonUnited States
| |
Collapse
|
2
|
Zhang Z, Zhao C, Sun L, Cheng C, Tian Q, Wu C, Xu Y, Dong X, Zhang B, Zhang L, Zhao Y. Trappc1 intrinsically prevents ferroptosis of naive T cells to avoid spontaneous autoinflammatory disease in mice. Eur J Immunol 2024; 54:e2350836. [PMID: 38234007 DOI: 10.1002/eji.202350836] [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: 10/15/2023] [Revised: 12/22/2023] [Accepted: 12/26/2023] [Indexed: 01/19/2024]
Abstract
T lymphocytes are pivotal in adaptive immunity. The role of the trafficking protein particle complex (TRAPPC) in regulating T-cell development and homeostasis is unknown. Using CD4cre -Trappc1flox/flox (Trappc1 cKO) mice, we found that Trappc1 deficiency in T cells significantly decreased cell number of naive T cells in the periphery, whereas thymic T-cell development in Trappc1 cKO mice was identical as WT mice. In the culture assays and mouse models with adoptive transfer of the sorted WT (CD45.1+ CD45.2+ ) and Trappc1 cKO naive T cells (CD45.2+ ) to CD45.1+ syngeneic mice, Trappc1-deficient naive T cells showed significantly reduced survival ability compared with WT cells. RNA-seq and molecular studies showed that Trappc1 deficiency in naive T cells reduced protein transport from the endoplasmic reticulum to the Golgi apparatus, enhanced unfolded protein responses, increased P53 transcription, intracellular Ca2+ , Atf4-CHOP, oxidative phosphorylation, and lipid peroxide accumulation, and subsequently led to ferroptosis. Trappc1 deficiency in naive T cells increased ferroptosis-related damage-associated molecular pattern molecules like high mobility group box 1 or lipid oxidation products like prostaglandin E2, leukotriene B4, leukotriene C4, and leukotriene D4. Functionally, the culture supernatant of Trappc1 cKO naive T cells significantly promoted neutrophils to express inflammatory cytokines like TNFα and IL-6, which was rescued by lipid peroxidation inhibitor Acetylcysteine. Importantly, Trappc1 cKO mice spontaneously developed severe autoinflammatory disease 4 weeks after birth. Thus, intrinsic expression of Trappc1 in naive T cells plays an integral role in maintaining T-cell homeostasis to avoid proinflammatory naive T-cell death-caused autoinflammatory syndrome in mice. This study highlights the importance of the TRAPPC in T-cell biology.
Collapse
Affiliation(s)
- Zhaoqi Zhang
- CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Chenxu Zhao
- CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Lingyun Sun
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Chen Cheng
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Qianchuan Tian
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Changhong Wu
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Yanan Xu
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Xue Dong
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Baojun Zhang
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Lianfeng Zhang
- Key Laboratory of Human Diseases Comparative Medicine, Ministry of Health, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Yong Zhao
- CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Faculty of Synthetic Biology, Shenzhen Institute of Advanced Technology, Shenzhen, China
| |
Collapse
|
3
|
Bin Masroni MS, Ling Eng GW, Jeon AJ, Gao Y, Cheng H, Leong SM, Cheong JK, Hue SSS, Tan SY. MicroRNA expression signature as a biomarker in the diagnosis of nodal T-cell lymphomas. Cancer Cell Int 2024; 24:48. [PMID: 38291429 PMCID: PMC10826179 DOI: 10.1186/s12935-024-03226-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 01/12/2024] [Indexed: 02/01/2024] Open
Abstract
BACKGROUND The diagnosis of T-cell lymphomas is typically established through a multiparameter approach that combines clinical, morphologic, immunophenotypic, and genetic features, utilizing a variety of histopathologic and molecular techniques. However, accurate diagnosis of such lymphomas and distinguishing them from reactive lymph nodes remains challenging due to their low prevalence and heterogeneous features, hence limiting the confidence of pathologists. We investigated the use of microRNA (miRNA) expression signatures as an adjunctive tool in the diagnosis and classification of T-cell lymphomas that involve lymph nodes. This study seeks to distinguish reactive lymph nodes (RLN) from two types of frequently occurring nodal T-cell lymphomas: nodal T-follicular helper (TFH) cell lymphomas (nTFHL) and peripheral T-cell lymphomas, not otherwise specified (nPTCL). METHODS From the formalin-fixed paraffin-embedded (FFPE) samples from a cohort of 88 subjects, 246 miRNAs were quantified and analyzed by differential expression. Two-class logistic regression and random forest plot models were built to distinguish RLN from the nodal T-cell lymphomas. Gene set enrichment analysis was performed on the target genes of the miRNA to identify pathways and transcription factors that may be regulated by the differentially expressed miRNAs in each subtype. RESULTS Using logistic regression analysis, we identified miRNA signatures that can distinguish RLN from nodal T-cell lymphomas (AUC of 0.92 ± 0.05), from nTFHL (AUC of 0.94 ± 0.05) and from nPTCL (AUC of 0.94 ± 0.08). Random forest plot modelling was also capable of distinguishing between RLN and nodal T-cell lymphomas, but performed worse than logistic regression. However, the miRNA signatures are not able to discriminate between nTFHL and nPTCL, owing to large similarity in miRNA expression patterns. Bioinformatic analysis of the gene targets of unique miRNA expression revealed the enrichment of both known and potentially understudied signalling pathways and genes in such lymphomas. CONCLUSION This study suggests that miRNA biomarkers may serve as a promising, cost-effective tool to aid the diagnosis of nodal T-cell lymphomas, which can be challenging. Bioinformatic analysis of differentially expressed miRNAs revealed both relevant or understudied signalling pathways that may contribute to the progression and development of each T-cell lymphoma entity. This may help us gain further insight into the biology of T-cell lymphomagenesis.
Collapse
Affiliation(s)
- Muhammad Sufyan Bin Masroni
- Department of Pathology, Yong Loo Lin School of Medicine, National University of Singapore (NUS), Singapore, 117596, Singapore
| | | | - Ah-Jung Jeon
- MiRXES Lab Pte Ltd, Singapore, 138667, Singapore
| | - Yuan Gao
- MiRXES Lab Pte Ltd, Singapore, 138667, Singapore
| | - He Cheng
- MiRXES Lab Pte Ltd, Singapore, 138667, Singapore
| | - Sai Mun Leong
- Department of Pathology, Yong Loo Lin School of Medicine, National University of Singapore (NUS), Singapore, 117596, Singapore
- NUS Centre for Cancer Research, Singapore, 117599, Singapore
| | - Jit Kong Cheong
- NUS Centre for Cancer Research, Singapore, 117599, Singapore
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore (NUS), Singapore, 117596, Singapore
| | - Susan Swee-Shan Hue
- Department of Pathology, National University Hospital, Singapore, 119077, Singapore.
- Department of Pathology, Yong Loo Lin School of Medicine, National University of Singapore (NUS), Singapore, 117596, Singapore.
- NUS Centre for Cancer Research, Singapore, 117599, Singapore.
| | - Soo Yong Tan
- Department of Pathology, National University Hospital, Singapore, 119077, Singapore.
- Department of Pathology, Yong Loo Lin School of Medicine, National University of Singapore (NUS), Singapore, 117596, Singapore.
- Advanced Molecular Pathology Laboratory, Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore, 138673, Singapore.
| |
Collapse
|
4
|
Mohammadi-Kordkhayli M, Sahraian MA, Ghorbani S, Mansouri F, Talebi F, Noorbakhsh F, Saboor-Yaraghi AA. Vitamins A and D Enhance the Expression of Ror-γ-Targeting miRNAs in a Mouse Model of Multiple Sclerosis. Mol Neurobiol 2023; 60:5853-5865. [PMID: 37353624 DOI: 10.1007/s12035-023-03427-3] [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: 01/05/2023] [Accepted: 06/05/2023] [Indexed: 06/25/2023]
Abstract
Autoreactive T cells, particularly those characterized by a Th17 phenotype, exert significant influence on the pathogenesis of multiple sclerosis (MS). The present study aimed to elucidate the impact of individual and combined administration of vitamin A and D on neuroinflammation, and microRNAs (miRNAs) involved in T helper (Th)17 development, utilizing a murine model of experimental autoimmune encephalomyelitis (EAE). EAE was induced in C57BL/6 mice, and 3 days prior to immunization, intraperitoneal injections of vitamins A and D or their combination were administered. Th17 cell percentages were determined in splenocytes utilizing intracellular staining and flow cytometry. Furthermore, the expression of Ror γ-t, miR-98-5p and Let-7a-5p, was measured in both splenocytes and spinal cord tissues using RT-PCR. Treatment with vitamin A and D resulted in a reduction in both disease severity in EAE mice. Treated mice showed a decreased frequency of Th17 cells and lower expression levels of IL17 and Ror γ-t in splenocytes and spinal cord. The spinal cord tissues and splenocytes of mice treated with vitamins A, D, and combined A+D showed a significant upregulation of miR-98-5p and Let-7a-5p compared to the EAE group. Statistical analysis indicated a strong negative correlation between miR-98-5p and Let-7a-5p levels in splenocytes and Ror-t expression. Our findings indicate that the administration of vitamins A and D exerts a suppressive effect on neuroinflammation in EAE that is associated with a reduction in the differentiation of T cells into the Th17 phenotype and is mediated by the upregulation of miR-98-5p and Let-7a-5p, which target the Ror γ-t.
Collapse
Affiliation(s)
- Marziyeh Mohammadi-Kordkhayli
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
- Hotchkiss Brain Institute and Department of Clinical Neurosciences, University of Calgary, Alberta, Canada
| | - Mohammad Ali Sahraian
- Sina MS Research Center, Sina Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Samira Ghorbani
- Hotchkiss Brain Institute and Department of Clinical Neurosciences, University of Calgary, Alberta, Canada
| | - Fatemeh Mansouri
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Farideh Talebi
- Immunoregulation Research Center, Shahed University, Tehran, Iran
| | - Farshid Noorbakhsh
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| | - Ali Akbar Saboor-Yaraghi
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
5
|
Wells AC, Hioki KA, Angelou CC, Lynch AC, Liang X, Ryan DJ, Thesmar I, Zhanybekova S, Zuklys S, Ullom J, Cheong A, Mager J, Hollander GA, Pobezinskaya EL, Pobezinsky LA. Let-7 enhances murine anti-tumor CD8 T cell responses by promoting memory and antagonizing terminal differentiation. Nat Commun 2023; 14:5585. [PMID: 37696797 PMCID: PMC10495470 DOI: 10.1038/s41467-023-40959-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 08/17/2023] [Indexed: 09/13/2023] Open
Abstract
The success of the CD8 T cell-mediated immune response against infections and tumors depends on the formation of a long-lived memory pool, and the protection of effector cells from exhaustion. The advent of checkpoint blockade therapy has significantly improved anti-tumor therapeutic outcomes by reversing CD8 T cell exhaustion, but fails to generate effector cells with memory potential. Here, using in vivo mouse models, we show that let-7 miRNAs determine CD8 T cell fate, where maintenance of let-7 expression during early cell activation results in memory CD8 T cell formation and tumor clearance. Conversely, let-7-deficiency promotes the generation of a terminal effector population that becomes vulnerable to exhaustion and cell death in immunosuppressive environments and fails to reject tumors. Mechanistically, let-7 restrains metabolic changes that occur during T cell activation through the inhibition of the PI3K/AKT/mTOR signaling pathway and production of reactive oxygen species, potent drivers of terminal differentiation and exhaustion. Thus, our results reveal a role for let-7 in the time-sensitive support of memory formation and the protection of effector cells from exhaustion. Overall, our data suggest a strategy in developing next-generation immunotherapies by preserving the multipotency of effector cells rather than enhancing the efficacy of differentiation.
Collapse
Affiliation(s)
- Alexandria C Wells
- Department of Veterinary and Animal science, University of Massachusetts, Amherst, MA, USA
| | - Kaito A Hioki
- Department of Veterinary and Animal science, University of Massachusetts, Amherst, MA, USA
- UMass Biotech Training Program (BTP), Amherst, MA, USA
| | - Constance C Angelou
- Department of Veterinary and Animal science, University of Massachusetts, Amherst, MA, USA
| | - Adam C Lynch
- Department of Veterinary and Animal science, University of Massachusetts, Amherst, MA, USA
| | - Xueting Liang
- Department of Veterinary and Animal science, University of Massachusetts, Amherst, MA, USA
| | - Daniel J Ryan
- Department of Veterinary and Animal science, University of Massachusetts, Amherst, MA, USA
| | - Iris Thesmar
- Department of Veterinary and Animal science, University of Massachusetts, Amherst, MA, USA
| | - Saule Zhanybekova
- Pediatric Immunology, Department of Biomedicine, University of Basel and University Children's Hospital Basel, Basel, Switzerland
| | - Saulius Zuklys
- Pediatric Immunology, Department of Biomedicine, University of Basel and University Children's Hospital Basel, Basel, Switzerland
| | - Jacob Ullom
- Department of Veterinary and Animal science, University of Massachusetts, Amherst, MA, USA
| | - Agnes Cheong
- Department of Veterinary and Animal science, University of Massachusetts, Amherst, MA, USA
| | - Jesse Mager
- Department of Veterinary and Animal science, University of Massachusetts, Amherst, MA, USA
| | - Georg A Hollander
- Pediatric Immunology, Department of Biomedicine, University of Basel and University Children's Hospital Basel, Basel, Switzerland
| | - Elena L Pobezinskaya
- Department of Veterinary and Animal science, University of Massachusetts, Amherst, MA, USA.
| | - Leonid A Pobezinsky
- Department of Veterinary and Animal science, University of Massachusetts, Amherst, MA, USA.
| |
Collapse
|
6
|
Ding X, Du Y, Sun B, Liu L, Le S, Wu C, Chen J, Chen X, Chen S, Xia J. MicroRNA let-7a mediates posttranscriptional inhibition of Nr4A1 and exacerbates cardiac allograft rejection. Cell Signal 2023:110783. [PMID: 37356602 DOI: 10.1016/j.cellsig.2023.110783] [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: 01/10/2023] [Revised: 05/23/2023] [Accepted: 06/22/2023] [Indexed: 06/27/2023]
Abstract
BACKGROUND Acute allograft rejection remains a major obstacle after heart transplantation, and CD4+ T cells play a crucial role in allograft rejection. Upregulation of Nr4A1 could regulate CD4+ T-cell function and alleviate allograft rejection. However, the regulatory mechanism of Nr4A1 in allograft rejection remains elusive. METHODS BCLb/c mouse hearts were transplanted into WT C57BL/6 mice, and dynamic detection of the changes in Nr4A1 expression revealed that Nr4A1 was regulated posttranscriptionally after heart transplantation. Potential upstream miRNAs of Nr4A1 were screened, and the transfection of cells with these miRNA mimics/inhibitors and dual-luciferase reporter experiments were performed to clarify the regulatory mechanism of miRNAs on Nr4A1 expression. The miRNA agomiR/antagomiR was applied in vivo to validate the role of the corresponding miRNA in heart transplantation. Finally, Nr4A1 knockout mice and an adoptive T-cell cotransfer model were used to confirm the specific effects of miRNA. RESULTS The expression of Nr4A1 protein (rather than mRNA) exhibited a trend of initially increasing and then decreasing rapidly, and this phenomenon could not be reversed by lysosomal or proteasomal inhibitors. The miRNA let-7a directly binds to the Nr4A1 3'UTR and posttranscriptionally regulates Nr4A1 expression. The let-7a antagomiR prolonged allograft survival and regulated CD4+ T-cell function by upregulating Nr4A1 protein expression in CD4+ T cells. CONCLUSIONS This study confirmed that let-7a is a potential target for interfering with Nr4A1 expression in CD4+ T cells and preventing the pathological progression of cardiac allograft rejection.
Collapse
Affiliation(s)
- Xiangchao Ding
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yifan Du
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Bing Sun
- Wuhan Third Hospital (Tongren Hospital of Wuhan University), Wuhan, China
| | - Liang Liu
- Department of Cardiovascular Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Sheng Le
- Department of Thoracic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Chuangyan Wu
- Departments of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jiuling Chen
- Departments of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xing Chen
- Department of Cardiovascular Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Shanshan Chen
- Key Laboratory for Molecular Diagnosis of Hubei Province and Central Laboratory, Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Jiahong Xia
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
7
|
Tokić S, Jirouš M, Plužarić V, Mihalj M, Šola M, Tolušić Levak M, Glavaš K, Balogh P, Štefanić M. The miR-20a/miR-92b Profile Is Associated with Circulating γδ T-Cell Perturbations in Mild Psoriasis. Int J Mol Sci 2023; 24:4323. [PMID: 36901753 PMCID: PMC10001743 DOI: 10.3390/ijms24054323] [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: 12/19/2022] [Revised: 02/17/2023] [Accepted: 02/19/2023] [Indexed: 02/24/2023] Open
Abstract
Psoriasis vulgaris (PV) is an autoinflammatory dermatosis of unknown etiology. Current evidence suggests a pathogenic role of γδT cells, but the growing complexity of this population has made the offending subset difficult to pinpoint. The work on γδTCRint and γδTCRhi subsets, which express intermediate and high levels of γδTCR at their surface, respectively, is particularly scarce, leaving their inner workings in PV essentially unresolved. We have shown here that the γδTCRint/γδTCRhi cell composition and their transcriptom are related to the differential miRNA expression by performing a targeted miRNA and mRNA quantification (RT-qPCR) in multiplexed, flow-sorted γδ blood T cells from healthy controls (n = 14) and patients with PV (n = 13). A significant loss of miR-20a in bulk γδT cells (~fourfold decrease, PV vs. controls) largely mirrored increasing Vδ1-Vδ2- and γδintVδ1-Vδ2- cell densities in the bloodstream, culminating in a relative excess of γδintVδ1-Vδ2- cells for PV. Transcripts encoding DNA-binding factors (ZBTB16), cytokine receptors (IL18R1), and cell adhesion molecules (SELPLG) were depleted in the process, closely tracking miR-20a availability in bulk γδ T-cell RNA. Compared to controls, PV was also associated with enhanced miR-92b expression (~13-fold) in bulk γδT cells that lacked association with the γδT cell composition. The miR-29a and let-7c expressions remained unaltered in case-control comparisons. Overall, our data expand the current landscape of the peripheral γδT cell composition, underlining changes in its mRNA/miRNA transcriptional circuits that may inform PV pathogenesis.
Collapse
Affiliation(s)
- Stana Tokić
- Department of Laboratory Medicine and Pharmacy, Faculty of Medicine, University of Osijek, 31000 Osijek, Croatia
| | - Maja Jirouš
- Department of Medical Chemistry, Biochemistry and Clinical Chemistry, Faculty of Medicine, University of Osijek, 31000 Osijek, Croatia
| | - Vera Plužarić
- Department of Dermatology and Venereology, University Hospital Osijek, 31000 Osijek, Croatia
| | - Martina Mihalj
- Department of Dermatology and Venereology, University Hospital Osijek, 31000 Osijek, Croatia
- Department of Physiology and Immunology, Faculty of Medicine, University of Osijek, 31000 Osijek, Croatia
| | - Marija Šola
- Department of Dermatology and Venereology, University Hospital Osijek, 31000 Osijek, Croatia
| | - Maja Tolušić Levak
- Department of Dermatology and Venereology, University Hospital Osijek, 31000 Osijek, Croatia
- Department of Histology and Embryology, Faculty of Medicine, University of Osijek, 31000 Osijek, Croatia
| | - Kristina Glavaš
- Department of Transfusion Medicine, Faculty of Medicine, University of Osijek, 31000 Osijek, Croatia
| | - Peter Balogh
- Department of Immunology and Biotechnology, Faculty of Medicine, University of Pecs, 7622 Pecs, Hungary
| | - Mario Štefanić
- Department of Nuclear Medicine and Oncology, Faculty of Medicine, University of Osijek, 31000 Osijek, Croatia
| |
Collapse
|
8
|
Rad SMAH, Halpin JC, Tawinwung S, Suppipat K, Hirankarn N, McLellan AD. MicroRNA‐mediated metabolic reprogramming of chimeric antigen receptor T cells. Immunol Cell Biol 2022; 100:424-439. [PMID: 35507473 PMCID: PMC9322280 DOI: 10.1111/imcb.12551] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 03/30/2022] [Accepted: 03/31/2022] [Indexed: 12/18/2022]
Affiliation(s)
- Seyed Mohammad Ali Hosseini Rad
- Department of Microbiology and Immunology School of Biomedical Science University of Otago Dunedin Otago New Zealand
- Center of Excellence in Immunology and Immune‐mediated Diseases Chulalongkorn University Bangkok Thailand
- Department of Microbiology Faculty of Medicine Chulalongkorn University Bangkok Thailand
| | - Joshua Colin Halpin
- Department of Microbiology and Immunology School of Biomedical Science University of Otago Dunedin Otago New Zealand
| | - Supannikar Tawinwung
- Center of Excellence in Immunology and Immune‐mediated Diseases Chulalongkorn University Bangkok Thailand
- Department of Pharmacology and Physiology Faculty of Pharmaceutical Sciences Chulalongkorn University Bangkok Thailand
| | - Koramit Suppipat
- Center of Excellence in Immunology and Immune‐mediated Diseases Chulalongkorn University Bangkok Thailand
| | - Nattiya Hirankarn
- Center of Excellence in Immunology and Immune‐mediated Diseases Chulalongkorn University Bangkok Thailand
- Department of Microbiology Faculty of Medicine Chulalongkorn University Bangkok Thailand
| | - Alexander D McLellan
- Department of Microbiology and Immunology School of Biomedical Science University of Otago Dunedin Otago New Zealand
| |
Collapse
|
9
|
Wells AC, Pobezinskaya EL, Pobezinsky LA. Non-coding RNAs in CD8 T cell biology. Mol Immunol 2020; 120:67-73. [PMID: 32085976 PMCID: PMC7093237 DOI: 10.1016/j.molimm.2020.01.023] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 01/17/2020] [Accepted: 01/29/2020] [Indexed: 12/12/2022]
Abstract
CD8 T cells are among the most vigorous soldiers of the immune system that fight viral infections and cancer. CD8 T cell development, maintenance, activation and differentiation are under the tight control of multiple transcriptional and post-transcriptional networks. Over the last two decades it has become clear that non-coding RNAs (ncRNAs), which consist of microRNAs (miRNAs) and long ncRNAs (lncRNAs), have emerged as global biological regulators. While our understanding of the function of specific miRNAs has increased since the discovery of RNA interference, it is still very limited, and the field of lncRNAs is just starting to blossom. Here we will summarize our knowledge on the role of ncRNAs in CD8 T cell biology, including differentiation into memory and exhausted cells.
Collapse
Affiliation(s)
- Alexandria C Wells
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20814, United States.
| | - Elena L Pobezinskaya
- Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, MA, 01003, United States.
| | - Leonid A Pobezinsky
- Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, MA, 01003, United States.
| |
Collapse
|
10
|
Angelou CC, Wells AC, Vijayaraghavan J, Dougan CE, Lawlor R, Iverson E, Lazarevic V, Kimura MY, Peyton SR, Minter LM, Osborne BA, Pobezinskaya EL, Pobezinsky LA. Differentiation of Pathogenic Th17 Cells Is Negatively Regulated by Let-7 MicroRNAs in a Mouse Model of Multiple Sclerosis. Front Immunol 2020; 10:3125. [PMID: 32010153 PMCID: PMC6978752 DOI: 10.3389/fimmu.2019.03125] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Accepted: 12/23/2019] [Indexed: 12/12/2022] Open
Abstract
Multiple sclerosis (MS) is a disabling demyelinating autoimmune disorder of the central nervous system (CNS) which is driven by IL-23- and IL-1β-induced autoreactive Th17 cells that traffic to the CNS and secrete proinflammatory cytokines. Th17 pathogenicity in MS has been correlated with the dysregulation of microRNA (miRNA) expression, and specific miRNAs have been shown to promote the pathogenic Th17 phenotype. In the present study, we demonstrate, using the animal model of MS, experimental autoimmune encephalomyelitis (EAE), that let-7 miRNAs confer protection against EAE by negatively regulating the proliferation, differentiation and chemokine-mediated migration of pathogenic Th17 cells to the CNS. Specifically, we found that let-7 miRNAs may directly target the cytokine receptors Il1r1 and Il23r, as well as the chemokine receptors Ccr2 and Ccr5. Therefore, our results identify a novel regulatory role for let-7 miRNAs in pathogenic Th17 differentiation during EAE development, suggesting a promising therapeutic application for disease treatment.
Collapse
Affiliation(s)
- Constance C. Angelou
- Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, MA, United States
| | - Alexandria C. Wells
- Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, MA, United States
| | - Jyothi Vijayaraghavan
- Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, MA, United States
| | - Carey E. Dougan
- Department of Chemical Engineering, University of Massachusetts, Amherst, MA, United States
| | - Rebecca Lawlor
- Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, MA, United States
| | - Elizabeth Iverson
- Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, MA, United States
| | - Vanja Lazarevic
- Experimental Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Motoko Y. Kimura
- Department of Immunology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Shelly R. Peyton
- Department of Chemical Engineering, University of Massachusetts, Amherst, MA, United States
| | - Lisa M. Minter
- Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, MA, United States
| | - Barbara A. Osborne
- Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, MA, United States
| | - Elena L. Pobezinskaya
- Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, MA, United States
| | - Leonid A. Pobezinsky
- Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, MA, United States
| |
Collapse
|