1
|
Zhang W, Kong D, Zhang X, Hu L, Nian Y, Shen Z. T cell aging and exhaustion: Mechanisms and clinical implications. Clin Immunol 2025; 275:110486. [PMID: 40120658 DOI: 10.1016/j.clim.2025.110486] [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: 01/25/2025] [Revised: 03/11/2025] [Accepted: 03/19/2025] [Indexed: 03/25/2025]
Abstract
T cell senescence and exhaustion represent critical aspects of adaptive immune system dysfunction, with profound implications for health and the development of disease prevention and therapeutic strategies. These processes, though distinct, are interconnected at the molecular level, leading to impaired effector functions and reduced proliferative capacity of T cells. Such impairments increase susceptibility to diseases and diminish the efficacy of vaccines and treatments. Importantly, T cell senescence and exhaustion can dynamically influence each other, particularly in the context of chronic diseases. A deeper understanding of the molecular mechanisms underlying T cell senescence and exhaustion, as well as their interplay, is essential for elucidating the pathogenesis of related diseases and restoring dysfunctional immune responses. This knowledge will pave the way for the development of targeted therapeutic interventions and strategies to enhance immune competence. This review aims to summarize the characteristics, mechanisms, and disease associations of T cell senescence and exhaustion, while also delineating the distinctions and intersections between these two states to enhance our comprehension.
Collapse
Affiliation(s)
- Weiqi Zhang
- School of Medicine, Nankai University, Tianjin, China; Research Institute of Transplant Medicine, Nankai University, Tianjin, China; Tianjin Key Laboratory for Organ Transplantation, Tianjin, China.
| | - Dejun Kong
- School of Medicine, Nankai University, Tianjin, China; Research Institute of Transplant Medicine, Nankai University, Tianjin, China; Tianjin Key Laboratory for Organ Transplantation, Tianjin, China.
| | - Xiaohan Zhang
- School of Medicine, Nankai University, Tianjin, China; Research Institute of Transplant Medicine, Nankai University, Tianjin, China; Tianjin Key Laboratory for Organ Transplantation, Tianjin, China.
| | - Lu Hu
- Tianjin Medical University First Central Clinical College, Tianjin, China.
| | - Yeqi Nian
- School of Medicine, Nankai University, Tianjin, China; Research Institute of Transplant Medicine, Nankai University, Tianjin, China; Tianjin Key Laboratory for Organ Transplantation, Tianjin, China; Key Laboratory of Transplant Medicine, Chinese Academy of Medical Science, Tianjin, China; Department of Kidney Transplant, Tianjin First Central Hospital, Tianjin, China.
| | - Zhongyang Shen
- School of Medicine, Nankai University, Tianjin, China; Research Institute of Transplant Medicine, Nankai University, Tianjin, China; Tianjin Key Laboratory for Organ Transplantation, Tianjin, China; Key Laboratory of Transplant Medicine, Chinese Academy of Medical Science, Tianjin, China.
| |
Collapse
|
2
|
Cimons JM, DeGolier KR, Burciaga SD, Yarnell MC, Novak AJ, Rivera-Reyes AM, Kohler ME, Fry TJ. T-bet overexpression enhances CAR T cell effector functions and antigen sensitivity. J Immunother Cancer 2025; 13:e010962. [PMID: 40246581 PMCID: PMC12007057 DOI: 10.1136/jitc-2024-010962] [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: 11/01/2024] [Accepted: 04/07/2025] [Indexed: 04/19/2025] Open
Abstract
BACKGROUND T cells modified to express a chimeric antigen receptor (CAR) are successful against B-lineage malignancies but fail to induce durable remissions in up to half of patients and have shown limited efficacy against other types of cancer. Strategies to improve CAR T cell potency and responses to low antigen densities without inducing CAR T cell dysfunction or limiting persistence are necessary to expand durability of remissions. METHODS We overexpressed T-bet in human and mouse CAR T cells to mimic exposure to signal 3 cytokines during T cell priming to promote T helper cell 1 (Th1) polarization of CD4+CAR T cells with the goal of enhancing antitumor activity. Using human CAR T cells and xenograft models we interrogated the impact of T-bet overexpression on CAR T cell antitumor activity in vitro and in vivo. We also used a syngeneic murine CAR T cell model to study the impact of T-bet overexpression on long-term persistence and secondary responses to tumor rechallenge. RESULTS T-bet overexpression reduced expression of the Th2 cytokine interleukin 4 and promoted polyfunctional production of Th1-associated cytokines in response to CAR stimulation. T-bet overexpression enhanced some effector functions in vitro but did not improve CAR T cell-mediated control of leukemia expressing high levels of antigen in vivo. T-bet overexpression also improved effector function of murine CD19 CAR T cells with no impairment to the persistence or ability of persistent CAR T cells to re-expand and clear a secondary leukemia challenge. Finally, T-bet overexpression promoted enhanced in vitro function against leukemia expressing low levels of CD19, which translated to improved control of CD19lo leukemia in vivo by human C19 CAR T cells containing a 4-1BB costimulatory domain. CONCLUSIONS Together, our data demonstrate that T-bet overexpression induces a reduction in Th2 cytokine production, an increase in polyfunctional Th1 cytokine production and enhances 4-1BB CAR T cell activity against cancers expressing low levels of target antigen without promoting a loss in functional CAR T cell persistence.
Collapse
Affiliation(s)
- Jennifer M Cimons
- Pediatrics Hematolgy/Oncology/Bone Marrow Transplant, University of Colorado Anschutz Medical Campus School of Medicine, Aurora, Colorado, USA
| | - Kole R DeGolier
- Pediatrics Hematolgy/Oncology/Bone Marrow Transplant, University of Colorado Anschutz Medical Campus School of Medicine, Aurora, Colorado, USA
| | - Samuel D Burciaga
- Pediatrics Hematolgy/Oncology/Bone Marrow Transplant, University of Colorado Anschutz Medical Campus School of Medicine, Aurora, Colorado, USA
| | - Michael C Yarnell
- Pediatrics Hematolgy/Oncology/Bone Marrow Transplant, University of Colorado Anschutz Medical Campus School of Medicine, Aurora, Colorado, USA
| | - Amanda J Novak
- Pediatrics Hematolgy/Oncology/Bone Marrow Transplant, University of Colorado Anschutz Medical Campus School of Medicine, Aurora, Colorado, USA
| | - Amalia M Rivera-Reyes
- Pediatrics Hematolgy/Oncology/Bone Marrow Transplant, University of Colorado Anschutz Medical Campus School of Medicine, Aurora, Colorado, USA
| | - M Eric Kohler
- Pediatrics Hematolgy/Oncology/Bone Marrow Transplant, University of Colorado Anschutz Medical Campus School of Medicine, Aurora, Colorado, USA
- Pediatrics, Children's Hospital Colorado, Aurora, Colorado, USA
| | - Terry J Fry
- University of Colorado Denver Children's Hospital Colorado Research Institute, Aurora, Colorado, USA
| |
Collapse
|
3
|
Hong Y, Wang Y, Shu W. Immunocyte phenotypes and childhood disease susceptibility: insights from bidirectional Mendelian randomization and implications for immunomodulatory therapies. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04091-1. [PMID: 40178601 DOI: 10.1007/s00210-025-04091-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 02/07/2025] [Accepted: 03/21/2025] [Indexed: 04/05/2025]
Abstract
Immune cells are essential for maintaining immune homeostasis during childhood and influence both growth and disease susceptibility. However, the causal relationships between immunocyte phenotypes and childhood diseases remain unclear. This study employed a two-sample Mendelian Randomization (MR) analysis to assess causal associations between 731 immunocyte phenotypes and four major childhood diseases: childhood obesity, childhood absence epilepsy, childhood asthma, and childhood allergies. Genome-wide association study (GWAS) data were used, and stringent instrumental variable (IV) selection and multiple sensitivity analyses, including MR-Egger, weighted median, and leave-one-out tests, were applied to validate the robustness of the results. Significant associations were identified between specific T cell, monocyte, and B cell phenotypes and childhood diseases. Notably, CD8bright T cells and CD19 + B cells were positively correlated with childhood obesity, while monocyte subtypes were strongly associated with asthma pathophysiology. Reverse MR analysis indicated no significant causal effects of childhood diseases on immune phenotypes, except for negative associations between childhood asthma and TCRgd AC, and childhood allergy and CD28 + CD45RA + CD4 + cells. These findings highlight the critical role of immune dysregulation in childhood disease etiology and suggest potential targets for immunomodulatory therapies. Understanding these immune-disease interactions may inform novel pharmacological interventions, particularly in immune-mediated disorders such as asthma and obesity. Further research into immune-targeted therapies could enhance treatment strategies for pediatric conditions associated with chronic inflammation and immune dysfunction.
Collapse
Affiliation(s)
- Yanggang Hong
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, 325035, Zhejiang Province, China.
| | - Yi Wang
- The First School of Medicine, Wenzhou Medical University, Wenzhou, 325035, Zhejiang Province, China
| | - Wanyi Shu
- School of Ophthalmology & Optometry, School of Biomedical Engineering, Wenzhou Medical University, Wenzhou, 325035, Zhejiang Province, China
| |
Collapse
|
4
|
Sahin F, Atasoy BT, Yalcin S, Bitirim VC. Membrane-targeted immunogenic compositions using exosome mimetic approach for vaccine development against SARS-CoV-2 and other pathogens. Sci Rep 2025; 15:10899. [PMID: 40157987 PMCID: PMC11954949 DOI: 10.1038/s41598-025-95503-y] [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: 01/21/2025] [Accepted: 03/21/2025] [Indexed: 04/01/2025] Open
Abstract
The COVID-19 pandemic has underscored the urgent need for a vaccine strategy that is safe, effective, rapid, cost-efficient, and scalable for large-scale deployment during widespread infectious outbreaks. Here, we present a new vaccination strategy that meets these critical requirements. The SARS-CoV-2 S protein consists of the S1 and S2 subunits. The S2 subunit acts as the viral cell membrane fusion protein, and mutations in its C-terminal region facilitate the transport of the entire S protein to the cell membrane. When we expressed the SARS-CoV-2 S protein with a deletion of 21 amino acids from its C-terminal region in various cell types, we observed a dense presence of the protein in the cell membrane, as determined by IHC, dot blot, and ELISA. In the cell membrane-SARS-CoV-2 S protein complex, the cell membrane functions as an exosome mimic, carrying protein antigens (S protein) in their most natural form, as no further protocols are used to attach antigens to the membrane. We demonstrate that using the membrane-S protein component as a vaccine yields a more robust and protective immune response, with enhanced safety compared to mRNA-based or inactivated virus-based vaccines against SARS-CoV-2. Additionally, we show that fusing the transmembrane domain of the Vesicular Stomatitis Virus (VSV) G protein with the SARS-CoV-2 S1 protein effectively transports the S1 protein to the cell membrane, similar to SARS-CoV-2 S Δ21. We propose that designing the S2 subunit of the SARS-CoV-2 S protein, or its analogues such as the VSV-G protein, as carriers for fusing bacterial, viral, or tumor proteins with antigenic properties-and transporting them to the cell membrane-could result in a comprehensive vaccination protocol applicable to all bacteria, viruses, and even tumors.
Collapse
Affiliation(s)
- Fikret Sahin
- Department of Medical Microbiology, University of Ankara, Ankara, 06100, Turkey.
- Faculty of Medicine, Department of Medical Microbiology, Ankara University, Morphology Building, Sihhiye, Ankara, 06100, Turkey.
| | - Buse Turegun Atasoy
- Department of Medical Microbiology, University of Ankara, Ankara, 06100, Turkey
| | - Suleyman Yalcin
- Microbiology Reference Laboratory, General Directorate of Public Health, Ankara, 06800, Turkey
| | | |
Collapse
|
5
|
Wang H, Ruan G, Li Y, Liu X. The Role and Potential Application of IL-12 in the Immune Regulation of Tuberculosis. Int J Mol Sci 2025; 26:3106. [PMID: 40243848 PMCID: PMC11988481 DOI: 10.3390/ijms26073106] [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: 02/22/2025] [Revised: 03/17/2025] [Accepted: 03/18/2025] [Indexed: 04/18/2025] Open
Abstract
Tuberculosis (TB), caused by Mycobacterium tuberculosis (Mtb), remains a significant global health challenge, affecting millions annually and leading to substantial mortality, particularly in developing countries. The pathogen's ability to persist latently and evade host immunity, combined with the emergence of drug-resistant strains, underscores the need for innovative therapeutic strategies. This review highlights the crucial role of interleukin-12 (IL-12) in coordinating immune responses against TB, focusing on its potential as an immunotherapy target. IL-12, a key Th1 cytokine, enhances cellular immunity by promoting Th1 cell differentiation and IFN-γ production, vital for Mtb clearance. By stimulating cytotoxic T lymphocytes and establishing immune memory, IL-12 supports robust host defense mechanisms. However, the complexity of IL-12 biology, including its roles in pro-inflammatory and regulatory pathways, necessitates a nuanced understanding for effective therapeutic use. Recent studies have shown how IL-12 impacts T cell synapse formation, exosome-mediated bystander activation, and interactions with other cytokines in shaping T cell memory. Genetic defects in the IL-12/IFN-γ axis link to susceptibility to mycobacterial diseases, highlighting its importance in TB immunity. The review also addresses challenges like cytokine imbalances seen in TNF-α/IFN-γ synergy, which exacerbate inflammation, and the implications for IL-12-based interventions. Research into modulating IL-12, including its use as an adjuvant and in recombinant vaccines, promises improved TB treatment outcomes and vaccine efficacy. The review concludes by stressing the need for continued investigation into IL-12's molecular mechanisms towards precision immunotherapies to combat TB and its complications.
Collapse
Affiliation(s)
- Hangxing Wang
- Division of Infectious Diseases, Department of Internal Medicine, State Key Laboratory of Complex Severe and Rare Disease, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China; (H.W.); (G.R.); (Y.L.)
| | - Guiren Ruan
- Division of Infectious Diseases, Department of Internal Medicine, State Key Laboratory of Complex Severe and Rare Disease, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China; (H.W.); (G.R.); (Y.L.)
| | - Yuanchun Li
- Division of Infectious Diseases, Department of Internal Medicine, State Key Laboratory of Complex Severe and Rare Disease, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China; (H.W.); (G.R.); (Y.L.)
| | - Xiaoqing Liu
- Division of Infectious Diseases, Department of Internal Medicine, State Key Laboratory of Complex Severe and Rare Disease, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China; (H.W.); (G.R.); (Y.L.)
- Clinical Epidemiology Unit, Peking Union Medical College, International Clinical Epidemiology Network, Beijing 100730, China
- Center for Tuberculosis Research, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| |
Collapse
|
6
|
Yamada A, Choules MP, Brightman FA, Takeshita S, Nakao S, Amino N, Nakayama T, Takeuchi M, Komatsu K, Ortega F, Mistry H, Orrell D, Chassagnole C, Bonate PL. A Multiple-Model-Informed Drug-Development Approach for Optimal Regimen Selection of an Oncolytic Virus in Combination With Pembrolizumab. CPT Pharmacometrics Syst Pharmacol 2025; 14:572-582. [PMID: 39776360 PMCID: PMC11919266 DOI: 10.1002/psp4.13297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 11/18/2024] [Accepted: 12/09/2024] [Indexed: 01/11/2025] Open
Abstract
The antitumor efficacy of an intratumoral injection of a genetically engineered oncolytic vaccinia virus carrying human IL-7 and murine IL-12 genes (hIL-7/mIL-12-VV) was demonstrated in CT26.WT-bearing mice. In the CT26.WT-bearing mouse model, the efficacy of the combination of hIL-7/mIL-12-VV plus the anti-programmed cell death protein (PD)-1 antibody was determined to be correlated with the timing of administration: greater efficacy was observed when hIL-7/mIL-12-VV was administered before the anti-PD-1 agent instead of simultaneous administration. To identify an optimal dosing regimen for first-in-human clinical trials, a multiple model-informed drug-development (MIDD) approach was used through development of a quantitative systems pharmacology (QSP) model and an agent-based model (ABM). All models were built and verified using available literature and preclinical study data. Multiple dosing scenarios were explored using virtual populations by altering the interval between hIL-7/hIL-12-VV and pembrolizumab administration. In contrast with observations from preclinical studies, both the QSP and the ABM models demonstrated no antagonistic effect on the dose-dependent antitumor efficacy of hIL-7/hIL-12-VV by pembrolizumab in simulations of clinical therapy. Based on the MIDD strategy, it was recommended that the highest dose of hIL-7/hIL-12-VV and pembrolizumab should be administered on the same day, but with pembrolizumab administration following hIL-7/hIL-12-VV administration. Multiple different modeling approaches uniquely supported and informed the first-in-human clinical trial design by guiding the optimal dose and regimen selection.
Collapse
|
7
|
Ali A, Bari MF, Arshad S, Wahid M, Safdar J, Anwar K, Farooqui WA. Tissue-resident memory T-cell expressions and their prognostic role in head and neck squamous cell carcinoma: a systematic review and meta-analysis. BMC Cancer 2025; 25:356. [PMID: 40011911 DOI: 10.1186/s12885-025-13764-2] [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: 10/22/2024] [Accepted: 02/18/2025] [Indexed: 02/28/2025] Open
Abstract
BACKGROUND CD8 + tissue-resident memory T lymphocytes (TRM) are a subset of tumor-infiltrating lymphocytes (TILs) that mediate innate immunity. Clinically, they can prevent tumor development, growth and metastasis and play a potential role in immunosurveillance and long-term immunity in head and neck squamous cell carcinoma (HNSCC). This systematic review and meta-analysis aimed to assess the prognostic significance of CD8 + TRM cells, identified by key immunophenotypic markers CD103, CD69, and CD49a linked to patient outcomes such as overall survival (OS) in HNSCC and its specified subcategory, OSCC. METHODS PubMed, Scopus, and Web of Science databases were searched systematically to include original research articles comprising cross-sectional, observational, experimental studies, and clinical trials. The characteristics of the studies were recorded for years of publication, research design, cancer types, HPV status, staging, diagnostic assays, immunophenotypic markers, and immune response regulators. Hazard ratios (HR) with confidence intervals (CI) and p-values were extracted for observing the association between CD103, CD69, and/or CD49a exhibiting CD8 + cytotoxic T lymphocytes with tissue-resident memory potential. The proportion of CD8 + TRM cells co-expressing CD103, CD69, and/or CD49a was estimated by extracting the actual percentage of expression in TME from graphical presentation of data in included studies. RESULTS Among the 276 studies, 11 studies were included by reviewing the abstract or title and full-text articles. The findings of these studies demonstrated a strong association between CD8 + TRM cells, characterized by the expression of CD103, CD69, or CD49a and improved OS in patients with HNSCC, and its subtype, OSCC. Notably, similar trends were observed within the included studies relative to oropharyngeal squamous cell carcinomas (OPSCC), another recognized subtype of HNSCC. The pooled HR was 0.49 (95% CI: 0.23-1.02, p < 0.001), indicating a potential prognostic benefit of CD8 + TRM cell infiltration in HNSCC and related subtypes of OSCC and OPSCC. However, the overall pooled findings at aggregate cancer incidences were not statistically significant (p > 0.05). CONCLUSION Increased infiltration of CD8 + TRM cells expressing CD103, CD69, and/or CD49a is associated with better prognosis and OS in HNSCC and its subtype, OSCC. TRIAL REGISTRATION This systematic review and meta-analysis were registered in the international database of systematic review protocols at https://www.crd.york.ac.uk/prospero/ under protocol identifier: CRD42024570177.
Collapse
Affiliation(s)
- Anwar Ali
- Department of Oral and Maxillofacial Surgery, Dr. Ishrat-Ul-Ebad Khan Institute of Oral Health Sciences, Dow University of Health Sciences, Karachi, Pakistan.
| | - Muhammad Furqan Bari
- Department of Pathology, Dr. Ishrat-Ul-Ebad Khan Institute of Oral Health Sciences, Dow University of Health Sciences, Karachi, Pakistan
| | - Saba Arshad
- Department of Oral Biology, Dr. Ishrat-Ul-Ebad, Khan Institute of Oral Health Sciences, Dow University of Health Sciences, Karachi, Pakistan.
| | - Mohsin Wahid
- Department of Pathology, Dow International Medical College, Dow Research Institute of Biotechnology and Biomedical Sciences, Dow University of Health Sciences, Karachi, Pakistan
| | - Jawad Safdar
- Department of Oral and Maxillofacial Surgery, Dow Dental College, Dow University of Health Sciences, Karachi, Pakistan
| | - Khadija Anwar
- Dow International Medical College, Dow University of Health Sciences, Karachi, Pakistan
| | - Waqas Ahmed Farooqui
- Department of Research, School of Public Health, Dow University of Health Sciences Karachi, Karachi, Pakistan
| |
Collapse
|
8
|
Wu X, Jiang D, Du Y, Chen C, Cao K, Yang M, Chen M, Zhou W, Qi J, Yan D, Miao Z, Yang S. Immune memory in hepatitis E virus: a comparative study of natural infection and vaccination in a nursing home population. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2025:vkae026. [PMID: 40073099 DOI: 10.1093/jimmun/vkae026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 11/20/2024] [Indexed: 03/14/2025]
Abstract
Immune memory is crucial for preventing hepatitis E virus (HEV) infection. Our study aims to investigate immunological memory characteristics and differences between vaccination and natural HEV infection, taking into account that both can induce immune memory. We recruited 60 HEV-infected patients, 58 contingency HEV-vaccinated individuals and 4 controls from an outbreak of hepatitis E in a nursing home between June and August 2023. Multicolor flow cytometry, ELISA, and quantitative polymerase chain reaction (qPCR) were employed to detect memory T-cell expression profiles, HEV-specific antibodies and cytokine expression. We observed that the level of HEV-specific IgM in acute jaundiced hepatitis E patients was greater than that in non-jaundiced patients (8.37 ± 1.27 vs. 4.27 ± 0.81, P < 0.05). No significant differences were detected in the HEV-specific IgG and memory T cell expression profiles among the different severities of hepatitis E. The percentage of CD8+ TEM at 6 months after recovery was significantly greater than that in acute jaundice patients (1.60% ± 0.30% vs. 1.15% ± 0.35%, P < 0.05). Compared with natural infection, three-dose vaccination increased the level of HEV-specific IgG (14.97 ± 0.21 vs. 12.75 ± 0.37, P < 0.05), IL-7 and IL-15 (28.50 ± 3.82 vs. 23.32 ± 6.37, 608.60 ± 143.30 vs. 257.50 ± 69.87, P < 0.05). Natural infection could effectively establish immune memory. During convalescence, infection severity impacted only HEV-specific IgM, while HEV-specific IgG and memory T-cell expression profiles were not affected. Compared with natural infection, three-dose vaccination results in equal effective humoral immune memory and weaker cellular immune memory with minimal side effects.
Collapse
Affiliation(s)
- Xiaoyue Wu
- Department of Emergency Medicine, Second Affiliated Hospital, Department of Epidemiology and Biostatistics, School of Public Health, The Key Laboratory of Intelligent Preventive Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Daixi Jiang
- Department of Emergency Medicine, Second Affiliated Hospital, Department of Epidemiology and Biostatistics, School of Public Health, The Key Laboratory of Intelligent Preventive Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Yuxia Du
- Department of Emergency Medicine, Second Affiliated Hospital, Department of Epidemiology and Biostatistics, School of Public Health, The Key Laboratory of Intelligent Preventive Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Can Chen
- Department of Emergency Medicine, Second Affiliated Hospital, Department of Epidemiology and Biostatistics, School of Public Health, The Key Laboratory of Intelligent Preventive Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Kexin Cao
- Department of Emergency Medicine, Second Affiliated Hospital, Department of Epidemiology and Biostatistics, School of Public Health, The Key Laboratory of Intelligent Preventive Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Mengya Yang
- Department of Emergency Medicine, Second Affiliated Hospital, Department of Epidemiology and Biostatistics, School of Public Health, The Key Laboratory of Intelligent Preventive Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Mengsha Chen
- Department of Emergency Medicine, Second Affiliated Hospital, Department of Epidemiology and Biostatistics, School of Public Health, The Key Laboratory of Intelligent Preventive Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Wenkai Zhou
- Department of Emergency Medicine, Second Affiliated Hospital, Department of Epidemiology and Biostatistics, School of Public Health, The Key Laboratory of Intelligent Preventive Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Jiaxing Qi
- Department of Emergency Medicine, Second Affiliated Hospital, Department of Epidemiology and Biostatistics, School of Public Health, The Key Laboratory of Intelligent Preventive Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Dong Yan
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ziping Miao
- Zhejiang Provincial Centre for Disease Control and Prevention, Hangzhou, China
| | - Shigui Yang
- Department of Emergency Medicine, Second Affiliated Hospital, Department of Epidemiology and Biostatistics, School of Public Health, The Key Laboratory of Intelligent Preventive Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, 310058, China
| |
Collapse
|
9
|
Zheng S, Che X, Zhang K, Bai Y, Deng H. Potentiating CAR-T cell function in the immunosuppressive tumor microenvironment by inverting the TGF-β signal. Mol Ther 2025; 33:688-702. [PMID: 39673127 PMCID: PMC11853376 DOI: 10.1016/j.ymthe.2024.12.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 10/05/2024] [Accepted: 12/10/2024] [Indexed: 12/16/2024] Open
Abstract
The immunosuppressive tumor microenvironment represents a key challenge for chimeric antigen receptor (CAR) T cells in solid tumors and includes the production of the inhibitory cytokine transforming growth factor β (TGF-β), which limits CAR-T cell persistence and function. Current strategies involving the blockade of TGF-β signaling have little benefit for solid tumor treatment. Here, we demonstrate a novel inverted cytokine receptor (ICR)-modified CAR-T cell strategy not only TGF-β signal blockade but also antitumor efficacy enhancement. The newly designed T cells carry an ICR construct that fuses the TGF-β receptor II extracellular domain to the interleukin-15 (IL-15) receptor α cytoplasmic domain (named TB15) and is directed to the tumor antigen epidermal growth factor receptor by a CAR construct. In mice with high TGF-β solid tumors, our signal-inverted CAR/TB15 T cells effectively treat tumors by blocking TGF-β and repurposing IL-15 stimulative signaling, resulting in enhanced CAR-T cell persistence and function. As a proof of concept, our study results extend synthetic receptor signaling beyond CAR-directed killing, which could endow adoptively transferred T cells with new functions that overcome major barriers in the treatment of solid tumors by using a chimeric ICR.
Collapse
Affiliation(s)
- Shen Zheng
- Department of Cell Biology and Stem Cell Research Center, School of Basic Medical Sciences, State Key Laboratory of Natural and Biomimetic Drugs, Peking University Health Science Center, Peking University, Beijing 100191, China
| | - Xuan Che
- Department of Cell Biology and Stem Cell Research Center, School of Basic Medical Sciences, State Key Laboratory of Natural and Biomimetic Drugs, Peking University Health Science Center, Peking University, Beijing 100191, China
| | - Kai Zhang
- Department of Dermatology and Venerology, Peking University First Hospital, National Clinical Research Center for Skin and Immune Diseases, Beijing Key Laboratory of Molecular Diagnosis of Dermatoses, Peking University, Beijing, China; Institute of Advanced Clinical Medicine, Peking University, Beijing, China
| | - Yun Bai
- Department of Cell Biology and Stem Cell Research Center, School of Basic Medical Sciences, State Key Laboratory of Natural and Biomimetic Drugs, Peking University Health Science Center, Peking University, Beijing 100191, China.
| | - Hongkui Deng
- Department of Cell Biology and Stem Cell Research Center, School of Basic Medical Sciences, State Key Laboratory of Natural and Biomimetic Drugs, Peking University Health Science Center, Peking University, Beijing 100191, China; MOE Key Laboratory of Cell Proliferation and Differentiation, College of Life Sciences, Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100191, China; Shenzhen Stem Cell Engineering Laboratory, Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen 518055, China.
| |
Collapse
|
10
|
Sun Z, Lin J, Sun X, Yun Z, Zhang X, Xu S, Duan J, Yao K. Bioinformatics combining machine learning and single-cell sequencing analysis to identify common mechanisms and biomarkers of rheumatoid arthritis and ischemic heart failure. Heliyon 2025; 11:e41641. [PMID: 39897930 PMCID: PMC11783397 DOI: 10.1016/j.heliyon.2025.e41641] [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: 12/14/2023] [Revised: 12/25/2024] [Accepted: 01/02/2025] [Indexed: 02/04/2025] Open
Abstract
Patients with rheumatoid arthritis (RA) have an increased risk of ischemic heart failure (IHF), but the shared mechanisms are unclear. This study analyzed RNA sequencing data from five RA and IHF datasets to identify common biological mechanisms and significant biomarkers. One hundred and seventy-seven common differentially expressed genes (CDEGs) were identified, with enrichment analysis highlighting pathways related to sarcomere organization, ventricular myocardial tissue morphogenesis, chondrocyte differentiation, prolactin signaling, hematopoietic cell lineage, and protein methyltransferases. Five hub genes (CD2, CD3D, CCL5, IL7R, and SPATA18) were identified through protein-protein interaction (PPI) network analysis and machine learning. Co-expression and immune cell infiltration analyses underscored the importance of the inflammatory immune response, with hub genes showing significant correlations with plasma cells, activated CD4+ T memory cells, monocytes, and T regulatory cells. Single-cell RNA sequencing (scRNA-seq) confirmed hub gene expression primarily in T cells, activated T cells, monocytes, and NK cells. The findings underscore the critical roles of sarcomere organization, prolactin signaling, protein methyltransferase activity, and immune responses in the progression of IHF in RA patients. These insights not only identify valuable biomarkers and therapeutic targets but also offer promising directions for early diagnosis, personalized treatments, and preventive strategies for IHF in the context of RA. Moreover, the results highlight opportunities for repurposing existing drugs and developing new therapeutic interventions, which could reduce the risk of IHF in RA patients and improve their overall prognosis.
Collapse
Affiliation(s)
- Ziyi Sun
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, No.5, Beixiangge, Xicheng District, Beijing, 100053, People's Republic of China
- Graduate School, Beijing University of Chinese Medicine, No.11 Beisanhuan East Road, Chaoyang District, Beijing, 100029, People's Republic of China
| | - Jianguo Lin
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, No.5, Beixiangge, Xicheng District, Beijing, 100053, People's Republic of China
| | - Xiaoning Sun
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, No.5, Beixiangge, Xicheng District, Beijing, 100053, People's Republic of China
| | - Zhangjun Yun
- Graduate School, Beijing University of Chinese Medicine, No.11 Beisanhuan East Road, Chaoyang District, Beijing, 100029, People's Republic of China
| | - Xiaoxiao Zhang
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, No.5, Beixiangge, Xicheng District, Beijing, 100053, People's Republic of China
| | - Siyu Xu
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, No.5, Beixiangge, Xicheng District, Beijing, 100053, People's Republic of China
- Graduate School, Beijing University of Chinese Medicine, No.11 Beisanhuan East Road, Chaoyang District, Beijing, 100029, People's Republic of China
| | - Jinlong Duan
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, No.5, Beixiangge, Xicheng District, Beijing, 100053, People's Republic of China
| | - Kuiwu Yao
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, No.5, Beixiangge, Xicheng District, Beijing, 100053, People's Republic of China
- Academic Administration Office, China Academy of Chinese Medical Sciences, No. 16, Nanxiaojie, Inside Dongzhimen, Dongcheng District, Beijing, 100700, People's Republic of China
| |
Collapse
|
11
|
Ma R, Li Z, Tang H, Wu X, Tian L, Shah Z, Liu N, Barr T, Zhang J, Wang S, Swaminathan S, Marcucci G, Peng Y, Caligiuri MA, Yu J. NKp46 enhances type 1 innate lymphoid cell proliferation and function and anti-acute myeloid leukemia activity. Nat Commun 2025; 16:989. [PMID: 39856052 PMCID: PMC11760942 DOI: 10.1038/s41467-025-55923-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 01/02/2025] [Indexed: 01/27/2025] Open
Abstract
NKp46 is a critical regulator of natural killer (NK) cell immunity, but its function in non-NK innate immune cells remains unclear. Here, we show that NKp46 is indispensable for expressing IL-2 receptor-α (IL-2Rα) by non-NK liver-resident type-1 innate lymphoid cells (ILC1s). Deletion of NKp46 reduces IL-2Rα on ILC1s by downregulating NF-κB signaling, thus impairing ILC1 proliferation and cytotoxicity in vitro and in vivo. The binding of anti-NKp46 antibody to NKp46 triggers the activation of NF-κB, the expression of IL-2Rα, interferon-γ (IFN-γ), tumor necrosis factor (TNF), proliferation, and cytotoxicity. Functionally, NKp46 expressed on mouse ILC1s interacts with tumor cells through cell-cell contact, increasing ILC1 production of IFN-γ and TNF, and enhancing cytotoxicity. In a mouse model of acute myeloid leukemia, deletion of NKp46 impairs the ability of ILC1s to control tumor growth and reduces survival. This can be reversed by injecting NKp46+ ILC1s into NKp46 knock-out mice. Human NKp46+ ILC1s exhibit stronger cytokine production and cytotoxicity than their NKp46- counterparts, suggesting that NKp46 plays a similar role in humans. These findings identify an NKp46-NF-κB-IL-2Rα axis and suggest that activating NKp46 with an anti-NKp46 antibody may provide a potential strategy for anti-tumor innate immunity.
Collapse
MESH Headings
- Animals
- Natural Cytotoxicity Triggering Receptor 1/genetics
- Natural Cytotoxicity Triggering Receptor 1/metabolism
- Natural Cytotoxicity Triggering Receptor 1/immunology
- Immunity, Innate
- Cell Proliferation
- Mice
- Lymphocytes/immunology
- Lymphocytes/metabolism
- Humans
- Leukemia, Myeloid, Acute/immunology
- Leukemia, Myeloid, Acute/pathology
- Leukemia, Myeloid, Acute/genetics
- NF-kappa B/metabolism
- Mice, Knockout
- Mice, Inbred C57BL
- Interferon-gamma/metabolism
- Interferon-gamma/immunology
- Killer Cells, Natural/immunology
- Tumor Necrosis Factor-alpha/metabolism
- Antigens, Ly/genetics
- Antigens, Ly/metabolism
- Antigens, Ly/immunology
- Cell Line, Tumor
- Cytotoxicity, Immunologic
- Signal Transduction
- Liver/immunology
Collapse
Affiliation(s)
- Rui Ma
- Center for Molecular Oncology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, China
- Department of Hematology & Hematopoietic Cell Transplantation, City of Hope National Medical Center, Los Angeles, CA, 91010, USA
| | - Zhenlong Li
- Department of Hematology & Hematopoietic Cell Transplantation, City of Hope National Medical Center, Los Angeles, CA, 91010, USA
| | - Hejun Tang
- Department of Hematology & Hematopoietic Cell Transplantation, City of Hope National Medical Center, Los Angeles, CA, 91010, USA
| | - Xiaojin Wu
- The First Affiliated Hospital of Soochow University, Suzhou, 215005, China
| | - Lei Tian
- Division of Hematology & Oncology, Department of Medicine, School of Medicine, University of California, Irvine, CA, 92697, USA
- The Clemons Family Center for Transformative Cancer Research, University of California, Irvine, CA, 92697, USA
| | - Zahir Shah
- Department of Hematology & Hematopoietic Cell Transplantation, City of Hope National Medical Center, Los Angeles, CA, 91010, USA
| | - Ningyuan Liu
- Department of Hematology & Hematopoietic Cell Transplantation, City of Hope National Medical Center, Los Angeles, CA, 91010, USA
| | - Tasha Barr
- Department of Hematology & Hematopoietic Cell Transplantation, City of Hope National Medical Center, Los Angeles, CA, 91010, USA
| | - Jianying Zhang
- Department of Computational and Quantitative Medicine, City of Hope National Medical Center, Los Angeles, CA, 91010, USA
| | - Sean Wang
- Division of Transfusion Medicine, City of Hope National Medical Center, Los Angeles, CA, 91010, USA
| | - Srividya Swaminathan
- Department of Systems Biology, City of Hope Beckman Research Institute, Los Angeles, CA, 91010, USA
| | - Guido Marcucci
- Gehr Family Center for Leukemia Research, Hematologic Malignancies Research Institute, Department of Hematological Malignancies Translational Science, City of Hope National Medical Center, Los Angeles, CA, 91010, USA
| | - Yong Peng
- Center for Molecular Oncology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, China.
| | - Michael A Caligiuri
- Department of Hematology & Hematopoietic Cell Transplantation, City of Hope National Medical Center, Los Angeles, CA, 91010, USA.
- Department of Systems Biology, City of Hope Beckman Research Institute, Los Angeles, CA, 91010, USA.
| | - Jianhua Yu
- Division of Hematology & Oncology, Department of Medicine, School of Medicine, University of California, Irvine, CA, 92697, USA.
- The Clemons Family Center for Transformative Cancer Research, University of California, Irvine, CA, 92697, USA.
| |
Collapse
|
12
|
Hu Q, Xuan J, Wang L, Shen K, Gao Z, Zhou Y, Wei C, Gu J. Application of adoptive cell therapy in malignant melanoma. J Transl Med 2025; 23:102. [PMID: 39844295 PMCID: PMC11752767 DOI: 10.1186/s12967-025-06093-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Accepted: 01/07/2025] [Indexed: 01/24/2025] Open
Abstract
Cutaneous melanoma is one of the most aggressive skin cancers originating from skin pigment cells. Patients with advanced melanoma suffer a poor prognosis and generally cannot benefit well from surgical resection and chemo/target therapy due to metastasis and drug resistance. Thus, adoptive cell therapy (ACT), employing immune cells with specific tumor-recognizing receptors, has emerged as a promising therapeutic approach to display on-tumor toxicity. This review discusses the application, efficacy, limitations, as well as future prospects of four commonly utilized approaches -including tumor-infiltrating lymphocytes, chimeric antigen receptor (CAR) T cell, engineered T-cell receptor T cells, and chimeric antigen receptor NK cells- in the context of malignant melanoma.
Collapse
Affiliation(s)
- Qianrong Hu
- Department of Plastic and Reconstructive Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, People's Republic of China
| | - Jiangying Xuan
- Department of Plastic and Reconstructive Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, People's Republic of China
| | - Lu Wang
- Department of Plastic and Reconstructive Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, People's Republic of China
| | - Kangjie Shen
- Department of Plastic and Reconstructive Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, People's Republic of China
| | - Zixu Gao
- Department of Plastic and Reconstructive Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, People's Republic of China
| | - Yuhong Zhou
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, 200032, People's Republic of China
| | - Chuanyuan Wei
- Department of Plastic and Reconstructive Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, People's Republic of China.
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, 200032, People's Republic of China.
| | - Jianying Gu
- Department of Plastic and Reconstructive Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, People's Republic of China.
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, 200032, People's Republic of China.
| |
Collapse
|
13
|
Ahmed A, Fujimura NA, Tahir S, Akram M, Abbas Z, Riaz M, Raza A, Abbas R, Ahmed N. Soluble and insoluble expression of recombinant human interleukin-2 protein using pET expression vector in Escherichia coli. Prep Biochem Biotechnol 2025; 55:45-57. [PMID: 38824503 DOI: 10.1080/10826068.2024.2361146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/03/2024]
Abstract
Interleukin-2 has emerged as a potent protein-based drug to treat various cancers, AIDS, and autoimmune diseases. Despite its immense requirement, the production procedures are inefficient to meet the demand. Therefore, efficient production procedures must be adopted to improve protein yield and decrease procedural loss. This study analyzed cytoplasmic and periplasmic IL-2 expression for increased protein yield and significant biological activity. The study is focused on cloning IL-2 into a pET-SUMO and pET-28a vector that expresses IL-2 in soluble form and inclusion bodies, respectively. Both constructs were expressed into different E. coli expression strains, but the periplasmic and cytoplasmic expression of IL-2 was highest in overnight culture in Rosetta 2 (DE3). Therefore, E. coli Rosetta 2 (DE3) was selected for large-scale production and purification. Purified IL-2 was characterized by SDS-PAGE and western blotting, while its biological activity was determined using MTT bioassay. The results depict that the periplasmic and cytoplasmic IL-2 achieved adequate purification, yielding 0.86 and 0.51 mg/mL, respectively, with significant cytotoxic activity of periplasmic and cytoplasmic IL-2. Periplasmic IL-2 has shown better yield and significant biological activity in vitro which describes its attainment of native protein structure and function.
Collapse
Affiliation(s)
- Atif Ahmed
- Centre of Excellence in Molecular Biology, University of the Punjab, Lahore, Pakistan
| | - Nao Akusa Fujimura
- Centre of Excellence in Molecular Biology, University of the Punjab, Lahore, Pakistan
| | - Saad Tahir
- Centre of Excellence in Molecular Biology, University of the Punjab, Lahore, Pakistan
| | - Muhammad Akram
- Centre of Excellence in Molecular Biology, University of the Punjab, Lahore, Pakistan
| | - Zaheer Abbas
- Centre of Excellence in Molecular Biology, University of the Punjab, Lahore, Pakistan
| | - Maira Riaz
- Centre of Excellence in Molecular Biology, University of the Punjab, Lahore, Pakistan
| | - Ali Raza
- Centre of Excellence in Molecular Biology, University of the Punjab, Lahore, Pakistan
| | - Rabia Abbas
- Centre of Excellence in Molecular Biology, University of the Punjab, Lahore, Pakistan
| | - Nadeem Ahmed
- Centre of Excellence in Molecular Biology, University of the Punjab, Lahore, Pakistan
| |
Collapse
|
14
|
Li W, Jin D, Takai S, Inoue N, Yamanishi K, Tanaka Y, Okamura H. IL-18 primes T cells with an antigen-inexperienced memory phenotype for proliferation and differentiation into effector cells through Notch signaling. J Leukoc Biol 2024; 117:qiae172. [PMID: 39213165 DOI: 10.1093/jleuko/qiae172] [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: 07/05/2024] [Indexed: 09/04/2024] Open
Abstract
Recent studies have revealed that a subset of CD8+ T cells exhibit innate features and can be activated by cytokines. However, the precise mechanisms underlying the proliferation and differentiation of these cells remain unclear. Here, we demonstrated that CD44highCD8+ T cells in the mouse spleen express functional interleukin-18 (IL-18) receptors, whereas CD44lowCD8+ T cells do not. In response to IL-18 stimulation, these cells activated various metabolic pathways, upregulated the expression of surface molecules, such as c-Kit (CD117), CD25, and PD-1, and induced progression through the G1/S phase in the cell cycle. IL-18-primed cells, expressing a high-affinity receptor for IL-2, exhibited robust proliferation in response to IL-2 and underwent differentiation into effector cells. The splenic CD44highCD8+ T cells exhibited high expression levels of CD122, CD62L, CCR7, and CXCR3, along with CD5, indicating their potential for migration to the lymph nodes, where they could undergo expansion and terminal differentiation into effector cells. Additionally, in a tumor model, administration of IL-18 increased the accumulation of CD8+ T cells in both the lymph nodes and tumors. It is noteworthy that stimulation of CD44highCD8+ T cells with IL-18 upregulated the Notch-1 receptor and c-Myc. Moreover, inclusion of γ-secretase inhibitors attenuated the effect of IL-18 on both proliferation and interferon-γ production in the cells. These results demonstrate that IL-18 primes CD44highCD122highCXCR3highCD62LhighCD8+ T cells for expansion and differentiation into effector cells in a Notch signaling-dependent manner.
Collapse
Affiliation(s)
- Wen Li
- Department of Innovation Medicine, Osaka Medical and Pharmaceutical University, 2-7 Daigaku-cho, Takatsuki, Osaka 569-8686, Japan
- International Cooperation for Medical Innovation Co., Ltd., 1-5-2 Minami-machi, Minatojima, Chuo-ku, Kobe, Hyogo 650-0047, Japan
| | - Denan Jin
- Department of Innovation Medicine, Osaka Medical and Pharmaceutical University, 2-7 Daigaku-cho, Takatsuki, Osaka 569-8686, Japan
| | - Shinji Takai
- Department of Innovation Medicine, Osaka Medical and Pharmaceutical University, 2-7 Daigaku-cho, Takatsuki, Osaka 569-8686, Japan
| | - Natsuko Inoue
- Department of Innovation Medicine, Osaka Medical and Pharmaceutical University, 2-7 Daigaku-cho, Takatsuki, Osaka 569-8686, Japan
| | - Kyosuke Yamanishi
- Department of Neuropsychiatry, Hyogo College of Medicine, 1-1 Mukogawa-cho, Nishinomiya, Hyogo 663-8501, Japan
| | - Yoshimasa Tanaka
- Center for Medical Innovation, Nagasaki University, 1-7-1 Sakamoto, Nagasaki 852-8588, Japan
| | - Haruki Okamura
- Department of Innovation Medicine, Osaka Medical and Pharmaceutical University, 2-7 Daigaku-cho, Takatsuki, Osaka 569-8686, Japan
- International Cooperation for Medical Innovation Co., Ltd., 1-5-2 Minami-machi, Minatojima, Chuo-ku, Kobe, Hyogo 650-0047, Japan
| |
Collapse
|
15
|
Zhou X, Ning J, Cai R, Liu J, Yang H, Bai Y. Single-Cell Sequencing and Machine Learning Integration to Identify Candidate Biomarkers in Psoriasis: INSIG1. J Inflamm Res 2024; 17:11485-11503. [PMID: 39735895 PMCID: PMC11681806 DOI: 10.2147/jir.s492875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Accepted: 12/15/2024] [Indexed: 12/31/2024] Open
Abstract
Background Psoriasis represents a persistent, immune-driven inflammatory condition affecting the skin, characterized by a lack of well-established biologic treatments without adverse events. Consequently, the identification of novel targets and therapeutic agents remains a pressing priority in the field of psoriasis research. Methods We collected single-cell RNA sequencing (scRNA-seq) datasets and inferred T cell differentiation trajectories through pseudotime analysis. Bulk transcriptome and scRNA-seq data were integrated to identify differentially expressed genes (DEGs). Machine learning was employed to screen candidate genes. Correlation analysis was used to predict the interactions between cells expressing insulin-induced gene 1 (INSIG1) and other immune cells. Finally, drug docking was performed on INSIG1, and the expression levels of INSIG1 in psoriasis were verified through clinical and in vivo experiments, and further in vivo experiments established the efficacy of tetrandrine in the treatment of psoriasis. Results T cells were initially categorized into seven states, with differentially expressed genes in T cells (TDEGs) identified and their functions and signaling pathways. INSIG1 emerged as a characteristic gene for psoriasis and was found to be downregulated in psoriasis and potentially negatively associated with T cells, influencing psoriasis fatty acid metabolism, as inferred from enrichment and immunoinfiltration analyses. In the cellular communication network, cells expressing INSIG1 exhibited close interactions with other immune cells through multiple signaling channels. Furthermore, drug sensitivity showed that tetrandrine stably binds to INSIG1, could be a potential therapeutic agent for psoriasis. Conclusion INSIG1 emerges as a specific candidate gene potentially regulating the fatty acid metabolism of patients with psoriasis. In addition, tetrandrine shows promise as a potential treatment for the condition.
Collapse
Affiliation(s)
- Xiangnan Zhou
- Department of Dermatology, China-Japan Friendship Hospital, National Center for Integrative Medicine, Beijing, 100029, People’s Republic of China
- Beijing University of Chinese Medicine, China-Japan Friendship Clinical School of Medicine, Beijing, 100029, People’s Republic of China
| | - Jingyuan Ning
- State Key Laboratory of Medical Molecular Biology & Department of Medical Genetics, Institute of Basic Medical Sciences & School of Basic Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100005, People’s Republic of China
| | - Rui Cai
- Beijing University of Chinese Medicine, China-Japan Friendship Clinical School of Medicine, Beijing, 100029, People’s Republic of China
| | - Jiayi Liu
- Beijing University of Chinese Medicine, China-Japan Friendship Clinical School of Medicine, Beijing, 100029, People’s Republic of China
| | - Haoyu Yang
- Department of Dermatology, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, 100010, People’s Republic of China
| | - Yanping Bai
- Department of Dermatology, China-Japan Friendship Hospital, National Center for Integrative Medicine, Beijing, 100029, People’s Republic of China
| |
Collapse
|
16
|
Tsogtsaikhan S, Inoue SI, Bayarsaikhan G, Macalinao ML, Kimura D, Miyakoda M, Yamamoto M, Hara H, Yoshida H, Yui K. Regulation of memory CD4+ T-cell generation by intrinsic and extrinsic IL-27 signaling during malaria infection. Int Immunol 2024; 36:629-640. [PMID: 38895753 DOI: 10.1093/intimm/dxae039] [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: 05/01/2024] [Accepted: 06/18/2024] [Indexed: 06/21/2024] Open
Abstract
The generation and maintenance of memory T cells are regulated by various factors, including cytokines. Previous studies have shown that IL-27 is produced during the early acute phase of Plasmodium chabaudi chabaudi AS (Pcc) infection and inhibits the development of Th1-type memory CD4+ T cells. However, whether IL-27 acts directly on its receptor on Plasmodium-specific CD4+ T cells or indirectly via its receptor on other immune cells remains unclear. We aimed to determine the role of IL-27 receptor signaling in different immune cell types in regulating the generation and phenotype of memory CD4+ T cells during Plasmodium infection. We utilized Plasmodium-specific T-cell antigen receptor (TCR) transgenic mice, PbT-II, and Il27rα-/- mice to assess the direct and indirect effects of IL-27 signaling on memory CD4+ T-cell generation. Mice were transferred with PbT-II or Il27rα-/- PbT-II cells and infected with Pcc. Conditional knockout mice lacking the IL-27 receptor in T cells or dendritic cells were employed to discern the specific immune cell types involved in IL-27 receptor signaling. High levels of memory in PbT-II cells with Th1-shift occurred only when both PbT-II and host cells lacked the IL-27 receptor, suggesting the predominant inhibitory role of IL-27 signaling in both cell types. Furthermore, IL-27 receptor signaling in T cells limited the number of memory CD4+ T cells, while signaling in both T and dendritic cells contributed to the Th1 dominance of memory CD4+ T cells. These findings underscore the complex cytokine signaling network regulating memory CD4+ T cells during Plasmodium infection.
Collapse
Affiliation(s)
- Sanjaadorj Tsogtsaikhan
- Division of Immunology, Department of Molecular Microbiology and Immunology, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki 852-8523, Japan
- Program for Nurturing Global Leaders in Tropical and Emerging Communicable Diseases, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki 852-8523, Japan
| | - Shin-Ichi Inoue
- Division of Immunology, Department of Molecular Microbiology and Immunology, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki 852-8523, Japan
- Program for Nurturing Global Leaders in Tropical and Emerging Communicable Diseases, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki 852-8523, Japan
| | - Ganchimeg Bayarsaikhan
- Division of Immunology, Department of Molecular Microbiology and Immunology, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki 852-8523, Japan
| | - Maria Lourdes Macalinao
- School of Tropical Medicine and Global Health, Nagasaki University, Nagasaki 852-8523, Japan
- Department of Infection Biology, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, Keppel Street, London WC1E 7HT, UK
| | - Daisuke Kimura
- Division of Immunology, Department of Molecular Microbiology and Immunology, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki 852-8523, Japan
| | - Mana Miyakoda
- Division of Immunology, Department of Molecular Microbiology and Immunology, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki 852-8523, Japan
| | - Masahiro Yamamoto
- Department of Immunoparasitology, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka 565-0871, Japan
| | - Hiromitsu Hara
- Department of Immunology, Graduate School of Medical and Dental Sciences, Kagoshima University, 8-35-1, Sakuragaoka, Kagoshima 890-8544, Japan
| | - Hiroki Yoshida
- Division of Molecular and Cellular Immunoscience, Department of Biomolecular Sciences, Faculty of Medicine, Saga University, Saga 849-8501, Japan
| | - Katsuyuki Yui
- Division of Immunology, Department of Molecular Microbiology and Immunology, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki 852-8523, Japan
- Program for Nurturing Global Leaders in Tropical and Emerging Communicable Diseases, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki 852-8523, Japan
- School of Tropical Medicine and Global Health, Nagasaki University, Nagasaki 852-8523, Japan
- Shionogi Global Infectious Diseases Division, Institute of Tropical Medicine, Nagasaki University, Nagasaki 852-8523, Japan
| |
Collapse
|
17
|
Guan T, Bian Z, Gao H, He Y, Yuan J, Wan H, Tang S, Li Y, Qiu J, Luo P, Ye X, Liang S, Chen S, Cai N, Guan Y, Liu J, Zhao Z, Jia H, Yang W, Li W. Altered CD8 + T cell subpopulation in the bone marrow microenvironment of cynomolgus monkeys with spontaneous ankylosing spondylitis. Ann Rheum Dis 2024; 83:1600-1602. [PMID: 39019571 DOI: 10.1136/ard-2024-226018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Accepted: 07/08/2024] [Indexed: 07/19/2024]
Affiliation(s)
- Tangming Guan
- Guangdong Laboratory Animals Monitoring Institute, Guangzhou, Guangdong, China
| | - Zhenhua Bian
- South China University of Technology, Guangzhou, Guangdong, China
| | - Hongbin Gao
- Guangdong Laboratory Animals Monitoring Institute, Guangzhou, Guangdong, China
| | - Yi He
- Department of Rheumatology and Immunology, The Third Affiliated Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Junru Yuan
- Guangdong Laboratory Animals Monitoring Institute, Guangzhou, Guangdong, China
| | - Hongping Wan
- Guangdong Blooming-spring Biological Technology Development Co., Ltd, Guangzhou, Guangdong, China
| | - Shuangjie Tang
- Guangdong Blooming-spring Biological Technology Development Co., Ltd, Guangzhou, Guangdong, China
| | - Yongfeng Li
- Guangdong Blooming-spring Biological Technology Development Co., Ltd, Guangzhou, Guangdong, China
| | - Jianming Qiu
- Guangdong Blooming-spring Biological Technology Development Co., Ltd, Guangzhou, Guangdong, China
| | - Panyue Luo
- South China University of Technology, Guangzhou, Guangdong, China
| | - Xiaolan Ye
- Guangdong Laboratory Animals Monitoring Institute, Guangzhou, Guangdong, China
| | - Shi Liang
- Guangdong Laboratory Animals Monitoring Institute, Guangzhou, Guangdong, China
| | - Siyu Chen
- Guangdong Laboratory Animals Monitoring Institute, Guangzhou, Guangdong, China
| | - Nvlue Cai
- Guangdong Laboratory Animals Monitoring Institute, Guangzhou, Guangdong, China
| | - Yezhi Guan
- Guangdong Laboratory Animals Monitoring Institute, Guangzhou, Guangdong, China
| | - Jianxin Liu
- Guangdong Laboratory Animals Monitoring Institute, Guangzhou, Guangdong, China
| | - Zhibin Zhao
- Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Huanhuan Jia
- Guangdong Laboratory Animals Monitoring Institute, Guangzhou, Guangdong, China
| | - Wei Yang
- The Center for Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou, Guangdong, China
| | - Wende Li
- Guangdong Laboratory Animals Monitoring Institute, Guangzhou, Guangdong, China
| |
Collapse
|
18
|
Miao N, Cao D, Jin J, Ma G, Yu H, Qu J, Li G, Gao C, Dong D, Xia F, Li W. Tumor cell-intrinsic Piezo2 drives radioresistance by impairing CD8+ T cell stemness maintenance. J Exp Med 2024; 221:e20231486. [PMID: 39167075 PMCID: PMC11338319 DOI: 10.1084/jem.20231486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 05/20/2024] [Accepted: 07/24/2024] [Indexed: 08/23/2024] Open
Abstract
Changes in mechanosensitive ion channels following radiation have seldom been linked to therapeutic sensitivity or specific factors involved in antitumor immunity. Here, in this study, we found that the mechanical force sensor, Piezo2, was significantly upregulated in tumor cells after radiation, and Piezo2 knockout in tumor cells enhanced tumor growth suppression by radiotherapy. Specifically, loss of Piezo2 in tumor cells induced their IL-15 expression via unleashing JAK2/STAT1/IRF-1 axis after radiation. This increase in IL-15 activates IL-15Rα on tumor-infiltrating CD8+ T cells, thereby leading to their augmented effector and stem cell-like properties, along with reduced terminal exhausted feature. Importantly, Piezo2 expression was negatively correlated with CD8 infiltration, as well as with radiosensitivity of patients with rectum adenocarcinoma receiving radiotherapy treatment. Together, our findings reveal that tumor cell-intrinsic Piezo2 induces radioresistance by dampening the IRF-1/IL-15 axis, thus leading to impaired CD8+ T cell-dependent antitumor responses, providing insights into the further development of combination strategies to treat radioresistant cancers.
Collapse
Affiliation(s)
- Naijun Miao
- Precision Research Center for Refractory Diseases, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Immunology and Microbiology, Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Dongqing Cao
- Department of Immunology and Microbiology, Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jingsi Jin
- Department of Immunology and Microbiology, Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Guizhi Ma
- Precision Research Center for Refractory Diseases, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Immunology and Microbiology, Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Haihui Yu
- School of Basic Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Junwen Qu
- Department of Urology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Guiping Li
- Core Facility of Basic Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Caixia Gao
- Precision Research Center for Refractory Diseases, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Dong Dong
- Department of Thoracic Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Fan Xia
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Wenwen Li
- Precision Research Center for Refractory Diseases, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Immunology and Microbiology, Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
19
|
Raeber ME, Caspar DP, Zurbuchen Y, Guo N, Schmid J, Michler J, Martin AC, Steiner UC, Moor AE, Koning F, Boyman O. Interleukin-2 immunotherapy reveals human regulatory T cell subsets with distinct functional and tissue-homing characteristics. Immunity 2024; 57:2232-2250.e10. [PMID: 39137779 DOI: 10.1016/j.immuni.2024.07.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 05/24/2024] [Accepted: 07/18/2024] [Indexed: 08/15/2024]
Abstract
Due to its stimulatory potential for immunomodulatory CD4+ regulatory T (Treg) cells, low-dose interleukin-2 (IL-2) immunotherapy has gained considerable attention for the treatment of autoimmune diseases. In this investigator-initiated single-arm non-placebo-controlled phase-2 clinical trial of low-dose IL-2 immunotherapy in systemic lupus erythematosus (SLE) patients, we generated a comprehensive atlas of in vivo human immune responses to low-dose IL-2. We performed an in-depth study of circulating and cutaneous immune cells by imaging mass cytometry, high-parameter flow cytometry, transcriptomics, and targeted serum proteomics. Low-dose IL-2 stimulated various circulating immune cells, including Treg cells with a skin-homing phenotype that appeared in the skin of SLE patients in close interaction with endothelial cells. Analysis of surface proteins and transcriptomes revealed different IL-2-driven Treg cell activation programs, including gut-homing CD38+, skin-homing HLA-DR+, and highly proliferative inflammation-homing CD38+ HLA-DR+ Treg cells. Collectively, these data define the distinct human Treg cell subsets that are responsive to IL-2 immunotherapy.
Collapse
Affiliation(s)
- Miro E Raeber
- Department of Immunology, University Hospital Zurich, 8091 Zurich, Switzerland; Faculty of Medicine, University of Zurich, 8032 Zurich, Switzerland; Center of Human Immunology, University of Zurich, 8006 Zurich, Switzerland
| | - Dominic P Caspar
- Department of Immunology, University Hospital Zurich, 8091 Zurich, Switzerland
| | - Yves Zurbuchen
- Department of Immunology, University Hospital Zurich, 8091 Zurich, Switzerland
| | - Nannan Guo
- Department of Immunology, Leiden University Medical Center, 2300 RC Leiden, the Netherlands
| | - Jonas Schmid
- Department of Immunology, University Hospital Zurich, 8091 Zurich, Switzerland
| | - Jan Michler
- Department of Biosystems Science and Engineering, ETH Zurich, 4056 Basel, Switzerland
| | - Alina C Martin
- Department of Immunology, University Hospital Zurich, 8091 Zurich, Switzerland
| | - Urs C Steiner
- Department of Immunology, University Hospital Zurich, 8091 Zurich, Switzerland
| | - Andreas E Moor
- Department of Biosystems Science and Engineering, ETH Zurich, 4056 Basel, Switzerland
| | - Frits Koning
- Department of Immunology, Leiden University Medical Center, 2300 RC Leiden, the Netherlands
| | - Onur Boyman
- Department of Immunology, University Hospital Zurich, 8091 Zurich, Switzerland; Faculty of Medicine, University of Zurich, 8032 Zurich, Switzerland; Center of Human Immunology, University of Zurich, 8006 Zurich, Switzerland; Faculty of Science, University of Zurich, 8057 Zurich, Switzerland.
| |
Collapse
|
20
|
Xia X, Yang Z, Lu Q, Liu Z, Wang L, Du J, Li Y, Yang DH, Wu S. Reshaping the tumor immune microenvironment to improve CAR-T cell-based cancer immunotherapy. Mol Cancer 2024; 23:175. [PMID: 39187850 PMCID: PMC11346058 DOI: 10.1186/s12943-024-02079-8] [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: 06/14/2024] [Accepted: 08/02/2024] [Indexed: 08/28/2024] Open
Abstract
In many hematologic malignancies, the adoptive transfer of chimeric antigen receptor (CAR) T cells has demonstrated notable success; nevertheless, further improvements are necessary to optimize treatment efficacy. Current CAR-T therapies are particularly discouraging for solid tumor treatment. The immunosuppressive microenvironment of tumors affects CAR-T cells, limiting the treatment's effectiveness and safety. Therefore, enhancing CAR-T cell infiltration capacity and resolving the immunosuppressive responses within the tumor microenvironment could boost the anti-tumor effect. Specific strategies include structurally altering CAR-T cells combined with targeted therapy, radiotherapy, or chemotherapy. Overall, monitoring the tumor microenvironment and the status of CAR-T cells is beneficial in further investigating the viability of such strategies and advancing CAR-T cell therapy.
Collapse
Affiliation(s)
- Xueting Xia
- The Second Clinical Medical School, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China
| | - Zongxin Yang
- The Second Clinical Medical School, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China
| | - Qisi Lu
- The First Affiliated Hospital of Soochow University, Suzhou, 215006, China
- Foresea Life Insurance Guangzhou General Hospital, Guangzhou, 511300, China
| | - Zhenyun Liu
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China
| | - Lei Wang
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China
| | - Jinwen Du
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China
| | - Yuhua Li
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China.
| | - Dong-Hua Yang
- New York College of Traditional Chinese Medicine, Mineola, NY, 11501, USA.
| | - Shaojie Wu
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China.
| |
Collapse
|
21
|
He J, Chen Y, Ding H, Zhou JA, Xing Z, Yang X, Fan Q, Zuo Y, Wang T, Cheng J. Autocrine VEGF-B signaling maintains lipid synthesis and mitochondrial fitness to support T cell immune responses. J Clin Invest 2024; 134:e176586. [PMID: 39145452 PMCID: PMC11324299 DOI: 10.1172/jci176586] [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: 10/11/2023] [Accepted: 06/20/2024] [Indexed: 08/16/2024] Open
Abstract
T cells rewire their metabolic activities to meet the demand of immune responses, but how to coordinate the immune response by metabolic regulators in activated T cells is unknown. Here, we identified autocrine VEGF-B as a metabolic regulator to control lipid synthesis and maintain the integrity of the mitochondrial inner membrane for the survival of activated T cells. Disruption of autocrine VEGF-B signaling in T cells reduced cardiolipin mass, resulting in mitochondrial damage, with increased apoptosis and reduced memory development. The addition of cardiolipin or modulating VEGF-B signaling improved T cell mitochondrial fitness and survival. Autocrine VEGF-B signaling through GA-binding protein α (GABPα) induced sentrin/SUMO-specific protease 2 (SENP2) expression, which further de-SUMOylated PPARγ and enhanced phospholipid synthesis, leading to a cardiolipin increase in activated T cells. These data suggest that autocrine VEGF-B mediates a signal to coordinate lipid synthesis and mitochondrial fitness with T cell activation for survival and immune response. Moreover, autocrine VEGF-B signaling in T cells provides a therapeutic target against infection and tumors as well as an avenue for the treatment of autoimmune diseases.
Collapse
Affiliation(s)
- Jianli He
- Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Department of Biochemistry and Molecular Cell Biology
| | - Yalan Chen
- Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Department of Biochemistry and Molecular Cell Biology
| | - Huihua Ding
- Department of Rheumatology, Renji Hospital
- Shanghai Institute of Rheumatology, Renji Hospital, and
| | - Jin-An Zhou
- State Key Laboratory of Systems Medicine for Cancer, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhengcao Xing
- Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Department of Biochemistry and Molecular Cell Biology
| | - Xinyu Yang
- Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Department of Biochemistry and Molecular Cell Biology
| | - Qiuju Fan
- Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Department of Biochemistry and Molecular Cell Biology
| | - Yong Zuo
- Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Department of Biochemistry and Molecular Cell Biology
| | - Tianshi Wang
- Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Department of Biochemistry and Molecular Cell Biology
| | - Jinke Cheng
- Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Department of Biochemistry and Molecular Cell Biology
- Hainan Medical University, Hainan Academy of Medical Sciences, Haikou, Hainan, China
| |
Collapse
|
22
|
Baldini C, Fulvio G, La Rocca G, Ferro F. Update on the pathophysiology and treatment of primary Sjögren syndrome. Nat Rev Rheumatol 2024; 20:473-491. [PMID: 38982205 DOI: 10.1038/s41584-024-01135-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/04/2024] [Indexed: 07/11/2024]
Abstract
Sjögren syndrome or Sjögren disease is a chronic form of autoimmune epithelitis characterized by lymphocytic infiltration of the exocrine glands, particularly the salivary and lacrimal glands, leading to progressive glandular dysfunction and subsequent xerostomia and xerophthalmia. Other common manifestations include pain and fatigue, various systemic manifestations and non-Hodgkin's lymphoma. Sjögren syndrome is therefore a complex and disabling disease associated with a reduced quality of life and with considerable long-term damage. Most of the available treatments are merely symptomatic with limited efficacy in both preventing glandular damage and suppressing systemic disease activity. In the past 10 years, great progress has been made in understanding the pathophysiology of Sjögren syndrome, opening new avenues towards a more targeted and individualized therapeutic approach to the disease. Indeed, several randomized controlled trials have just been completed or are poised to commence evaluating the effectiveness of novel drugs targeting both innate and adaptive immune pathways, including pro-inflammatory cytokines, the type I interferon system, B cell activation, B cell and T cell co-stimulation pathway, and ectopic germinal centre formation. Novel clinical trials are also ongoing exploring various targeted approaches (that is, IgG recycling inhibition, nuclease therapy and CAR-T cell therapy) for Sjögren syndrome.
Collapse
Affiliation(s)
- Chiara Baldini
- Rheumatology Unit, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy.
| | - Giovanni Fulvio
- Rheumatology Unit, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Gaetano La Rocca
- Rheumatology Unit, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Francesco Ferro
- Rheumatology Unit, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| |
Collapse
|
23
|
Goleij P, Rahimi M, Pourshahroudi M, Tabari MAK, Muhammad S, Suteja RC, Daglia M, Majma Sanaye P, Hadipour M, Khan H, Sadeghi P. The role of IL-2 cytokine family in asthma. Cytokine 2024; 180:156638. [PMID: 38761716 DOI: 10.1016/j.cyto.2024.156638] [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: 03/09/2024] [Revised: 04/25/2024] [Accepted: 05/02/2024] [Indexed: 05/20/2024]
Abstract
BACKGROUND The interleukin-2 (IL-2) family of cytokines, including IL-2, IL-4, IL-7, IL-9, IL-15, and IL-21, are pivotal regulators of the immune response, impacting both innate and adaptive immunity. Understanding their molecular characteristics, receptor interactions, and signalling pathways is essential for elucidating their roles in health and disease. OBJECTIVES This review provides a comprehensive overview of the IL-2 family of cytokines, highlighting their molecular biology, receptor interactions, and signalling mechanisms. Furthermore, it explores the involvement of IL-2 family cytokines in the pathogenesis of chronic respiratory diseases, with a specific focus on chronic obstructive pulmonary disease (COPD) and asthma. METHODS A thorough literature review was conducted to gather insights into the molecular biology, receptor interactions, and signalling pathways of IL-2 family cytokines. Additionally, studies investigating the roles of these cytokines in chronic respiratory diseases, particularly COPD and asthma, were analysed to discern their implications in wider pathophysiology of disease. RESULTS IL-2 family cytokines exert pleiotropic effects on immune cells, modulating cellular proliferation, differentiation, and survival. Dysregulation of IL-2 family cytokines has been implicated in the pathogenesis of chronic respiratory illnesses, including COPD and asthma. Elevated levels of IL-2 and IL-9 have been associated with disease severity in COPD, while IL-4 and IL-9 play crucial roles in asthma pathogenesis by promoting airway inflammation and remodelling. CONCLUSION Understanding the intricate roles of IL-2 family cytokines in chronic respiratory diseases provides valuable insights into potential therapeutic targets for these conditions. Targeting specific cytokines or their receptors may offer novel treatment modalities to attenuate disease progression and improve clinical outcomes in patients with COPD and asthma.
Collapse
Affiliation(s)
- Pouya Goleij
- USERN Office, Kermanshah University of Medical Sciences, Kermanshah, Iran; Immunology Board for Transplantation and Cell-Based Therapeutics (ImmunoTACT), Universal Scientific Education and Research Network (USERN), Tehran, Iran.
| | - Mohammad Rahimi
- Student Research Committee, School of Medicine, Mazandaran University of Medical Sciences, Mazandaran, Iran.
| | - Motahareh Pourshahroudi
- Department of Public Health, Faculty of Health, Education and Life Sciences, Birmingham City University, Birmingham, United Kingdom.
| | - Mohammad Amin Khazeei Tabari
- Student Research Committee, School of Medicine, Mazandaran University of Medical Sciences, Mazandaran, Iran; Network of Interdisciplinarity in Neonates and Infants (NINI), Universal Scientific Education and Research Network (USERN), Tehran, Iran.
| | - Syed Muhammad
- Farooqia College of Pharmacy, Mysuru, Karnataka, India.
| | | | - Maria Daglia
- Department of Pharmacy, University of Naples "Federico II", Via D. Montesano 49, 80131 Naples, Italy; International Research Center for Food Nutrition and Safety, Jiangsu University, Zhenjiang 212013, China.
| | | | - Mahboube Hadipour
- Department of Biochemistry, School of Medicine, Hormozgan University of Medical Sciences, Bandar Abbas, Iran.
| | - Haroon Khan
- Department of Pharmacy, Faculty of Chemical and Life Sciences, Abdul Wali Khan University Mardan, Mardan 23200, Pakistan.
| | - Parniyan Sadeghi
- Network of Interdisciplinarity in Neonates and Infants (NINI), Universal Scientific Education and Research Network (USERN), Tehran, Iran; School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
24
|
Hu J, Liu X. Generation of CAR-T SCM: CAR-T with super clutch. Int Immunopharmacol 2024; 136:112379. [PMID: 38833844 DOI: 10.1016/j.intimp.2024.112379] [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: 03/18/2024] [Revised: 05/26/2024] [Accepted: 05/28/2024] [Indexed: 06/06/2024]
Abstract
CAR-T therapy has demonstrated effectiveness in hematological malignancies and is now striding into solid tumor areas. One of the main roadblocks of CAR-T therapy is T cell exhaustion normally aroused by T cell terminal differentiation due to persistent contact with antigen in vivo or in vitro manufacturing process. TSCM positions as the first, and pivotal step of naïve T cell differentiation to downstream memory and effector stages. Researchers highly seek to restrain CAR-T cells at the TSCM stage during manufacture as TSCM percentage in CAR-T products is strongly associated with better treatment response. We reviewed the recent strategies regarding CAR-TSCM generation from aspects of starting source, manufacturing process, CAR assembly, transcription factor and metabolism regulation, etc.
Collapse
Affiliation(s)
- Jinhui Hu
- Department of Laboratory Medicine, Gongli Hospital, No. 219, Miaopu Road, Pudong, Shanghai, 200135, China.
| | - Xiang Liu
- TriArm Therapeutics Inc, Building 5, Niudun Road, Pudong New District, Shanghai, 201203, China.
| |
Collapse
|
25
|
Xiong H, Shen Z. Tissue-resident memory T cells in immunotherapy and immune-related adverse events by immune checkpoint inhibitor. Int J Cancer 2024; 155:193-202. [PMID: 38554117 DOI: 10.1002/ijc.34940] [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: 10/20/2023] [Revised: 03/05/2024] [Accepted: 03/07/2024] [Indexed: 04/01/2024]
Abstract
Tissue-resident memory T cells (TRM) are a specialized subset of T cells that reside in tissues and provide long-term protective immunity against pathogens that enter the body through that specific tissue. TRM cells have specific phenotype and reside preferentially in barrier tissues. Recent studies have revealed that TRM cells are the main target of immune checkpoint inhibitor immunotherapy since their role in cancer immunosurveillance. Furthermore, TRM cells also play a crucial part in pathogenesis of immune-related adverse events (irAEs). Here, we provide a concise review of biological characteristics of TRM cells, and the major advances and recent findings regarding their involvement in immune checkpoint inhibitor immunotherapy and the corresponding irAEs.
Collapse
Affiliation(s)
- Hao Xiong
- Department of Dermatology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, China
| | - Zhu Shen
- Department of Dermatology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, China
| |
Collapse
|
26
|
Gray-Gaillard SL, Solis SM, Chen HM, Monteiro C, Ciabattoni G, Samanovic MI, Cornelius AR, Williams T, Geesey E, Rodriguez M, Ortigoza MB, Ivanova EN, Koralov SB, Mulligan MJ, Herati RS. SARS-CoV-2 inflammation durably imprints memory CD4 T cells. Sci Immunol 2024; 9:eadj8526. [PMID: 38905326 PMCID: PMC11824880 DOI: 10.1126/sciimmunol.adj8526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 05/30/2024] [Indexed: 06/23/2024]
Abstract
Memory CD4 T cells are critical to human immunity, yet it is unclear whether viral inflammation during memory formation has long-term consequences. Here, we compared transcriptional and epigenetic landscapes of Spike (S)-specific memory CD4 T cells in 24 individuals whose first exposure to S was via SARS-CoV-2 infection or mRNA vaccination. Nearly 2 years after memory formation, S-specific CD4 T cells established by infection remained enriched for transcripts related to cytotoxicity and for interferon-stimulated genes, likely because of a chromatin accessibility landscape altered by inflammation. Moreover, S-specific CD4 T cells primed by infection had reduced proliferative capacity in vitro relative to vaccine-primed cells. Furthermore, the transcriptional state of S-specific memory CD4 T cells was minimally altered by booster immunization and/or breakthrough infection. Thus, infection-associated inflammation durably imprints CD4 T cell memory, which affects the function of these cells and may have consequences for long-term immunity.
Collapse
Affiliation(s)
| | - Sabrina M. Solis
- Department of Medicine, New York University Grossman School
of Medicine; New York, NY, USA
| | - Han M. Chen
- Department of Medicine, New York University Grossman School
of Medicine; New York, NY, USA
| | - Clarice Monteiro
- Department of Medicine, New York University Grossman School
of Medicine; New York, NY, USA
| | - Grace Ciabattoni
- Department of Microbiology, New York University School of
Medicine; New York, NY, USA
| | - Marie I. Samanovic
- Department of Medicine, New York University Grossman School
of Medicine; New York, NY, USA
| | - Amber R. Cornelius
- Department of Medicine, New York University Grossman School
of Medicine; New York, NY, USA
| | - Tijaana Williams
- Department of Medicine, New York University Grossman School
of Medicine; New York, NY, USA
| | - Emilie Geesey
- Department of Medicine, New York University Grossman School
of Medicine; New York, NY, USA
| | - Miguel Rodriguez
- Department of Medicine, New York University Grossman School
of Medicine; New York, NY, USA
| | - Mila Brum Ortigoza
- Department of Medicine, New York University Grossman School
of Medicine; New York, NY, USA
| | - Ellie N. Ivanova
- Department of Pathology, New York University School of
Medicine; New York, NY, USA
| | - Sergei B. Koralov
- Department of Pathology, New York University School of
Medicine; New York, NY, USA
| | - Mark J. Mulligan
- Department of Medicine, New York University Grossman School
of Medicine; New York, NY, USA
- Department of Microbiology, New York University School of
Medicine; New York, NY, USA
| | - Ramin Sedaghat Herati
- Department of Medicine, New York University Grossman School
of Medicine; New York, NY, USA
- Department of Microbiology, New York University School of
Medicine; New York, NY, USA
| |
Collapse
|
27
|
Zuo D, Zhu Y, Wang K, Qin Y, Su Y, Lan S, Li Y, Dong S, Liang Y, Feng M. A novel LAG3 neutralizing antibody improves cancer immunotherapy by dual inhibition of MHC-II and FGL1 ligand binding. Biomed Pharmacother 2024; 175:116782. [PMID: 38776682 DOI: 10.1016/j.biopha.2024.116782] [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: 04/05/2024] [Revised: 05/08/2024] [Accepted: 05/17/2024] [Indexed: 05/25/2024] Open
Abstract
LAG3 is an inhibitory immune checkpoint expressed on activated T and NK cells. Blocking the interaction of LAG3 with its ligands MHC-II and FGL1 renders T cells improved cytotoxicity to cancer cells. Current study generated a panel of LAG3 monoclonal antibodies (mAbs) through immunization of mice followed by phage display. Some of them bound to the D1-D2 domain of LAG3, which is known for the engagement of its ligands FGL1 and MHC-II. Three outperformers, M208, M226, and M234, showed stronger blocking activity than Relatlimab in the FGL1 binding. Furthermore, M234 showed dual inhibition of FGL1 (IC50 of 20.6 nM) and MHC-II binding (IC50 of 6.2 nM) to LAG3. In vitro functional tests showed that M234 significantly stimulated IFN-γ secretion from activated PBMC cells. In vivo studies in a mouse model of hepatocellular carcinoma xenografts demonstrated that combining M234 IgG with GPC3-targeted bispecific antibodies significantly improved efficacy. In addition, GPC3-targeted CAR-T cells secreting IL-21-M234 scFv fusion protein exhibited enhanced activity in inhibiting tumor growth and greatly increased the survival rate of mice. Taken together, M234 has potential in cancer immunotherapy and warrants further clinical trial.
Collapse
MESH Headings
- Animals
- Lymphocyte Activation Gene 3 Protein
- Humans
- Mice
- Antigens, CD/immunology
- Antigens, CD/metabolism
- Antibodies, Neutralizing/pharmacology
- Antibodies, Neutralizing/immunology
- Ligands
- Immunotherapy/methods
- Cell Line, Tumor
- Histocompatibility Antigens Class II/immunology
- Histocompatibility Antigens Class II/metabolism
- Xenograft Model Antitumor Assays
- Liver Neoplasms/immunology
- Liver Neoplasms/drug therapy
- Liver Neoplasms/pathology
- Liver Neoplasms/therapy
- Carcinoma, Hepatocellular/immunology
- Carcinoma, Hepatocellular/drug therapy
- Carcinoma, Hepatocellular/therapy
- Carcinoma, Hepatocellular/pathology
- Mice, Inbred BALB C
- Protein Binding
- Female
- Antibodies, Monoclonal/pharmacology
Collapse
Affiliation(s)
- Dianbao Zuo
- College of Life Science and Technology, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Yuankui Zhu
- College of Life Science and Technology, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Ke Wang
- College of Life Science and Technology, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Youjia Qin
- College of Life Science and Technology, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Yiyi Su
- College of Life Science and Technology, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Sina Lan
- College of Life Science and Technology, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Yunyi Li
- College of Life Science and Technology, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Shuang Dong
- Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430079, China; Hubei Provincial Clinical Research Center for Colorectal Cancer, China; Wuhan Clinical Research Center for Colorectal Cancer, China
| | - Yinming Liang
- Center of Disease Model and Immunology, Hunan Academy of Chinese Medicine, Changsha, Hunan 410013, China.
| | - Mingqian Feng
- Hubei Provincial Clinical Research Center for Colorectal Cancer, China; College of Biomedicine and Health, Huazhong Agricultural University, Wuhan, Hubei 430070, China.
| |
Collapse
|
28
|
Wang L, Wei Y, Jin Z, Liu F, Li X, Zhang X, Bai X, Jia Q, Zhu B, Chu Q. IFN-α/β/IFN-γ/IL-15 pathways identify GBP1-expressing tumors with an immune-responsive phenotype. Clin Exp Med 2024; 24:102. [PMID: 38758367 PMCID: PMC11101573 DOI: 10.1007/s10238-024-01328-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 03/09/2024] [Indexed: 05/18/2024]
Abstract
Immunotherapy is widely used in cancer treatment; however, only a subset of patients responds well to it. Significant efforts have been made to identify patients who will benefit from immunotherapy. Successful anti-tumor immunity depends on an intact cancer-immunity cycle, especially long-lasting CD8+ T-cell responses. Interferon (IFN)-α/β/IFN-γ/interleukin (IL)-15 pathways have been reported to be involved in the development of CD8+ T cells. And these pathways may predict responses to immunotherapy. Herein, we aimed to analyze multiple public databases to investigate whether IFN-α/β/IFN-γ/IL-15 pathways could be used to predict the response to immunotherapy. Results showed that IFN-α/β/IFN-γ/IL-15 pathways could efficiently predict immunotherapy response, and guanylate-binding protein 1 (GBP1) could represent the IFN-α/β/IFN-γ/IL-15 pathways. In public and private cohorts, we further demonstrated that GBP1 could efficiently predict the response to immunotherapy. Functionally, GBP1 was mainly expressed in macrophages and strongly correlated with chemokines involved in T-cell migration. Therefore, our study comprehensively investigated the potential role of GBP1 in immunotherapy, which could serve as a novel biomarker for immunotherapy and a target for drug development.
Collapse
Affiliation(s)
- Lei Wang
- Department of Oncology, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, People's Republic of China
| | - Yuxuan Wei
- Department of Oncology, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, People's Republic of China
| | - Zheng Jin
- Institute of Life Sciences, Chongqing Medical University, Chongqing, 400032, People's Republic of China
- Research Institute, GloriousMed Clinical Laboratory (Shanghai) Co., Ltd, Shanghai, 201318, People's Republic of China
| | - Fangfang Liu
- Department of Oncology, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, People's Republic of China
| | - Xuchang Li
- Department of Oncology, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, People's Republic of China
| | - Xiao Zhang
- Army 953 Hospital, Shigatse Branch of Xinqiao Hospital, Army Medical University, Shigatse, 857000, People's Republic of China
| | - Xiumei Bai
- Army 953 Hospital, Shigatse Branch of Xinqiao Hospital, Army Medical University, Shigatse, 857000, People's Republic of China
| | - Qingzhu Jia
- Department of Oncology, Xinqiao Hospital, Army Medical University, Chongqing, 400037, People's Republic of China
- Chongqing Key Laboratory of Immunotherapy, Chongqing, 400037, People's Republic of China
| | - Bo Zhu
- Department of Oncology, Xinqiao Hospital, Army Medical University, Chongqing, 400037, People's Republic of China
- Chongqing Key Laboratory of Immunotherapy, Chongqing, 400037, People's Republic of China
| | - Qian Chu
- Department of Oncology, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, People's Republic of China.
| |
Collapse
|
29
|
Guo Y, Mao W, Bai N, Jin L, Tang S, Lin X, Ni J, Liu X, Fu H, Shou Q. Integrated network pharmacological analysis revealed that Smilax glabra Roxb. alleviates IMQ-induced psoriatic skin inflammation through regulating T cell immune response. JOURNAL OF ETHNOPHARMACOLOGY 2024; 325:117836. [PMID: 38301985 DOI: 10.1016/j.jep.2024.117836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 01/15/2024] [Accepted: 01/25/2024] [Indexed: 02/03/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Psoriasis is an autoimmune disease characterized by dysfunctional T cells and dysregulated immune responses. Smilax glabra Roxb. (SGR) is a formulation used in Traditional Chinese Medicine for the treatment of inflammatory skin disorders, including psoriasis. This study explores the scientific basis for its use by examining the effects of SGR on T cell differentiation and insulin receptor signaling, relevant pathways implicated in the pathophysiology of psoriasis. AIM OF THE STUDY This study investigates the therapeutic potential of SGR (a Chinese medicine) in psoriasis and its impact on T cell differentiation. MATERIALS AND METHODS An integrated network pharmacology and bioinformatics approach was employed to elucidate the mechanisms of SGR in regulating T cell differentiation. A psoriasis mouse model was utilized to evaluate the effects of SGR on T cell subsets. Immunohistochemistry and gene expression analyses were conducted to investigate the modulation of insulin receptor signaling pathways by SGR. RESULTS SGR treatment effectively reset the expression of various T cell subsets in the psoriasis mouse model, suggesting its ability to regulate T cell differentiation and immune function. Furthermore, SGR treatment inhibited insulin receptor signaling and downstream pathways, including PI3K/AKT and ERK, in psoriatic skin lesions. This indicates that SGR may exert its therapeutic effects through modulation of the insulin receptor signaling pathway. CONCLUSIONS This study provides novel insights into the therapeutic potential of SGR in psoriasis. By modulating T cell differentiation and targeting the insulin receptor signaling pathway, SGR holds promise as a potential treatment option for psoriasis.
Collapse
Affiliation(s)
- Yingxue Guo
- Second Clinical Medical College, Jinhua Academy, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, China
| | - Weiye Mao
- Second Clinical Medical College, Jinhua Academy, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, China; Zhezhong Laboratory, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, China
| | - Ningning Bai
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 311402, China
| | - Lu Jin
- Second Clinical Medical College, Jinhua Academy, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, China
| | - Shuiyan Tang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 311402, China
| | - Xiaochen Lin
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 311402, China
| | - Jianyu Ni
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 311402, China
| | - Xia Liu
- Second Clinical Medical College, Jinhua Academy, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, China
| | - Huiying Fu
- Second Clinical Medical College, Jinhua Academy, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, China.
| | - Qiyang Shou
- Second Clinical Medical College, Jinhua Academy, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, China; Zhezhong Laboratory, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, China.
| |
Collapse
|
30
|
Gharib E, Rejali L, Piroozkhah M, Zonoobi E, Nasrabadi PN, Arabsorkhi Z, Baghdar K, Shams E, Sadeghi A, Kuppen PJK, Salehi Z, Nazemalhosseini-Mojarad E. IL-2RG as a possible immunotherapeutic target in CRC predicting poor prognosis and regulated by miR-7-5p and miR-26b-5p. J Transl Med 2024; 22:439. [PMID: 38720389 PMCID: PMC11080123 DOI: 10.1186/s12967-024-05251-2] [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] [Received: 12/19/2023] [Accepted: 04/29/2024] [Indexed: 05/12/2024] Open
Abstract
Despite advances in treatment strategies, colorectal cancer (CRC) continues to cause significant morbidity and mortality, with mounting evidence a close link between immune system dysfunctions issued. Interleukin-2 receptor gamma (IL-2RG) plays a pivotal role as a common subunit receptor in the IL-2 family cytokines and activates the JAK-STAT pathway. This study delves into the role of Interleukin-2 receptor gamma (IL-2RG) within the tumor microenvironment and investigates potential microRNAs (miRNAs) that directly inhibit IL-2RG, aiming to discern their impact on CRC clinical outcomes. Bioinformatics analysis revealed a significant upregulation of IL-2RG mRNA in TCGA-COAD samples and showed strong correlations with the infiltration of various lymphocytes. Single-cell analysis corroborated these findings, highlighting IL-2RG expression in critical immune cell subsets. To explore miRNA involvement in IL-2RG dysregulation, mRNA was isolated from the tumor tissues and lymphocytes of 258 CRC patients and 30 healthy controls, and IL-2RG was cloned into the pcDNA3.1/CT-GFP-TOPO vector. Human embryonic kidney cell lines (HEK-293T) were transfected with this construct. Our research involved a comprehensive analysis of miRPathDB, miRWalk, and Targetscan databases to identify the miRNAs associated with the 3' UTR of human IL-2RG. The human microRNA (miRNA) molecules, hsa-miR-7-5p and hsa-miR-26b-5p, have been identified as potent suppressors of IL-2RG expression in CRC patients. Specifically, the downregulation of hsa-miR-7-5p and hsa-miR-26b-5p has been shown to result in the upregulation of IL-2RG mRNA expression in these patients. Prognostic evaluation of IL-2RG, hsa-miR-7-5p, and hsa-miR-26b-5p, using TCGA-COAD data and patient samples, established that higher IL-2RG expression and lower expression of both miRNAs were associated with poorer outcomes. Additionally, this study identified several long non-coding RNAs (LncRNAs), such as ZFAS1, SOX21-AS1, SNHG11, SNHG16, SNHG1, DLX6-AS1, GAS5, SNHG6, and MALAT1, which may act as competing endogenous RNA molecules for IL2RG by sequestering shared hsa-miR-7-5p and hsa-miR-26b-5p. In summary, this investigation underscores the potential utility of IL-2RG, hsa-miR-7-5p, and hsa-miR-26b-5p as serum and tissue biomarkers for predicting CRC patient prognosis while also offering promise as targets for immunotherapy in CRC management.
Collapse
Affiliation(s)
- Ehsan Gharib
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Leili Rejali
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Moein Piroozkhah
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Elham Zonoobi
- Department of Surgery, Leiden University Medical Center, Leiden, Netherlands
| | - Parinaz Nasri Nasrabadi
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Zahra Arabsorkhi
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Kaveh Baghdar
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Elahe Shams
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Amir Sadeghi
- Gastroenterology and Liver Diseases Research Centre, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Yeman Street, Chamran Expressway, P.O. Box: 19857-17411, Tehran, Iran
| | - Peter J K Kuppen
- Department of Surgery, Leiden University Medical Center, Leiden, Netherlands
| | - Zahra Salehi
- Hematology, Oncology and Stem Cell Transplantation Research Center, Tehran University of Medical Sciences, Tehran, Iran.
- Research Institute for Oncology, Hematology and Cell Therapy, Tehran University of Medical Sciences, Tehran, Iran.
| | - Ehsan Nazemalhosseini-Mojarad
- Department of Surgery, Leiden University Medical Center, Leiden, Netherlands.
- Gastroenterology and Liver Diseases Research Centre, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Yeman Street, Chamran Expressway, P.O. Box: 19857-17411, Tehran, Iran.
| |
Collapse
|
31
|
Zhang J, Li K, Cao Y, Wang D, Cheng J, Gao H, Geng M, Yang J, Wei X. Inducible IL-2 production and IL-2 + cell expansion are landmark events for T-cell activation of teleost. FISH & SHELLFISH IMMUNOLOGY 2024; 148:109515. [PMID: 38499218 DOI: 10.1016/j.fsi.2024.109515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Revised: 03/14/2024] [Accepted: 03/15/2024] [Indexed: 03/20/2024]
Abstract
As a multipotent cytokine, interleukin (IL)-2 plays important roles in activation, differentiation and survival of the lymphocytes. Although biological characteristics and function of IL-2 have been clarified in several teleost species, evidence regarding IL-2 production at the cellular and protein levels is still scarce in fish due to the lack of reliable antibody. In this study, we developed a mouse anti-Nile tilapia IL-2 monoclonal antibody (mAb), which could specifically recognize IL-2 protein and identify IL-2-producing lymphocytes of tilapia. Using this mAb, we found that CD3+ T cells, but not CD3- lymphocytes, are the main cellular source of IL-2 in tilapia. Under resting condition, both CD3+CD4-1+ T cells and CD3+CD4-1- T cells of tilapia produce IL-2. Moreover, the IL-2 protein level and the frequency of IL-2+ T cells significantly increased once T cells were activated by phytohemagglutinin (PHA) or CD3 plus CD28 mAbs in vitro. In addition, Edwardsiella piscicida infection also induces the IL-2 production and the expansion of IL-2+ T cells in the spleen lymphocytes. These findings demonstrate that IL-2 takes part in the T-cell activation and anti-bacterial adaptive immune response of tilapia, and can serve as an important marker for T-cell activation of teleost fish. Our study has enriched the knowledge regarding T-cell response in fish species, and also provide novel perspective for understanding the evolution of adaptive immune system.
Collapse
Affiliation(s)
- Jiansong Zhang
- State Key Laboratory of Estuarine and Coastal Research, School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Kang Li
- State Key Laboratory of Estuarine and Coastal Research, School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Yi Cao
- State Key Laboratory of Estuarine and Coastal Research, School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Ding Wang
- State Key Laboratory of Estuarine and Coastal Research, School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Jie Cheng
- State Key Laboratory of Estuarine and Coastal Research, School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Haiyou Gao
- State Key Laboratory of Estuarine and Coastal Research, School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Ming Geng
- State Key Laboratory of Estuarine and Coastal Research, School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Jialong Yang
- State Key Laboratory of Estuarine and Coastal Research, School of Life Sciences, East China Normal University, Shanghai, 200241, China; Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China.
| | - Xiumei Wei
- State Key Laboratory of Estuarine and Coastal Research, School of Life Sciences, East China Normal University, Shanghai, 200241, China.
| |
Collapse
|
32
|
Feng J, Li Y, Wang C, Wang Y, Wan Y, Zheng M, Chen T, Xiao X. Peripheral blood transcriptomic analysis identifies potential inflammation and immune signatures for central retinal artery occlusion. Sci Rep 2024; 14:7398. [PMID: 38548806 PMCID: PMC10978867 DOI: 10.1038/s41598-024-57052-8] [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] [Received: 10/12/2023] [Accepted: 03/13/2024] [Indexed: 04/01/2024] Open
Abstract
Central retinal artery occlusion (CRAO) is an acute retinal ischaemic disease, but early diagnosis is challenging due to a lack of biomarkers. Blood samples were collected from CRAO patients and cataract patients. Gene expression profiles were distinct between arterial/venous CRAO blood (A-V group) and venous CRAO/control blood (V-C group) samples. Differentially expressed genes (DEGs) were subjected to GO and KEGG enrichment analyses. Hub genes were identified by Cytoscape and used to predict gene interactions via GeneMANIA. Immune cell infiltration was analysed by CIBERSORT. More than 1400 DEGs were identified in the A-V group and 112 DEGs in the V-C group compared to controls. The DEGs in both groups were enriched in the ribosome pathway, and those in the V-C group were also enriched in antigen processing/MHC pathways. Network analysis identified ribosomal proteins (RPS2 and RPS5) as the core genes of the A-V group and MHC genes (HLA-F) as the core genes of the V-C group. Coexpression networks showed ribosomal involvement in both groups, with additional immune responses in the V-C group. Immune cell analysis indicated increased numbers of neutrophils and T cells. Ribosomal and MHC-related genes were identified as potential CRAO biomarkers, providing research directions for prevention, diagnosis, treatment and prognosis.
Collapse
Affiliation(s)
- Jiaqing Feng
- Department of Ophthalmology, Renmin Hospital of Wuhan University, No. 238 Jie Fang Road, Wuhan, 430060, Hubei, China
| | - Ying Li
- Department of Ophthalmology, Renmin Hospital of Wuhan University, No. 238 Jie Fang Road, Wuhan, 430060, Hubei, China
| | - Chuansen Wang
- Department of Ophthalmology, Renmin Hospital of Wuhan University, No. 238 Jie Fang Road, Wuhan, 430060, Hubei, China
| | - Yuedan Wang
- Department of Ophthalmology, Renmin Hospital of Wuhan University, No. 238 Jie Fang Road, Wuhan, 430060, Hubei, China
| | - Yuwei Wan
- Department of Ophthalmology, Renmin Hospital of Wuhan University, No. 238 Jie Fang Road, Wuhan, 430060, Hubei, China
| | - Mengxue Zheng
- Department of Ophthalmology, Renmin Hospital of Wuhan University, No. 238 Jie Fang Road, Wuhan, 430060, Hubei, China
| | - Ting Chen
- Department of Ophthalmology, Renmin Hospital of Wuhan University, No. 238 Jie Fang Road, Wuhan, 430060, Hubei, China.
| | - Xuan Xiao
- Department of Ophthalmology, Renmin Hospital of Wuhan University, No. 238 Jie Fang Road, Wuhan, 430060, Hubei, China.
- Department of Clinical Laboratory, Institute of Translational Medicine, Renmin Hospital of Wuhan University, Wuhan, China.
| |
Collapse
|
33
|
Guo Q, Jin Y, Chen X, Ye X, Shen X, Lin M, Zeng C, Zhou T, Zhang J. NF-κB in biology and targeted therapy: new insights and translational implications. Signal Transduct Target Ther 2024; 9:53. [PMID: 38433280 PMCID: PMC10910037 DOI: 10.1038/s41392-024-01757-9] [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: 10/19/2023] [Revised: 01/16/2024] [Accepted: 01/19/2024] [Indexed: 03/05/2024] Open
Abstract
NF-κB signaling has been discovered for nearly 40 years. Initially, NF-κB signaling was identified as a pivotal pathway in mediating inflammatory responses. However, with extensive and in-depth investigations, researchers have discovered that its role can be expanded to a variety of signaling mechanisms, biological processes, human diseases, and treatment options. In this review, we first scrutinize the research process of NF-κB signaling, and summarize the composition, activation, and regulatory mechanism of NF-κB signaling. We investigate the interaction of NF-κB signaling with other important pathways, including PI3K/AKT, MAPK, JAK-STAT, TGF-β, Wnt, Notch, Hedgehog, and TLR signaling. The physiological and pathological states of NF-κB signaling, as well as its intricate involvement in inflammation, immune regulation, and tumor microenvironment, are also explicated. Additionally, we illustrate how NF-κB signaling is involved in a variety of human diseases, including cancers, inflammatory and autoimmune diseases, cardiovascular diseases, metabolic diseases, neurological diseases, and COVID-19. Further, we discuss the therapeutic approaches targeting NF-κB signaling, including IKK inhibitors, monoclonal antibodies, proteasome inhibitors, nuclear translocation inhibitors, DNA binding inhibitors, TKIs, non-coding RNAs, immunotherapy, and CAR-T. Finally, we provide an outlook for research in the field of NF-κB signaling. We hope to present a stereoscopic, comprehensive NF-κB signaling that will inform future research and clinical practice.
Collapse
Affiliation(s)
- Qing Guo
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, No. 270, Dong'an Road, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yizi Jin
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, No. 270, Dong'an Road, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xinyu Chen
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med-X Stem Cell Research Center, Shanghai Cancer Institute & Department of Urology, Ren Ji Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200127, PR China
| | - Xiaomin Ye
- Department of Cardiology, the First Affiliated Hospital of Sun Yat-Sen University, 58 Zhongshan 2nd Road, Guangzhou, 510080, China
| | - Xin Shen
- Department of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Mingxi Lin
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, No. 270, Dong'an Road, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Cheng Zeng
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, No. 270, Dong'an Road, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Teng Zhou
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, No. 270, Dong'an Road, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jian Zhang
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, No. 270, Dong'an Road, Shanghai, 200032, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.
| |
Collapse
|
34
|
Zhang F, Ge Q, Meng J, Chen J, Liang C, Zhang M. Characterizing CD8+ TEMRA Cells in CP/CPPS Patients: Insights from Targeted Single-Cell Transcriptomic and Functional Investigations. Immunotargets Ther 2024; 13:111-121. [PMID: 38435982 PMCID: PMC10906729 DOI: 10.2147/itt.s451199] [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: 11/22/2023] [Accepted: 02/17/2024] [Indexed: 03/05/2024] Open
Abstract
Background The specific involvement of the CD8+ T effector memory RA (TEMRA) subset in patients with chronic prostatitis/chronic pelvic pain syndrome (CP/CPPS) has largely not been explored in the literature. Methods Targeted single-cell RNA sequencing (scRNA-seq) profiles were generated from peripheral blood mononuclear cells (PBMCs) obtained from two CP/CPPS patients and two healthy controls (HCs) in our recent study. Pseudotime series algorithms were used to reveal the differentiation trajectory, CellChat analysis was used to explore the communication between individual cells, and the SCENIC program was used to identify potential transcription factors (TFs). Based on the cosine similarity, clusters of differentially expressed genes (DEGs) were considered to be further enriched in different pathways. To confirm the functional role of the critical clusters, flow cytometry was employed. Results The results revealed the molecular landscape of these clusters, with TEMRA cells exhibiting pronounced cytokine-mediated signaling pathway enrichment. Pseudotime trajectory analysis further mapped the evolution from naïve T cells to that of TEMRA cells, elucidating the developmental pathways involved in the immune context. A significant finding from CellChat analysis was the differential expression of ligands and receptors, with CD8+ TEMRA cells showing enhanced signaling, particularly in the CP/CPPS context, compared to HCs. Flow cytometry confirmed these results, revealing a heightened proinflammatory cytokine profile in patients with chronic prostatitis-like symptoms (CP-LS), suggesting that TEMRA cells play a significant role in disease pathogenesis. TF profiling across the T-cell clusters identified key regulators of cellular identity, identifying novel therapeutic targets. Elevated TNF signaling activity in CD8+ TEMRA cells underscored the involvement of these cells in disease mechanisms. Conclusion This study elucidates the pivotal role of the CD8+ TEMRA cell subset in CP/CPPS, which is characterized by increased TNF signaling and proinflammatory factor expression, highlighting potential biomarkers and opening new avenues for therapeutic intervention.
Collapse
Affiliation(s)
- Fei Zhang
- Department of Urology, The First Affiliated Hospital of Anhui Medical University; Institute of Urology, Anhui Medical University; Anhui Province Key Laboratory of Urological and Andrological Diseases Research and Medical Transformation, Anhui Medical University, Hefei, 230022, People's Republic of China
| | - Qintao Ge
- Department of Urology, The First Affiliated Hospital of Anhui Medical University; Institute of Urology, Anhui Medical University; Anhui Province Key Laboratory of Urological and Andrological Diseases Research and Medical Transformation, Anhui Medical University, Hefei, 230022, People's Republic of China
| | - Jialin Meng
- Department of Urology, The First Affiliated Hospital of Anhui Medical University; Institute of Urology, Anhui Medical University; Anhui Province Key Laboratory of Urological and Andrological Diseases Research and Medical Transformation, Anhui Medical University, Hefei, 230022, People's Republic of China
| | - Jia Chen
- Department of Urology, The First Affiliated Hospital of Anhui Medical University; Institute of Urology, Anhui Medical University; Anhui Province Key Laboratory of Urological and Andrological Diseases Research and Medical Transformation, Anhui Medical University, Hefei, 230022, People's Republic of China
| | - Chaozhao Liang
- Department of Urology, The First Affiliated Hospital of Anhui Medical University; Institute of Urology, Anhui Medical University; Anhui Province Key Laboratory of Urological and Andrological Diseases Research and Medical Transformation, Anhui Medical University, Hefei, 230022, People's Republic of China
| | - Meng Zhang
- Department of Urology, The First Affiliated Hospital of Anhui Medical University; Institute of Urology, Anhui Medical University; Anhui Province Key Laboratory of Urological and Andrological Diseases Research and Medical Transformation, Anhui Medical University, Hefei, 230022, People's Republic of China
| |
Collapse
|
35
|
Katakai T, Okazaki T. A battle between two biological singularities: Immune response vs. cancer. Biophys Physicobiol 2024; 21:e211006. [PMID: 39175864 PMCID: PMC11338675 DOI: 10.2142/biophysico.bppb-v21.s006] [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: 12/28/2023] [Accepted: 02/07/2024] [Indexed: 08/24/2024] Open
Abstract
In a post-growth multicellular organism, the phenomenon in which a small number of rare cells can be the starting point for inducing a dramatic change in the entire system is considered a "biological singularity." The immune response and cancer can be regarded as singularity phenomena in mammals, but their nature is fundamentally different. The immune response is considered a "programmed" singularity, whereas cancer is an "unprogrammed" singularity. These two systems perpetually engage in a cycle of attack and defense within the organism. The outcome is depending on the wining system, which determines whether the individual experiences a state resembling light or darkness. However, the overall mechanism of the competition remains unclear and is expected to be elucidated with future innovations in bioimaging technologies. Immune checkpoint blockade therapy is a means by which the two singularity balances can be artificially manipulated; therefore, mechanistic insight is necessary for cancer treatment strategies. Altogether, these findings provide a different perspective crucial for understanding the behavior of dynamic cell populations in multicellular organisms.
Collapse
Affiliation(s)
- Tomoya Katakai
- Department of Immunology, Niigata University Graduate School of Medical and Dental Sciences, Niigata 950-8510, Japan
| | - Taku Okazaki
- Laboratory of Molecular Immunology, Institute for Quantitative Biosciences, The University of Tokyo, Tokyo, 113-0032, Japan
| |
Collapse
|
36
|
Sprent J, Boyman O. Optimising IL-2 for Cancer Immunotherapy. Immune Netw 2024; 24:e5. [PMID: 38455463 PMCID: PMC10917570 DOI: 10.4110/in.2024.24.e5] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 01/01/2024] [Accepted: 01/08/2024] [Indexed: 03/09/2024] Open
Abstract
The key role of T cells in cancer immunotherapy is well established and is highlighted by the remarkable capacity of Ab-mediated checkpoint blockade to overcome T-cell exhaustion and amplify anti-tumor responses. However, total or partial tumor remission following checkpoint blockade is still limited to only a few types of tumors. Hence, concerted attempts are being made to devise new methods for improving tumor immunity. Currently, much attention is being focused on therapy with IL-2. This cytokine is a powerful growth factor for T cells and optimises their effector functions. When used at therapeutic doses for cancer treatment, however, IL-2 is highly toxic. Nevertheless, recent work has shown that modifying the structure or presentation of IL-2 can reduce toxicity and lead to effective anti-tumor responses in synergy with checkpoint blockade. Here, we review the complex interaction of IL-2 with T cells: first during normal homeostasis, then during responses to pathogens, and finally in anti-tumor responses.
Collapse
Affiliation(s)
- Jonathan Sprent
- Immunology Division, Garvan Institute of Medical Research, Darlinghurst 2010, Australia
- St. Vincent’s Clinical School, University of New South Wales, Sydney 1466, Australia
- Menzies Institute of Medical Research, Hobart 7000, Australia
| | - Onur Boyman
- Department of Immunology, University Hospital Zurich, Zurich 8091, Switzerland
- Faculty of Medicine and Faculty of Science, University of Zurich, Zurich 8057, Switzerland
| |
Collapse
|
37
|
Cunha SMF, Lam S, Mallard B, Karrow NA, Cánovas Á. Genomic Regions Associated with Resistance to Gastrointestinal Nematode Parasites in Sheep-A Review. Genes (Basel) 2024; 15:187. [PMID: 38397178 PMCID: PMC10888242 DOI: 10.3390/genes15020187] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 01/27/2024] [Accepted: 01/27/2024] [Indexed: 02/25/2024] Open
Abstract
Gastrointestinal nematodes (GINs) can be a major constraint and global challenge to the sheep industry. These nematodes infect the small intestine and abomasum of grazing sheep, causing symptoms such as weight loss, diarrhea, hypoproteinemia, and anemia, which can lead to death. The use of anthelmintics to treat infected animals has led to GIN resistance, and excessive use of these drugs has resulted in residue traced in food and the environment. Resistance to GINs can be measured using multiple traits, including fecal egg count (FEC), Faffa Malan Chart scores, hematocrit, packed cell volume, eosinophilia, immunoglobulin (Ig), and dagginess scores. Genetic variation among animals exists, and understanding these differences can help identify genomic regions associated with resistance to GINs in sheep. Genes playing important roles in the immune system were identified in several studies in this review, such as the CFI and MUC15 genes. Results from several studies showed overlapping quantitative trait loci (QTLs) associated with multiple traits measuring resistance to GINs, mainly FEC. The discovery of genomic regions, positional candidate genes, and QTLs associated with resistance to GINs can help increase and accelerate genetic gains in sheep breeding programs and reveal the genetic basis and biological mechanisms underlying this trait.
Collapse
Affiliation(s)
- Samla Marques Freire Cunha
- Centre for Genetic Improvement of Livestock, Department of Animal Biosciences, University of Guelph, 50 Stone Rd E, Guelph, ON N1G 2W1, Canada; (S.M.F.C.); (S.L.); (B.M.); (N.A.K.)
| | - Stephanie Lam
- Centre for Genetic Improvement of Livestock, Department of Animal Biosciences, University of Guelph, 50 Stone Rd E, Guelph, ON N1G 2W1, Canada; (S.M.F.C.); (S.L.); (B.M.); (N.A.K.)
| | - Bonnie Mallard
- Centre for Genetic Improvement of Livestock, Department of Animal Biosciences, University of Guelph, 50 Stone Rd E, Guelph, ON N1G 2W1, Canada; (S.M.F.C.); (S.L.); (B.M.); (N.A.K.)
- Department of Pathobiology, University of Guelph, 50 Stone Rd E, Guelph, ON N1G 2W1, Canada
| | - Niel A. Karrow
- Centre for Genetic Improvement of Livestock, Department of Animal Biosciences, University of Guelph, 50 Stone Rd E, Guelph, ON N1G 2W1, Canada; (S.M.F.C.); (S.L.); (B.M.); (N.A.K.)
| | - Ángela Cánovas
- Centre for Genetic Improvement of Livestock, Department of Animal Biosciences, University of Guelph, 50 Stone Rd E, Guelph, ON N1G 2W1, Canada; (S.M.F.C.); (S.L.); (B.M.); (N.A.K.)
| |
Collapse
|
38
|
Martin-Salgado M, Ochoa-Echeverría A, Mérida I. Diacylglycerol kinases: A look into the future of immunotherapy. Adv Biol Regul 2024; 91:100999. [PMID: 37949728 DOI: 10.1016/j.jbior.2023.100999] [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: 10/23/2023] [Accepted: 11/01/2023] [Indexed: 11/12/2023]
Abstract
Cancer still represents the second leading cause of death right after cardiovascular diseases. According to the World Health Organization (WHO), cancer provoked around 10 million deaths in 2020, with lung and colon tumors accounting for the deadliest forms of cancer. As tumor cells become resistant to traditional therapeutic approaches, immunotherapy has emerged as a novel strategy for tumor control. T lymphocytes are key players in immune responses against tumors. Immunosurveillance allows identification, targeting and later killing of cancerous cells. Nevertheless, tumors evolve through different strategies to evade the immune response and spread in a process called metastasis. The ineffectiveness of traditional strategies to control tumor growth and expansion has led to novel approaches considering modulation of T cell activation and effector functions. Program death receptor 1 (PD-1) and cytotoxic T-lymphocyte antigen 4 (CTLA-4) showed promising results in the early 90s and nowadays are still being exploited together with other drugs for several cancer types. Other negative regulators of T cell activation are diacylglycerol kinases (DGKs) a family of enzymes that catalyze the conversion of diacylglycerol (DAG) into phosphatidic acid (PA). In T cells, DGKα and DGKζ limit the PLCγ/Ras/ERK axis thus attenuating DAG mediated signaling and T cell effector functions. Upregulation of either of both isoforms results in impaired Ras activation and anergy induction, whereas germline knockdown mice showed enhanced antitumor properties and more effective immune responses against pathogens. Here we review the mechanisms used by DGKs to ameliorate T cell activation and how inhibition could be used to reinvigorate T cell functions in cancer context. A better knowledge of the molecular mechanisms involved upon T cell activation will help to improve current therapies with DAG promoting agents.
Collapse
Affiliation(s)
- Miguel Martin-Salgado
- Department of Immunology and Oncology. National Centre for Biotechnology. Spanish Research Council (CNB-CSIC), Spain
| | - Ane Ochoa-Echeverría
- Department of Immunology and Oncology. National Centre for Biotechnology. Spanish Research Council (CNB-CSIC), Spain
| | - Isabel Mérida
- Department of Immunology and Oncology. National Centre for Biotechnology. Spanish Research Council (CNB-CSIC), Spain.
| |
Collapse
|
39
|
Wu C, Jiang ML, Pang T, Zhang CJ. T Cell Subsets and Immune Homeostasis. Methods Mol Biol 2024; 2782:39-63. [PMID: 38622391 DOI: 10.1007/978-1-0716-3754-8_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] [Indexed: 04/17/2024]
Abstract
T cells are a heterogeneous group of cells that can be classified into different subtypes according to different classification methods. The body's immune system has a highly complex and effective regulatory network that allows for the relative stability of immune system function. Maintaining proper T cell homeostasis is essential for promoting protective immunity and limiting autoimmunity and tumor formation. Among the T cell family members, more and more T cell subsets have gradually been characterized. In this chapter, we summarize the functions of some key T cell subsets and their impact on immune homeostasis.
Collapse
Affiliation(s)
- Chuyu Wu
- Department of Neurology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Screening, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, China Pharmaceutical University, Nanjing, China
| | - Mei-Ling Jiang
- Department of Neurology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Tao Pang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Screening, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, China Pharmaceutical University, Nanjing, China
| | - Cun-Jin Zhang
- Department of Neurology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| |
Collapse
|
40
|
Zhu S, Liu J, Patel V, Zhao X, Peng W, Xue HH. Antigen exposure reshapes chromatin architecture in central memory CD8 + T cells and imprints enhanced recall capacity. Proc Natl Acad Sci U S A 2023; 120:e2313476120. [PMID: 38085779 PMCID: PMC10742382 DOI: 10.1073/pnas.2313476120] [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: 08/05/2023] [Accepted: 11/07/2023] [Indexed: 12/18/2023] Open
Abstract
CD62L+ central memory CD8+ T (TCM) cells provide enhanced protection than naive cells; however, the underlying mechanism, especially the contribution of higher-order genomic organization, remains unclear. Systematic Hi-C analyses reveal that antigen-experienced CD8+ T cells undergo extensive rewiring of chromatin interactions (ChrInt), with TCM cells harboring specific interaction hubs compared with naive CD8+ T cells, as observed at cytotoxic effector genes such as Ifng and Tbx21. TCM cells also acquire de novo CTCF (CCCTC-binding factor) binding sites, which are not only strongly associated with TCM-specific hubs but also linked to increased activities of local gene promoters and enhancers. Specific ablation of CTCF in TCM cells impairs rapid induction of genes in cytotoxic program, energy supplies, transcription, and translation by recall stimulation. Therefore, acquisition of CTCF binding and ChrInt hubs by TCM cells serves as a chromatin architectural basis for their transcriptomic dynamics in primary response and for imprinting the code of "recall readiness" against secondary challenge.
Collapse
Affiliation(s)
- Shaoqi Zhu
- Department of Physics, The George Washington University, Washington, DC20052
| | - Jia Liu
- Center for Discovery and Innovation, Hackensack University Medical Center, Nutley, NJ07110
| | - Vanita Patel
- Center for Discovery and Innovation, Hackensack University Medical Center, Nutley, NJ07110
| | - Xiuyi Zhao
- Center for Discovery and Innovation, Hackensack University Medical Center, Nutley, NJ07110
- Solon High School, Solon, OH44139
| | - Weiqun Peng
- Department of Physics, The George Washington University, Washington, DC20052
| | - Hai-Hui Xue
- Center for Discovery and Innovation, Hackensack University Medical Center, Nutley, NJ07110
- New Jersey Veterans Affairs Health Care System, East Orange, NJ07018
| |
Collapse
|
41
|
Elalouf A, Elalouf H, Rosenfeld A. Modulatory immune responses in fungal infection associated with organ transplant - advancements, management, and challenges. Front Immunol 2023; 14:1292625. [PMID: 38143753 PMCID: PMC10748506 DOI: 10.3389/fimmu.2023.1292625] [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: 09/11/2023] [Accepted: 11/20/2023] [Indexed: 12/26/2023] Open
Abstract
Organ transplantation stands as a pivotal achievement in modern medicine, offering hope to individuals with end-stage organ diseases. Advancements in immunology led to improved organ transplant survival through the development of immunosuppressants, but this heightened susceptibility to fungal infections with nonspecific symptoms in recipients. This review aims to establish an intricate balance between immune responses and fungal infections in organ transplant recipients. It explores the fundamental immune mechanisms, recent advances in immune response dynamics, and strategies for immune modulation, encompassing responses to fungal infections, immunomodulatory approaches, diagnostics, treatment challenges, and management. Early diagnosis of fungal infections in transplant patients is emphasized with the understanding that innate immune responses could potentially reduce immunosuppression and promise efficient and safe immuno-modulating treatments. Advances in fungal research and genetic influences on immune-fungal interactions are underscored, as well as the potential of single-cell technologies integrated with machine learning for biomarker discovery. This review provides a snapshot of the complex interplay between immune responses and fungal infections in organ transplantation and underscores key research directions.
Collapse
Affiliation(s)
- Amir Elalouf
- Department of Management, Bar-Ilan University, Ramat Gan, Israel
| | - Hadas Elalouf
- Information Science Department, Bar-Ilan University, Ramat Gan, Israel
| | - Ariel Rosenfeld
- Information Science Department, Bar-Ilan University, Ramat Gan, Israel
| |
Collapse
|
42
|
Macalinao ML, Inoue SI, Tsogtsaikhan S, Matsumoto H, Bayarsaikhan G, Jian JY, Kimura K, Yasumizu Y, Inoue T, Yoshida H, Hafalla J, Kimura D, Yui K. IL-27 produced during acute malaria infection regulates Plasmodium-specific memory CD4 + T cells. EMBO Mol Med 2023; 15:e17713. [PMID: 37855243 DOI: 10.15252/emmm.202317713] [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: 03/14/2023] [Revised: 09/27/2023] [Accepted: 09/28/2023] [Indexed: 10/20/2023] Open
Abstract
Malaria infection elicits both protective and pathogenic immune responses, and IL-27 is a critical cytokine that regulate effector responses during infection. Here, we identified a critical window of CD4+ T cell responses that is targeted by IL-27. Neutralization of IL-27 during acute infection with Plasmodium chabaudi expanded specific CD4+ T cells, which were maintained at high levels thereafter. In the chronic phase, Plasmodium-specific CD4+ T cells in IL-27-neutralized mice consisted mainly of CD127+ KLRG1- and CD127- KLRG1+ subpopulations that displayed distinct cytokine production, proliferative capacity, and are maintained in a manner independent of active infection. Single-cell RNA-seq analysis revealed that these CD4+ T cell subsets formed independent clusters that express unique Th1-type genes. These IL-27-neutralized mice exhibited enhanced cellular and humoral immune responses and protection. These findings demonstrate that IL-27, which is produced during the acute phase of malaria infection, inhibits the development of unique Th1 memory precursor CD4+ T cells, suggesting potential implications for the development of vaccines and other strategic interventions.
Collapse
Affiliation(s)
- Maria Lourdes Macalinao
- School of Tropical Medicine and Global Health, Nagasaki University, Nagasaki, Japan
- Department of Infection Biology, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, UK
| | - Shin-Ichi Inoue
- Division of Immunology, Department of Molecular Microbiology and Immunology, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Sanjaadorj Tsogtsaikhan
- Division of Immunology, Department of Molecular Microbiology and Immunology, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Hirotaka Matsumoto
- School of Information and Data Sciences, Nagasaki University, Nagasaki, Japan
| | - Ganchimeg Bayarsaikhan
- Division of Immunology, Department of Molecular Microbiology and Immunology, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Jiun-Yu Jian
- Division of Immunology, Department of Molecular Microbiology and Immunology, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Kazumi Kimura
- Division of Immunology, Department of Molecular Microbiology and Immunology, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Yoshiaki Yasumizu
- Department of Experimental Immunology, Immunology Frontier Research Center, Osaka University, Suita, Japan
- Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives (OTRI), Osaka University, Osaka, Japan
| | - Tsuyoshi Inoue
- Department of Physiology of Visceral Function and Body Fluid, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Hiroki Yoshida
- Division of Molecular and Cellular Immunoscience, Department of Biomolecular Sciences, Faculty of Medicine, Saga University, Saga, Japan
| | - Julius Hafalla
- Department of Infection Biology, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, UK
| | - Daisuke Kimura
- Division of Immunology, Department of Molecular Microbiology and Immunology, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Katsuyuki Yui
- School of Tropical Medicine and Global Health, Nagasaki University, Nagasaki, Japan
- Division of Immunology, Department of Molecular Microbiology and Immunology, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
- Shionogi Global Infectious Diseases Division, Institute of Tropical Medicine, Nagasaki University, Nagasaki, Japan
| |
Collapse
|
43
|
Brito-de-Sousa JP, Lima-Silva ML, Costa-Rocha IAD, Júnior LRADO, Campi-Azevedo AC, Peruhype-Magalhães V, Quetz JDS, Coelho-Dos-Reis JGA, Costa-Pereira C, Garcia CC, Antonelli LRDV, Fonseca CT, Lemos JAC, Mambrini JVDM, Souza-Fagundes EM, Teixeira-Carvalho A, Faria AMDC, Gomes AO, Torres KCDL, Martins-Filho OA. Rhythmic profile of memory T and B-cells along childhood and adolescence. Sci Rep 2023; 13:20978. [PMID: 38017254 PMCID: PMC10684863 DOI: 10.1038/s41598-023-48115-3] [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: 08/02/2023] [Accepted: 11/22/2023] [Indexed: 11/30/2023] Open
Abstract
Immunobiography describes the life-long effects of exogenous or endogenous stimuli on remodeling of immune cell biology, including the development of memory T and B-cells. The present study aimed at investigating the rhythms of changes in phenotypic features of memory T and B-cells along childhood and adolescence. A descriptive-observational investigation was conducted including 812 healthy volunteers, clustered into six consecutive age groups (9Mths-1Yr; 2Yrs; 3-4Yrs; 5-7Yrs; 8-10Yrs; 11-18Yrs). Immunophenotypic analysis of memory T-cell (CD4+ and CD8+) and B-cell subsets were performed by flow cytometry. The results pointed out that memory-related biomarkers of T and B-cells displayed a bimodal profile along healthy childhood and adolescence, regardless of sex. The first stage of changes occurs around 2Yrs, with predominance of naive cells, while the second and more prominent wave occurs around the age 8-10Yrs, with the prevalence of memory phenotypes. The neighborhood connectivity profile analysis demonstrated that the number of correlations reaches a peak at 11-18Yrs and lower values along the childhood. Males presented higher and conserved number of correlations when compared to females. Altogether, our results provide new insights into immunobiography and a better understanding of interactions among the cellular subsets studied here during childhood and adolescence.
Collapse
Affiliation(s)
- Joaquim Pedro Brito-de-Sousa
- Programa de Pós-graduação em Imunologia e Parasitologia Aplicadas, Universidade Federal de Uberlândia, Uberlândia, MG, Brazil
| | - Maria Luiza Lima-Silva
- Instituto René Rachou, Fundação Oswaldo Cruz (FIOCRUZ-Minas), Avenida Augusto de Lima, 1715, Barro Preto, Belo Horizonte, MG, 30190-002, Brazil
| | - Ismael Artur da Costa-Rocha
- Instituto René Rachou, Fundação Oswaldo Cruz (FIOCRUZ-Minas), Avenida Augusto de Lima, 1715, Barro Preto, Belo Horizonte, MG, 30190-002, Brazil
| | | | - Ana Carolina Campi-Azevedo
- Instituto René Rachou, Fundação Oswaldo Cruz (FIOCRUZ-Minas), Avenida Augusto de Lima, 1715, Barro Preto, Belo Horizonte, MG, 30190-002, Brazil
| | - Vanessa Peruhype-Magalhães
- Instituto René Rachou, Fundação Oswaldo Cruz (FIOCRUZ-Minas), Avenida Augusto de Lima, 1715, Barro Preto, Belo Horizonte, MG, 30190-002, Brazil
| | - Josiane da Silva Quetz
- Instituto René Rachou, Fundação Oswaldo Cruz (FIOCRUZ-Minas), Avenida Augusto de Lima, 1715, Barro Preto, Belo Horizonte, MG, 30190-002, Brazil
- Universidade Professor Edson Antônio Velano, UNIFENAS, Belo Horizonte, MG, Brazil
| | - Jordana Grazziela Alves Coelho-Dos-Reis
- Laboratório de Virologia Básica e Aplicada, Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Christiane Costa-Pereira
- Instituto René Rachou, Fundação Oswaldo Cruz (FIOCRUZ-Minas), Avenida Augusto de Lima, 1715, Barro Preto, Belo Horizonte, MG, 30190-002, Brazil
| | - Cristiana Couto Garcia
- Instituto René Rachou, Fundação Oswaldo Cruz (FIOCRUZ-Minas), Avenida Augusto de Lima, 1715, Barro Preto, Belo Horizonte, MG, 30190-002, Brazil
| | - Lis Ribeiro do Vale Antonelli
- Instituto René Rachou, Fundação Oswaldo Cruz (FIOCRUZ-Minas), Avenida Augusto de Lima, 1715, Barro Preto, Belo Horizonte, MG, 30190-002, Brazil
| | - Cristina Toscano Fonseca
- Instituto René Rachou, Fundação Oswaldo Cruz (FIOCRUZ-Minas), Avenida Augusto de Lima, 1715, Barro Preto, Belo Horizonte, MG, 30190-002, Brazil
| | | | - Juliana Vaz de Melo Mambrini
- Instituto René Rachou, Fundação Oswaldo Cruz (FIOCRUZ-Minas), Avenida Augusto de Lima, 1715, Barro Preto, Belo Horizonte, MG, 30190-002, Brazil
| | - Elaine Maria Souza-Fagundes
- Laboratório de Virologia Básica e Aplicada, Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Andréa Teixeira-Carvalho
- Instituto René Rachou, Fundação Oswaldo Cruz (FIOCRUZ-Minas), Avenida Augusto de Lima, 1715, Barro Preto, Belo Horizonte, MG, 30190-002, Brazil
| | - Ana Maria de Caetano Faria
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | | | - Karen Cecília de Lima Torres
- Instituto René Rachou, Fundação Oswaldo Cruz (FIOCRUZ-Minas), Avenida Augusto de Lima, 1715, Barro Preto, Belo Horizonte, MG, 30190-002, Brazil.
- Universidade Professor Edson Antônio Velano, UNIFENAS, Belo Horizonte, MG, Brazil.
| | - Olindo Assis Martins-Filho
- Programa de Pós-graduação em Imunologia e Parasitologia Aplicadas, Universidade Federal de Uberlândia, Uberlândia, MG, Brazil.
- Instituto René Rachou, Fundação Oswaldo Cruz (FIOCRUZ-Minas), Avenida Augusto de Lima, 1715, Barro Preto, Belo Horizonte, MG, 30190-002, Brazil.
| |
Collapse
|
44
|
Huldani H, Abdul-Jabbar Ali S, Al-Dolaimy F, Hjazi A, Denis Andreevich N, Oudaha KH, Almulla AF, Alsaalamy A, Kareem Oudah S, Mustafa YF. The potential role of interleukins and interferons in ovarian cancer. Cytokine 2023; 171:156379. [PMID: 37757536 DOI: 10.1016/j.cyto.2023.156379] [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: 09/03/2023] [Revised: 09/20/2023] [Accepted: 09/21/2023] [Indexed: 09/29/2023]
Abstract
Ovarian cancer poses significant challenges and remains a highly lethal disease with limited treatment options. In the context of ovarian cancer, interleukins (ILs) and interferons (IFNs), important cytokines that play crucial roles in regulating the immune system, have emerged as significant factors influencing its development. This article provides a comprehensive review of the involvement of various ILs, including those from the IL-1 family, IL-2 family, IL-6 family, IL-8 family, IL-10 family, and IL-17 family, in ovarian cancer. The focus is on their impact on tumor growth, metastasis, and their role in evading immune responses within the tumor microenvironment. Additionally, the article conducts an in-depth examination of the oncogenic or antitumor roles of each IL in the context of ovarian cancer pathogenesis and progression. Besides, we elucidated the enhancements in the treatment of ovarian cancer through the utilization of type-I IFN and type-II IFN. Recent research has shed light on the intricate mechanisms through which specific ILs and IFNs contribute to the advancement of the disease. By incorporating recent findings, this review also seeks to inspire further investigations into unexplored mechanisms, fostering ongoing research to develop more effective therapeutic strategies for ovarian cancer. Moreover, through an in-depth analysis of IL- and IFN-associated clinical trials, we have highlighted their promising potential of in the treatment of ovarian cancer. These clinical trials serve to reinforce the significant outlook for utilizing ILs and IFNs as therapeutic agents in combating this disease.
Collapse
Affiliation(s)
- Huldani Huldani
- Department of Physiology, Faculty of Medicine, Lambung Mangkurat University, Banjarmasin, South Kalimantan, Indonesia
| | | | | | - Ahmed Hjazi
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Prince Sattam bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia
| | | | - Khulood H Oudaha
- Pharmaceutical Chemistry Department, College of Pharmacy, Al-Ayen University, Thi-Qar, Iraq
| | - Abbas F Almulla
- College of Technical Engineering, the Islamic University, Najaf, Iraq; College of Technical Engineering, the Islamic University of Al Diwaniyah, Iraq; College of Technical Engineering, the Islamic University of Babylon, Iraq
| | - Ali Alsaalamy
- College of Technical Engineering, Imam Ja'afar Al-Sadiq University, Al-Muthanna 66002, Iraq
| | - Shamam Kareem Oudah
- College of Pharmacy, National University of Science and Technology, Dhi Qar, Iraq
| | - Yasser Fakri Mustafa
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Mosul, Mosul 41001, Iraq
| |
Collapse
|
45
|
Liu XQ, Zhou PL, Yin XY, Wang AX, Wang DH, Yang Y, Liu Q. Circulating inflammatory cytokines and psoriasis risk: A systematic review and meta-analysis. PLoS One 2023; 18:e0293327. [PMID: 37883350 PMCID: PMC10602280 DOI: 10.1371/journal.pone.0293327] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 10/10/2023] [Indexed: 10/28/2023] Open
Abstract
BACKGROUND Psoriasis is a systemic immune-mediated chronic inflammatory skin disease; its systemic manifestations and periodic recurrence negatively affect a patient's quality of life. Inflammatory cytokines are known to have an important role in the onset and progression of psoriasis, however, data on the association between circulating inflammatory cytokines and psoriasis risk is inconclusive. Here, we explore the relevance of circulating proinflammatory factors to the pathogenesis of psoriasis using a meta-analysis. OBJECTIVE To explore the association between circulating levels of inflammatory factors and psoriasis to elucidate the mechanisms underlying psoriasis and improve clinical diagnosis and treatment. METHODS We systematically retrieved articles published in PubMed, EMBASE, the Cochrane Library and the Web of Science from the establishment of each database to January 2023. The standard mean difference (SMD) in cytokine levels of individuals with psoriasis and healthy controls was used to check for correlations between circulating inflammatory factor levels and psoriasis. RESULTS Fifty-seven studies, with data from 2838 patients, were retrieved and included in the meta-analysis. Eleven inflammatory factors were studied (circulating interleukin-2 (IL-2), IL-4, IL-12, IL-17, IL-18, IL-22, IL-23, IL-35, IL-36, transforming growth factor-beta (TGF-β) and gamma-interferon (IFN-γ)). Of these, IL-2 [SMD = 1.29 (95% CI: 0.61-1.97; P <0.001)], IL-17 [SMD = 0.71 (95% CI: 0.12-1.30; P = 0.018)], IL-18 [SMD = 1.27 (95% CI: 0.64-1.90; P <0.001)], and IFN-γ [SMD = 1.90 (95% CI: 1.27-2.52; P <0.001)] levels had significant correlations with psoriasis. CONCLUSION Increased serum concentrations of the circulating inflammatory cytokines IL-2, IL-17, IL-18 and IFN-γ were significantly correlated with psoriasis.
Collapse
Affiliation(s)
- Xiao-Qing Liu
- Department of Dermatology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Pei-Lin Zhou
- Department of Dermatology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Xin-Yu Yin
- Department of Dermatology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Ai-Xue Wang
- Department of Dermatology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Da-Hu Wang
- Department of Dermatology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Yun Yang
- Department of Dermatology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Qiang Liu
- Department of Dermatology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| |
Collapse
|
46
|
Sengupta S, Shaw SK, Chatterjee S, Bhattacharya G, Barik PK, Chattopadhyay S, Devadas S. Perturbations in spike-specific peripheral T follicular helper cells in SARS-CoV2 breakthrough convalescent individuals immunized by BBV152 vaccine. J Med Virol 2023; 95:e29053. [PMID: 37650214 DOI: 10.1002/jmv.29053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 07/22/2023] [Accepted: 08/14/2023] [Indexed: 09/01/2023]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-Cov2) infection has caused an increase in mortality and morbidity, but with vaccination, the disease severity has significantly reduced. With the emergence of various variants of concern (VOCs), the vaccine breakthrough infection has also increased. Here we studied circulating spike-specific T follicular response (cTfh) in infection-naïve vaccinees and convalescent vaccinees (individuals who got the Delta breakthrough infection after two doses of BBV152 vaccine) to understand their response as they are the most crucial cells that are involved in vaccine-mediated protection by helping in B-cell maturation. Our results indicated that cTfh cells in both the groups recognized the wild-type and Delta spike protein but memory response to the wild-type spike was superior in infection-naïve than in the convalescent group. The cytokine response, particularly interleukin-21 (IL-21) from cTfh, was also higher in infection-naïve than in convalescent vaccinees, indicating a dampened cTfh response in convalescent vaccinees after breakthrough infection. Also, there was a positive correlation between IL-21 from cTfh cells and neutralizing antibodies of infection-naïve vaccinees. Multiple cytokine analysis also revealed higher inflammation in convalescent vaccinees. Our data indicated that the necessity of a third booster dose may be individual-specific depending on the steady-state functional phenotype of immune cells.
Collapse
Affiliation(s)
- Soumya Sengupta
- Institute of Life Science, Nalco Square, Bhubaneswar, Odisha, India
- Regional Centre for Biotechnology (RCB), 3rd Milestone, Faridabad-Gurgaon, Haryana, India
- T cell and Immune Response Lab, Department of Infectious Disease Biology, Institute of Life Science (Autonomous Institute of Dept of Biotechnology, Govt. of India), Bhubaneswar, Odisha, India
| | - Shubham K Shaw
- Institute of Life Science, Nalco Square, Bhubaneswar, Odisha, India
- Regional Centre for Biotechnology (RCB), 3rd Milestone, Faridabad-Gurgaon, Haryana, India
- T cell and Immune Response Lab, Department of Infectious Disease Biology, Institute of Life Science (Autonomous Institute of Dept of Biotechnology, Govt. of India), Bhubaneswar, Odisha, India
| | - Sanchari Chatterjee
- Institute of Life Science, Nalco Square, Bhubaneswar, Odisha, India
- Regional Centre for Biotechnology (RCB), 3rd Milestone, Faridabad-Gurgaon, Haryana, India
- Molecular Virology Lab, Institute of Life Science, Nalco Square, Bhubaneswar, Odisha, India
| | - Gargee Bhattacharya
- Institute of Life Science, Nalco Square, Bhubaneswar, Odisha, India
- Regional Centre for Biotechnology (RCB), 3rd Milestone, Faridabad-Gurgaon, Haryana, India
- T cell and Immune Response Lab, Department of Infectious Disease Biology, Institute of Life Science (Autonomous Institute of Dept of Biotechnology, Govt. of India), Bhubaneswar, Odisha, India
| | - Prakash K Barik
- Institute of Life Science, Nalco Square, Bhubaneswar, Odisha, India
- T cell and Immune Response Lab, Department of Infectious Disease Biology, Institute of Life Science (Autonomous Institute of Dept of Biotechnology, Govt. of India), Bhubaneswar, Odisha, India
| | - Soma Chattopadhyay
- Institute of Life Science, Nalco Square, Bhubaneswar, Odisha, India
- Regional Centre for Biotechnology (RCB), 3rd Milestone, Faridabad-Gurgaon, Haryana, India
- Molecular Virology Lab, Institute of Life Science, Nalco Square, Bhubaneswar, Odisha, India
| | - Satish Devadas
- Institute of Life Science, Nalco Square, Bhubaneswar, Odisha, India
- Regional Centre for Biotechnology (RCB), 3rd Milestone, Faridabad-Gurgaon, Haryana, India
- T cell and Immune Response Lab, Department of Infectious Disease Biology, Institute of Life Science (Autonomous Institute of Dept of Biotechnology, Govt. of India), Bhubaneswar, Odisha, India
| |
Collapse
|
47
|
Mao Y, Ge H, Chen W, Wang Y, Liu H, Li Z, Bai Y, Wang D, Yu Y, Zhen Q, Li B, Sun L. RasGRP1 influences imiquimod-induced psoriatic inflammation via T-cell activation in mice. Int Immunopharmacol 2023; 122:110590. [PMID: 37429143 DOI: 10.1016/j.intimp.2023.110590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 06/21/2023] [Accepted: 06/29/2023] [Indexed: 07/12/2023]
Abstract
The vascular endothelial growth factor (VEGF) signal transduction pathway has been shown to be a potential target for the treatment of psoriasis. Ras guanyl-releasing protein 1 (RasGRP1), a downstream target gene of VEGF, regulates the development, homeostasis, and differentiation of T cells, but the contribution of RasGRP1 to psoriasis is limited. In this manuscript, we aimed to investigate the role of RasGRP1 in psoriasis. The RNA-Seq transcriptome sequencing data from the mouse model of psoriasis treated with IMQ (imiquimod) were analyzed. The effect of RasGRP1 was investigated through in vivo injection of activators or small molecular inhibitors, as well as adeno-associated virus injections. Gene knockout and NB-UVB (narrow-band ultraviolet B) treatments were utilized to interfere with the psoriatic mouse model. By transfection of lentivirus in vitro, the effect of RasGRP1 gene function on the secretion of psoriasis-related cytokines by T cells was confirmed. We showed that cutaneous VEGF and RasGRP1 were strongly activated in human psoriatic lesions and the skin of mice with IMQ-induced psoriasis. RasGRP1 deficiency and overexpression influence IMQ-induced psoriasis-like manifestations and skin inflammation in mice. VEGF, secreted mainly by epidermal cells, mediates psoriatic inflammation through the RasGRP1-AKT-NF-κB pathway. RasGRP1 is required for psoriasis development mediated by VEGF. These results confirmed the role of RasGRP1 in the pathogenesis of psoriasis and provided potential targets for clinical psoriasis treatment.
Collapse
Affiliation(s)
- Yiwen Mao
- Department of Dermatology, the First Affiliated Hospital of Anhui Medical University, Hefei, China; Institute of Dermatology, Anhui Medical University, Hefei, China; Key Laboratory of Dermatology (Anhui Medical University), Ministry of Education, Hefei, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, China; Anhui Provincial Institute of Translational Medicine, Hefei, China
| | - Huiyao Ge
- Department of Dermatology, the First Affiliated Hospital of Anhui Medical University, Hefei, China; Institute of Dermatology, Anhui Medical University, Hefei, China; Key Laboratory of Dermatology (Anhui Medical University), Ministry of Education, Hefei, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, China; Anhui Provincial Institute of Translational Medicine, Hefei, China
| | - Weiwei Chen
- Department of Dermatology, the First Affiliated Hospital of Anhui Medical University, Hefei, China; Institute of Dermatology, Anhui Medical University, Hefei, China; Key Laboratory of Dermatology (Anhui Medical University), Ministry of Education, Hefei, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, China; Anhui Provincial Institute of Translational Medicine, Hefei, China
| | - YiRui Wang
- Department of Dermatology, the First Affiliated Hospital of Anhui Medical University, Hefei, China; Institute of Dermatology, Anhui Medical University, Hefei, China; Key Laboratory of Dermatology (Anhui Medical University), Ministry of Education, Hefei, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, China; Anhui Provincial Institute of Translational Medicine, Hefei, China
| | - Hao Liu
- Department of Dermatology, the First Affiliated Hospital of Anhui Medical University, Hefei, China; Institute of Dermatology, Anhui Medical University, Hefei, China; Key Laboratory of Dermatology (Anhui Medical University), Ministry of Education, Hefei, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, China; Anhui Provincial Institute of Translational Medicine, Hefei, China
| | - Zhuo Li
- Department of Dermatology, the First Affiliated Hospital of Anhui Medical University, Hefei, China; Institute of Dermatology, Anhui Medical University, Hefei, China; Key Laboratory of Dermatology (Anhui Medical University), Ministry of Education, Hefei, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, China; Anhui Provincial Institute of Translational Medicine, Hefei, China
| | - Yuanming Bai
- Department of Dermatology, the First Affiliated Hospital of Anhui Medical University, Hefei, China; Institute of Dermatology, Anhui Medical University, Hefei, China; Key Laboratory of Dermatology (Anhui Medical University), Ministry of Education, Hefei, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, China; Anhui Provincial Institute of Translational Medicine, Hefei, China
| | - Daiyue Wang
- Department of Dermatology, the First Affiliated Hospital of Anhui Medical University, Hefei, China; Institute of Dermatology, Anhui Medical University, Hefei, China; Key Laboratory of Dermatology (Anhui Medical University), Ministry of Education, Hefei, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, China; Anhui Provincial Institute of Translational Medicine, Hefei, China
| | - Yafen Yu
- Department of Dermatology, the First Affiliated Hospital of Anhui Medical University, Hefei, China; Institute of Dermatology, Anhui Medical University, Hefei, China; Key Laboratory of Dermatology (Anhui Medical University), Ministry of Education, Hefei, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, China; Anhui Provincial Institute of Translational Medicine, Hefei, China
| | - Qi Zhen
- Department of Dermatology, the First Affiliated Hospital of Anhui Medical University, Hefei, China; Institute of Dermatology, Anhui Medical University, Hefei, China; Key Laboratory of Dermatology (Anhui Medical University), Ministry of Education, Hefei, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, China; Anhui Provincial Institute of Translational Medicine, Hefei, China
| | - Bao Li
- Integrated Laboratory, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Liangdan Sun
- Department of Dermatology, the First Affiliated Hospital of Anhui Medical University, Hefei, China; Key Laboratory of Dermatology (Anhui Medical University), Ministry of Education, Hefei, China; Health Science Center, North China University of Science and Technology, Tangshan 063210, China; North China University of Science and Technology Affiliated Hospital, Tangshan 063000, China; Inflammation and Immune Diseases Laboratory of North China University of Science and Technology, Tangshan 063210, China; School of Public Health, North China University of Science and Technology, Tangshan, Hebei, 063210, China.
| |
Collapse
|
48
|
Mise-Omata S, Ando M, Srirat T, Nakagawara K, Hayakawa T, Iizuka-Koga M, Nishimasu H, Nureki O, Ito M, Yoshimura A. SOCS3 deletion in effector T cells confers an anti-tumorigenic role of IL-6 to the pro-tumorigenic cytokine. Cell Rep 2023; 42:112940. [PMID: 37582370 DOI: 10.1016/j.celrep.2023.112940] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 06/26/2023] [Accepted: 07/20/2023] [Indexed: 08/17/2023] Open
Abstract
Interleukin (IL)-6 is abundantly expressed in the tumor microenvironment and is associated with poor patient outcomes. Here, we demonstrate that the deletion of the suppressor of cytokine signaling 3 (SOCS3) in T cells potentiates anti-tumor immune responses by conferring the anti-tumorigenic function of IL-6 in mouse and human models. In Socs3-deficient CD8+ T cells, IL-6 upregulates the expression of type I interferon (IFN)-regulated genes and enhances the anti-tumor effector function of T cells, while also modifying mitochondrial fitness to increase mitochondrial membrane potential and reactive oxygen species (ROS) levels and to promote metabolic glycolysis in the energy state. Furthermore, Socs3 deficiency reduces regulatory T cells and increases T helper 1 (Th1) cells. SOCS3 knockdown in human chimeric antigen receptor T (CAR-T) cells exhibits a strong anti-tumor response in humanized mice. Thus, genetic disruption of SOCS3 offers an avenue to improve the therapeutic efficacy of adoptive T cell therapy.
Collapse
Affiliation(s)
- Setsuko Mise-Omata
- Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo, Japan.
| | - Makoto Ando
- Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo, Japan
| | - Tanakorn Srirat
- Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo, Japan
| | - Kensuke Nakagawara
- Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo, Japan
| | - Taeko Hayakawa
- Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo, Japan
| | - Mana Iizuka-Koga
- Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo, Japan
| | - Hiroshi Nishimasu
- Structural Biology Division, Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan
| | - Osamu Nureki
- Department of Biological Science, Graduate School of Science, The University of Tokyo, Tokyo, Japan
| | - Minako Ito
- Division of Allergy and Immunology, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Akihiko Yoshimura
- Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo, Japan.
| |
Collapse
|
49
|
Schoonhoven AOV, de Bruijn MJ, Stikker B, Brouwer RW, Braunstahl GJ, van IJcken WF, Graf T, Huylebroeck D, Hendriks RW, Stik G, Stadhouders R. 3D chromatin reprogramming primes human memory T H2 cells for rapid recall and pathogenic dysfunction. Sci Immunol 2023; 8:eadg3917. [PMID: 37418545 PMCID: PMC7617366 DOI: 10.1126/sciimmunol.adg3917] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Accepted: 06/13/2023] [Indexed: 07/09/2023]
Abstract
Memory T cells provide long-lasting defense responses through their ability to rapidly reactivate, but how they efficiently "recall" an inflammatory transcriptional program remains unclear. Here, we show that human CD4+ memory T helper 2 (TH2) cells carry a chromatin landscape synergistically reprogrammed at both one-dimensional (1D) and 3D levels to accommodate recall responses, which is absent in naive T cells. In memory TH2 cells, recall genes were epigenetically primed through the maintenance of transcription-permissive chromatin at distal (super)enhancers organized in long-range 3D chromatin hubs. Precise transcriptional control of key recall genes occurred inside dedicated topologically associating domains ("memory TADs"), in which activation-associated promoter-enhancer interactions were preformed and exploited by AP-1 transcription factors to promote rapid transcriptional induction. Resting memory TH2 cells from patients with asthma showed premature activation of primed recall circuits, linking aberrant transcriptional control of recall responses to chronic inflammation. Together, our results implicate stable multiscale reprogramming of chromatin organization as a key mechanism underlying immunological memory and dysfunction in T cells.
Collapse
Affiliation(s)
- Anne Onrust-van Schoonhoven
- Department of Pulmonary Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
- Department of Cell Biology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Marjolein J.W. de Bruijn
- Department of Pulmonary Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Bernard Stikker
- Department of Pulmonary Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Rutger W.W. Brouwer
- Center for Biomics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Gert-Jan Braunstahl
- Department of Pulmonary Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
- Department of Respiratory Medicine, Franciscus Gasthuis and Vlietland, Rotterdam, Netherlands
| | | | - Thomas Graf
- Centre for Genomic Regulation (CRG) and Institute of Science and Technology (BIST), Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Danny Huylebroeck
- Department of Cell Biology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Rudi W. Hendriks
- Department of Pulmonary Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Grégoire Stik
- Josep Carreras Leukaemia Research Institute (IJC), Badalona, Spain
| | - Ralph Stadhouders
- Department of Pulmonary Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
- Department of Cell Biology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| |
Collapse
|
50
|
Shireman JM, Gonugunta N, Zhao L, Pattnaik A, Distler E, Her S, Wang X, Das R, Galipeau J, Dey M. GM-CSF and IL-7 fusion cytokine engineered tumor vaccine generates long-term Th-17 memory cells and increases overall survival in aged syngeneic mouse models of glioblastoma. Aging Cell 2023; 22:e13864. [PMID: 37165998 PMCID: PMC10352573 DOI: 10.1111/acel.13864] [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: 02/02/2023] [Revised: 04/18/2023] [Accepted: 04/20/2023] [Indexed: 05/12/2023] Open
Abstract
Age-related immune dysfunctions, such as decreased T-cell output, are closely related to pathologies like cancers and lack of vaccine efficacy among the elderly. Engineered fusokine, GIFT-7, a fusion of interleukin 7 (IL-7) and GM-CSF, can reverse aging-related lymphoid organ atrophy. We generated a GIFT-7 fusokine tumor vaccine and employed it in aged syngeneic mouse models of glioblastoma and found that peripheral vaccination with GIFT-7TVax resulted in thymic regeneration and generated durable long-term antitumor immunity specifically in aged mice. Global cytokine analysis showed increased pro-inflammatory cytokines including IL-1β in the vaccinated group that resulted in hyperactivation of dendritic cells. In addition, GIFT-7 vaccination resulted in increased T-cell trafficking to the brain and robust Th-17 long-term effector memory T-cell formation. TCR-seq analysis showed increased productive frequency among detected rearrangements within the vaccinated group. Overall, our data demonstrate that aging immune system can be therapeutically augmented to generate lasting antitumor immunity.
Collapse
Affiliation(s)
- Jack M. Shireman
- Department of NeurosurgeryUniversity of Wisconsin School of Medicine & Public Health, UW Carbone Cancer Center,MadisonWisconsinUSA
| | - Nikita Gonugunta
- Department of NeurosurgeryUniversity of Wisconsin School of Medicine & Public Health, UW Carbone Cancer Center,MadisonWisconsinUSA
| | - Lei Zhao
- Department of NeurosurgeryUniversity of Wisconsin School of Medicine & Public Health, UW Carbone Cancer Center,MadisonWisconsinUSA
| | - Akshita Pattnaik
- Department of NeurosurgeryUniversity of Wisconsin School of Medicine & Public Health, UW Carbone Cancer Center,MadisonWisconsinUSA
| | - Emily Distler
- Department of NeurosurgeryUniversity of Wisconsin School of Medicine & Public Health, UW Carbone Cancer Center,MadisonWisconsinUSA
| | - Skyler Her
- Department of NeurosurgeryUniversity of Wisconsin School of Medicine & Public Health, UW Carbone Cancer Center,MadisonWisconsinUSA
| | - Xiaohu Wang
- Department of NeurosurgeryUniversity of Wisconsin School of Medicine & Public Health, UW Carbone Cancer Center,MadisonWisconsinUSA
| | - Rahul Das
- Department of Medicine, Division of Hematology and OncologyUniversity of Wisconsin School of Medicine & Public Health, UW Carbone Cancer Center,MadisonWisconsinUSA
| | - Jaques Galipeau
- Department of Medicine, Division of Hematology and OncologyUniversity of Wisconsin School of Medicine & Public Health, UW Carbone Cancer Center,MadisonWisconsinUSA
| | - Mahua Dey
- Department of NeurosurgeryUniversity of Wisconsin School of Medicine & Public Health, UW Carbone Cancer Center,MadisonWisconsinUSA
| |
Collapse
|