1
|
Xie Y, Wang X, Wang W, Pu N, Liu L. Epithelial-mesenchymal transition orchestrates tumor microenvironment: current perceptions and challenges. J Transl Med 2025; 23:386. [PMID: 40176117 PMCID: PMC11963649 DOI: 10.1186/s12967-025-06422-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Accepted: 03/25/2025] [Indexed: 04/04/2025] Open
Abstract
The epithelial-mesenchymal transition (EMT) is a critical process in cancer progression, facilitating tumor cells to develop invasive traits and augmenting their migratory capabilities. EMT is primed by tumor microenvironment (TME)-derived signals, whereupon cancer cells undergoing EMT in turn remodel the TME, thereby modulating tumor progression and therapeutic response. This review discusses the mechanisms by which EMT coordinates TME dynamics, including secretion of soluble factors, direct cell contact, release of exosomes and enzymes, as well as metabolic reprogramming. Recent evidence also indicates that cells undergoing EMT may differentiate into cancer-associated fibroblasts, thereby establishing themselves as functional constituents of the TME. Elucidating the relationship between EMT and the TME offers novel perspectives for therapeutic strategies to enhance cancer treatment efficacy. Although EMT-directed therapies present significant therapeutic potential, the current lack of effective targeting approaches-attributable to EMT complexity and its microenvironmental context dependency-underscores the necessity for mechanistic investigations and translational clinical validation.
Collapse
Affiliation(s)
- Yuqi Xie
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, People's Republic of China
| | - Xuan Wang
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, People's Republic of China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Wenquan Wang
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, People's Republic of China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Ning Pu
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, People's Republic of China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Liang Liu
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, People's Republic of China.
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
2
|
Ravishankar S, Towlerton AM, Tiamiyu IL, Mooka P, Nankoma J, Kafeero J, Mubiru D, Sekitene S, Aicher LD, Miller CP, Coffey DG, Okoche L, Atwinirembabazi P, Okonye J, White J, Koelle DM, Jing L, Phipps WT, Warren EH. T-cells specific for KSHV and HIV migrate to Kaposi sarcoma tumors and persist over time. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.02.06.579223. [PMID: 38370623 PMCID: PMC10871354 DOI: 10.1101/2024.02.06.579223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/20/2024]
Abstract
Kaposi sarcoma-associated herpesvirus (KSHV) is the etiologic agent of Kaposi sarcoma (KS), which causes significant morbidity and mortality worldwide, particularly in people living with HIV (PLWH) and in sub-Saharan Africa where KSHV seroprevalence is high. Postulating that T-cells specific for KSHV and HIV would be attracted to KS tumors, we performed transcriptional profiling and T-cell receptor (TCR) repertoire analysis of tumor biopsies from 144 Ugandan adults with KS, 106 of whom were also living with HIV. We show that CD8+ T-cells and M2-polarized macrophages are the most common immune cells in KS tumors. The TCR repertoire of T-cells associated with KS tumors is shared across spatially and temporally distinct tumors from the same individual. Clusters of T-cells with predicted shared specificity for uncharacterized antigens, potentially encoded by KSHV or HIV, comprise ~25% of the T-cells in KS tumors. Single-cell RNA-sequencing of blood from a subset of 9 adults captured 4,283 unique αβ TCRs carried in 14,698 putative KSHV- or HIV-specific T-cells, which carried an antigen-experienced effector phenotype. T-cells engineered to express a representative sample of these TCRs showed high-avidity recognition of KSHV- or HIV-encoded antigens. These results suggest that a poly-specific, high-avidity KSHV- and HIV-specific T-cell response, potentially inhibited by M2 macrophages, migrates to and localizes with KS tumors. Further analysis of KSHV- and HIV-specific T-cells in KS tumors will provide insight into the pathogenesis of KS and could guide the development of specific immune therapy based on adoptive transfer or vaccination.
Collapse
Affiliation(s)
- Shashidhar Ravishankar
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, Washington, United States of America
| | - Andrea M.H. Towlerton
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, Washington, United States of America
- Hutchinson Centre Research Institute – Uganda, Kampala, Uganda
| | - Iyabode L. Tiamiyu
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, Washington, United States of America
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington, United States of America
| | - Peter Mooka
- Hutchinson Centre Research Institute – Uganda, Kampala, Uganda
| | - Janet Nankoma
- Hutchinson Centre Research Institute – Uganda, Kampala, Uganda
| | - James Kafeero
- Hutchinson Centre Research Institute – Uganda, Kampala, Uganda
- Uganda Cancer Institute, Kampala, Uganda
| | - Dennis Mubiru
- Hutchinson Centre Research Institute – Uganda, Kampala, Uganda
- Uganda Cancer Institute, Kampala, Uganda
| | - Semei Sekitene
- Hutchinson Centre Research Institute – Uganda, Kampala, Uganda
- Uganda Cancer Institute, Kampala, Uganda
| | - Lauri D. Aicher
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, Washington, United States of America
| | - Chris P. Miller
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, Washington, United States of America
| | - David G. Coffey
- Division of Myeloma, Sylvester Comprehensive Cancer Center, University of Miami, Miami, Florida, United States of America
| | - Lazarus Okoche
- Hutchinson Centre Research Institute – Uganda, Kampala, Uganda
| | | | - Joseph Okonye
- Hutchinson Centre Research Institute – Uganda, Kampala, Uganda
| | - Jessica White
- Department of Medicine, University of Washington, Seattle, Washington, United States
| | - David M. Koelle
- Department of Medicine, University of Washington, Seattle, Washington, United States
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington, United States of America
| | - Lichen Jing
- Department of Medicine, University of Washington, Seattle, Washington, United States
| | - Warren T. Phipps
- Hutchinson Centre Research Institute – Uganda, Kampala, Uganda
- Department of Medicine, University of Washington, Seattle, Washington, United States
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington, United States of America
| | - Edus H. Warren
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, Washington, United States of America
- Hutchinson Centre Research Institute – Uganda, Kampala, Uganda
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington, United States of America
- Department of Medicine, University of Washington, Seattle, Washington, United States
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington, United States of America
| |
Collapse
|
3
|
Heiß A, Krammer S, Kuhnt C, Draßner C, Beck P, Geiger A, Schliep S, Geppert C, Steinkasserer A, Wild AB. Tissue-Resident Regulatory T Cells Expressing CD83 Maintain Local Homeostasis and Restrict Th2 Responses in Asthma. Eur J Immunol 2025; 55:e202451525. [PMID: 39955650 PMCID: PMC11830382 DOI: 10.1002/eji.202451525] [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: 09/19/2024] [Revised: 01/29/2025] [Accepted: 01/30/2025] [Indexed: 02/17/2025]
Abstract
Non-lymphoid tissue Tregs (NLT-Tregs) are critical for tissue homeostasis, inflammation control, and induction of tissue repair. Recent single-cell RNA sequencing data identified the expression of CD83 as part of an NLT-Treg signature, which is an essential molecule for the stability and differentiation of lymphoid Tregs. However, the biological significance of CD83 expression for NLT Tregs has not yet been elucidated. The present study explores for the first time the role of CD83 expression by lung-resident Tregs in the steady state and during asthma to understand its importance in barrier tissues. We evaluated the effect of Treg-specific CD83 deletion (CD83cKO) on the lung-resident T-cell compartment and cytokine profile. CD83-deficient lung Tregs are less differentiated but more activated, resulting in unrestrained T-cell activation. Further, CD83cKO mice were challenged in an asthma model and showed an accelerated disease progression, driven by Th2-biased T-cell responses. CD83cKO Tregs exhibited enhanced responsiveness to IL-4, leading to insufficient control of Th2-differentiation from naïve T cells. These findings underscore the pivotal role of CD83 in the NLT-Treg-mediated modulation of Th2 responses. Overall, our results highlight CD83 as a key player in tissue homeostasis and inflammatory responses, suggesting potential therapeutic implications for inflammatory disorders such as asthma.
Collapse
Affiliation(s)
- Anita Heiß
- Department of Immune ModulationUniversitätsklinikum ErlangenFriedrich‐Alexander‐Universität Erlangen‐Nürnberg (FAU)ErlangenGermany
| | - Susanne Krammer
- Department of Molecular PneumologyUniversitätsklinikum ErlangenFriedrich‐Alexander‐Universität Erlangen‐Nürnberg (FAU)ErlangenGermany
| | - Christine Kuhnt
- Department of Immune ModulationUniversitätsklinikum ErlangenFriedrich‐Alexander‐Universität Erlangen‐Nürnberg (FAU)ErlangenGermany
| | - Christina Draßner
- Department of Immune ModulationUniversitätsklinikum ErlangenFriedrich‐Alexander‐Universität Erlangen‐Nürnberg (FAU)ErlangenGermany
| | - Philipp Beck
- Department of Immune ModulationUniversitätsklinikum ErlangenFriedrich‐Alexander‐Universität Erlangen‐Nürnberg (FAU)ErlangenGermany
| | - Adriana Geiger
- Department of Molecular PneumologyUniversitätsklinikum ErlangenFriedrich‐Alexander‐Universität Erlangen‐Nürnberg (FAU)ErlangenGermany
| | - Stefan Schliep
- Department of DermatologyUniversitätsklinikum ErlangenFriedrich‐Alexander‐Universität Erlangen‐Nürnberg (FAU)ErlangenGermany
| | - Carol‐Immanuel Geppert
- Institute of PathologyUniversitätsklinikum ErlangenFriedrich‐Alexander‐Universität Erlangen‐Nürnberg (FAU)ErlangenGermany
| | - Alexander Steinkasserer
- Department of Immune ModulationUniversitätsklinikum ErlangenFriedrich‐Alexander‐Universität Erlangen‐Nürnberg (FAU)ErlangenGermany
| | - Andreas B. Wild
- Department of Immune ModulationUniversitätsklinikum ErlangenFriedrich‐Alexander‐Universität Erlangen‐Nürnberg (FAU)ErlangenGermany
| |
Collapse
|
4
|
Feng P, Yang Q, Luo L, Guan Z, Fu J, Zhao M, Meng W, Wan S, He J, Li Z, Wang G, Sun G, Dong Z, Yang M. Vps34 sustains Treg cell survival and function via regulating intracellular redox homeostasis. Cell Death Differ 2024; 31:1519-1533. [PMID: 39117783 PMCID: PMC11519664 DOI: 10.1038/s41418-024-01353-y] [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: 08/13/2023] [Revised: 07/28/2024] [Accepted: 07/31/2024] [Indexed: 08/10/2024] Open
Abstract
The survival and suppressive function of regulatory T (Treg) cells rely on various intracellular metabolic and physiological processes. Our study demonstrates that Vps34 plays a critical role in maintaining Treg cell homeostasis and function by regulating cellular metabolic activities. Disruption of Vps34 in Treg cells leads to spontaneous fatal systemic autoimmune disorder and multi-tissue inflammatory damage, accompanied by a reduction in the number of Treg cells, particularly eTreg cells with highly immunosuppressive activity. Mechanistically, the poor survival of Vps34-deficient Treg cells is attributed to impaired endocytosis, intracellular vesicular trafficking and autophagosome formation, which further results in enhanced mitochondrial respiration and excessive ROS production. Removal of excessive ROS can effectively rescue the death of Vps34-deficient Treg cells. Functionally, acute deletion of Vps34 within established Treg cells enhances anti-tumor immunity in a malignant melanoma model by boosting T-cell-mediated anti-tumor activity. Overall, our results underscore the pivotal role played by Vps34 in orchestrating Treg cell homeostasis and function towards establishing immune homeostasis and tolerance.
Collapse
Affiliation(s)
- Peiran Feng
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai People's Hospital (Zhuhai Clinical Medical College of Jinan University), Jinan University, Zhuhai, China
- Guangdong Provincial Key Laboratory of Spine and Spinal Cord Reconstruction, The Fifth Affiliated Hospital of Jinan University (Heyuan Shenhe People's Hospital), Jinan University, Heyuan, China
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, The Biomedical Translational Research Institute, Health Science Center (School of Medicine), Jinan University, Guangzhou, China
| | - Quanli Yang
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai People's Hospital (Zhuhai Clinical Medical College of Jinan University), Jinan University, Zhuhai, China
| | - Liang Luo
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai People's Hospital (Zhuhai Clinical Medical College of Jinan University), Jinan University, Zhuhai, China
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, The Biomedical Translational Research Institute, Health Science Center (School of Medicine), Jinan University, Guangzhou, China
| | - Zerong Guan
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai People's Hospital (Zhuhai Clinical Medical College of Jinan University), Jinan University, Zhuhai, China
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, The Biomedical Translational Research Institute, Health Science Center (School of Medicine), Jinan University, Guangzhou, China
| | - Jiamin Fu
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai People's Hospital (Zhuhai Clinical Medical College of Jinan University), Jinan University, Zhuhai, China
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, The Biomedical Translational Research Institute, Health Science Center (School of Medicine), Jinan University, Guangzhou, China
| | - Mingyue Zhao
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai People's Hospital (Zhuhai Clinical Medical College of Jinan University), Jinan University, Zhuhai, China
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, The Biomedical Translational Research Institute, Health Science Center (School of Medicine), Jinan University, Guangzhou, China
| | - Wanqing Meng
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai People's Hospital (Zhuhai Clinical Medical College of Jinan University), Jinan University, Zhuhai, China
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, The Biomedical Translational Research Institute, Health Science Center (School of Medicine), Jinan University, Guangzhou, China
| | - Shuo Wan
- International Joint Laboratory for Embryonic Development & Prenatal Medicine, Division of Histology and Embryology, School of Medicine, Jinan University, Guangzhou, China
| | - Junming He
- The First Affiliated Hospital of Anhui Medical University and Institute for Clinical Immunology, Anhui Medical University, Hefei, Anhui, China
| | - Zhizhong Li
- Guangdong Provincial Key Laboratory of Spine and Spinal Cord Reconstruction, The Fifth Affiliated Hospital of Jinan University (Heyuan Shenhe People's Hospital), Jinan University, Heyuan, China
| | - Guang Wang
- International Joint Laboratory for Embryonic Development & Prenatal Medicine, Division of Histology and Embryology, School of Medicine, Jinan University, Guangzhou, China
| | - Guodong Sun
- Guangdong Provincial Key Laboratory of Spine and Spinal Cord Reconstruction, The Fifth Affiliated Hospital of Jinan University (Heyuan Shenhe People's Hospital), Jinan University, Heyuan, China
| | - Zhongjun Dong
- The First Affiliated Hospital of Anhui Medical University and Institute for Clinical Immunology, Anhui Medical University, Hefei, Anhui, China.
| | - Meixiang Yang
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai People's Hospital (Zhuhai Clinical Medical College of Jinan University), Jinan University, Zhuhai, China.
- Guangdong Provincial Key Laboratory of Spine and Spinal Cord Reconstruction, The Fifth Affiliated Hospital of Jinan University (Heyuan Shenhe People's Hospital), Jinan University, Heyuan, China.
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, The Biomedical Translational Research Institute, Health Science Center (School of Medicine), Jinan University, Guangzhou, China.
- Key Laboratory of Ministry of Education for Viral Pathogenesis & Infection Prevention and Control (Jinan University). Guangzhou Key Laboratory for Germ-Free Animals and Microbiota Application. Institute of Laboratory Animal Science, School of Medicine, Jinan University, Guangzhou, China.
| |
Collapse
|
5
|
Bessoles S, Chiron A, Sarrabayrouse G, De La Grange P, Abina AM, Hacein-Bey-Abina S. Erythropoietin induces tumour progression and CD39 expression on immune cells in a preclinical model of triple-negative breast cancer. Immunology 2024; 173:360-380. [PMID: 38953295 DOI: 10.1111/imm.13832] [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: 03/21/2024] [Accepted: 06/14/2024] [Indexed: 07/04/2024] Open
Abstract
The adverse effects observed in some cancer patients treated with erythropoiesis-stimulating agents such as erythropoietin (EPO) might be due to the latter's well-known immunosuppressive functions. Here, we used a mouse model of syngeneic triple-negative breast cancer to explore EPO's immunomodulatory role in a tumour setting. Our results showed that EPO treatment promotes tumour growth, exacerbates the 'immune desert', and results in a 'cold tumour'. EPO treatment changed the immune cell distribution in peripheral blood, secondary lymphoid organs, and the tumour microenvironment (TME). Our in-depth analysis showed that EPO mainly impacts CD4 T cells by accelerating their activation in the spleen and thus their subsequent exhaustion in the TME. This process is accompanied by a general elevation of CD39 expression by several immune cells (notably CD4 T cells in the tumour and spleen), which promotes an immunosuppressive TME. Lastly, we identified a highly immunosuppressive CD39+ regulatory T cell population (ICOS+, CTLA4+, Ki67+) as a potential biomarker of the risk of EPO-induced tumour progression. EPO displays pleiotropic immunosuppressive functions and enhances mammary tumour progression in mice.
Collapse
Affiliation(s)
- Stéphanie Bessoles
- Université Paris Cité, CNRS, INSERM, UTCBS, Unité des Technologies Chimiques et Biologiques pour la Santé, Paris, France
| | - Andrada Chiron
- Université Paris Cité, CNRS, INSERM, UTCBS, Unité des Technologies Chimiques et Biologiques pour la Santé, Paris, France
- Clinical Immunology Laboratory, Groupe Hospitalier Universitaire Paris-Sud, Hôpital Kremlin-Bicêtre, Assistance Publique-Hôpitaux de Paris, Le-Kremlin-Bicêtre, France
| | - Guillaume Sarrabayrouse
- Université Paris Cité, CNRS, INSERM, UTCBS, Unité des Technologies Chimiques et Biologiques pour la Santé, Paris, France
| | | | - Amine M Abina
- Université Paris Cité, CNRS, INSERM, UTCBS, Unité des Technologies Chimiques et Biologiques pour la Santé, Paris, France
| | - Salima Hacein-Bey-Abina
- Université Paris Cité, CNRS, INSERM, UTCBS, Unité des Technologies Chimiques et Biologiques pour la Santé, Paris, France
- Clinical Immunology Laboratory, Groupe Hospitalier Universitaire Paris-Sud, Hôpital Kremlin-Bicêtre, Assistance Publique-Hôpitaux de Paris, Le-Kremlin-Bicêtre, France
| |
Collapse
|
6
|
Sang J, Liu P, Wang M, Xu F, Ma J, Wei Z, Ye X. Dynamic Changes in the Immune Microenvironment in Tumor-Draining Lymph Nodes of a Lewis Lung Cancer Mouse Model After Microwave Ablation. J Inflamm Res 2024; 17:4175-4186. [PMID: 38979433 PMCID: PMC11228081 DOI: 10.2147/jir.s462650] [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: 02/02/2024] [Accepted: 06/12/2024] [Indexed: 07/10/2024] Open
Abstract
PURPOSE Microwave ablation (MWA) is a minimally invasive technique for treating lung cancer. It can induce immune response; however, its effect on the immune microenvironment in tumor-draining lymph nodes (TdLN) is not well understood. This study aims to identify changes in the immune microenvironment in TdLN following MWA in a Lewis lung cancer (LLC) mouse model. METHODS LLC mouse model was established and followed by MWA. TdLN were collected at various time points, including pre-MWA and days 1, 2, 4, and 8 post-MWA. Flow cytometry was used to determine the frequencies of CD4+ T cells, CD8+ T cells, regulatory T (Treg) cells, natural killer (NK) cells, dendritic cells (DCs) and other immune cells in the TdLN. Certain cytokines were also detected. RESULTS Compared with pre-MWA, the frequency of CD4+ T cells significantly increased from day 1 to day 8 post-MWA. The frequency of CD8+ T cells decreased significantly on days 2 and 4, but no significant changes occurred on days 1 and 8. Significant decreases in the frequencies of Treg cells and Klrg1+ Treg cells were observed from day 1 to day 4. On days 4 and 8, there was a significant increase in the frequency of NK cells. The frequency of resident cDC2 significantly increased on day 4, whereas CD11b+ migratory cDCs increased on day 1. Additionally, on day 4, a notable rise was observed in the frequency of NK cells secreting IFN-γ, while on day 8, there was a significant increase in the frequency of CD8+ T cells secreting both IFN-γ and TNF-α. CONCLUSION MWA of lung cancer can alter the immune microenvironment in the TdLN, triggering immune responses. These changes are particularly evident and intricate within the initial 4 days post-MWA. Treatment combined with MWA within a certain period may significantly enhance anti-tumor immunity.
Collapse
Affiliation(s)
- Jing Sang
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Lung Cancer Institute, Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Jinan, People’s Republic of China
- Department of Pathology, The Affiliated Taian City Central Hospital of Qingdao University, Taian, People’s Republic of China
| | - Peng Liu
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Lung Cancer Institute, Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Jinan, People’s Republic of China
- Yuncheng Central Hospital Affiliated to Shanxi Medical University, Yuncheng, People’s Republic of China
| | - Meixiang Wang
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Lung Cancer Institute, Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Jinan, People’s Republic of China
| | - Fengkuo Xu
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Lung Cancer Institute, Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Jinan, People’s Republic of China
| | - Ji Ma
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Lung Cancer Institute, Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Jinan, People’s Republic of China
| | - Zhigang Wei
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Lung Cancer Institute, Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Jinan, People’s Republic of China
| | - Xin Ye
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Lung Cancer Institute, Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Jinan, People’s Republic of China
| |
Collapse
|
7
|
Zhang Y, Chen S, Tang X, Peng Y, Jiang T, Zhang X, Li J, Liu Y, Yang Z. The role of KLRG1: a novel biomarker and new therapeutic target. Cell Commun Signal 2024; 22:337. [PMID: 38898461 PMCID: PMC11186184 DOI: 10.1186/s12964-024-01714-7] [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: 04/07/2024] [Accepted: 06/13/2024] [Indexed: 06/21/2024] Open
Abstract
Killer cell lectin-like receptor G1 (KLRG1) is an immune checkpoint receptor expressed predominantly in NK and T-cell subsets that downregulates the activation and proliferation of immune cells and participates in cell-mediated immune responses. Accumulating evidence has demonstrated the importance of KLRG1 as a noteworthy disease marker and therapeutic target that can influence disease onset, progression, and prognosis. Blocking KLRG1 has been shown to effectively mitigate the effects of downregulation in various mouse tumor models, including solid tumors and hematologic malignancies. However, KLRG1 inhibitors have not yet been approved for human use, and the understanding of KLRG1 expression and its mechanism of action in various diseases remains incomplete. In this review, we explore alterations in the distribution, structure, and signaling pathways of KLRG1 in immune cells and summarize its expression patterns and roles in the development and progression of autoimmune diseases, infectious diseases, and cancers. Additionally, we discuss the potential applications of KLRG1 as a tool for tumor immunotherapy.
Collapse
Affiliation(s)
- Yakun Zhang
- School of Medicine, Chongqing University, Chongqing, 400030, China
- Department of Hematology-Oncology, Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing, 400030, China
| | - Shuang Chen
- Department of Hematology-Oncology, Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing, 400030, China
| | - Xinyi Tang
- School of Medicine, Chongqing University, Chongqing, 400030, China
- Department of Hematology-Oncology, Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing, 400030, China
| | - Yu Peng
- Department of Hematology-Oncology, Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing, 400030, China
| | - Tingting Jiang
- Department of Hematology-Oncology, Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing, 400030, China
| | - Xiaomei Zhang
- Department of Hematology-Oncology, Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing, 400030, China
| | - Jun Li
- Department of Hematology-Oncology, Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing, 400030, China
| | - Yao Liu
- Department of Hematology-Oncology, Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing, 400030, China.
| | - Zailin Yang
- Department of Hematology-Oncology, Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing, 400030, China.
| |
Collapse
|
8
|
Morel D, Robert C, Paragios N, Grégoire V, Deutsch E. Translational Frontiers and Clinical Opportunities of Immunologically Fitted Radiotherapy. Clin Cancer Res 2024; 30:2317-2332. [PMID: 38477824 PMCID: PMC11145173 DOI: 10.1158/1078-0432.ccr-23-3632] [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: 11/21/2023] [Revised: 01/09/2024] [Accepted: 02/13/2024] [Indexed: 03/14/2024]
Abstract
Ionizing radiation can have a wide range of impacts on tumor-immune interactions, which are being studied with the greatest interest and at an accelerating pace by the medical community. Despite its undeniable immunostimulatory potential, it clearly appears that radiotherapy as it is prescribed and delivered nowadays often alters the host's immunity toward a suboptimal state. This may impair the full recovery of a sustained and efficient antitumor immunosurveillance posttreatment. An emerging concept is arising from this awareness and consists of reconsidering the way of designing radiation treatment planning, notably by taking into account the individualized risks of deleterious radio-induced immune alteration that can be deciphered from the planned beam trajectory through lymphocyte-rich organs. In this review, we critically appraise key aspects to consider while planning immunologically fitted radiotherapy, including the challenges linked to the identification of new dose constraints to immune-rich structures. We also discuss how pharmacologic immunomodulation could be advantageously used in combination with radiotherapy to compensate for the radio-induced loss, for example, with (i) agonists of interleukin (IL)2, IL4, IL7, IL9, IL15, or IL21, similarly to G-CSF being used for the prophylaxis of severe chemo-induced neutropenia, or with (ii) myeloid-derived suppressive cell blockers.
Collapse
Affiliation(s)
- Daphné Morel
- Department of Radiation Oncology, Gustave Roussy, Villejuif, France
- INSERM U1030, Molecular Radiotherapy, Villejuif, France
| | - Charlotte Robert
- Department of Radiation Oncology, Gustave Roussy, Villejuif, France
- INSERM U1030, Molecular Radiotherapy, Villejuif, France
- Paris-Saclay University, School of Medicine, Le Kremlin Bicêtre, France
| | - Nikos Paragios
- Therapanacea, Paris, France
- CentraleSupélec, Gif-sur-Yvette, France
| | - Vincent Grégoire
- Department of Radiation Oncology, Centre Léon Bérard, Lyon, France
| | - Eric Deutsch
- Department of Radiation Oncology, Gustave Roussy, Villejuif, France
- INSERM U1030, Molecular Radiotherapy, Villejuif, France
- Paris-Saclay University, School of Medicine, Le Kremlin Bicêtre, France
| |
Collapse
|
9
|
Varma G, Diefenbach CS. KLRG1, Another Opportunity for a Breakthrough in MTCL. Clin Cancer Res 2024; 30:2300-2302. [PMID: 38568191 DOI: 10.1158/1078-0432.ccr-24-0374] [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: 02/21/2024] [Revised: 03/12/2024] [Accepted: 03/21/2024] [Indexed: 06/04/2024]
Abstract
Outcomes in mature T-cell lymphomas remain poor, with previous attempts at developing mAbs compromised by limited efficacy and significant immunocompromise. Anti-killer cell lectin-like receptor G1 mAbs may have greater selectivity and specificity for malignant T cells and avoid the toxicity concerns with previous agents. See related article by Assatova et al., p. 2514.
Collapse
Affiliation(s)
- Gaurav Varma
- Division of Hematology and Medical Oncology, Department of Medicine, NYU Grossman School of Medicine, New York, New York
- Laura and Isaac Perlmutter Cancer Center, NYU Langone Health, New York, New York
| | - Catherine S Diefenbach
- Division of Hematology and Medical Oncology, Department of Medicine, NYU Grossman School of Medicine, New York, New York
- Laura and Isaac Perlmutter Cancer Center, NYU Langone Health, New York, New York
| |
Collapse
|
10
|
Li T, Han B, Wang L, Sun L, Cai Y, Yu M, Xiao W, Yang H. Activation of mucosal insulin receptor exacerbates intestinal inflammation by promoting tissue resident memory T cells differentiation through EZH2. J Transl Med 2024; 22:78. [PMID: 38243324 PMCID: PMC10797971 DOI: 10.1186/s12967-023-04789-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Accepted: 12/09/2023] [Indexed: 01/21/2024] Open
Abstract
BACKGROUND Inflammatory Bowel Diseases (IBD), an autoimmune disease characterised by abnormal intestinal immunity, are related to vital morbidity around the world. However, therapeutic agents for IBD have not achieved desired benefit. Exploring new therapeutic targets for IBD, especially based on its abnormally intestinal immunity, could alleviate the flare-up and worsening of IBD. Tissue resident memory T cells (TRM) are core of multiple autoimmune diseases, including IBD. However, the mechanism of TRM differentiation remains to be investigated. METHODS The alterations in mRNA and lncRNA profile of intestinal intraepithelial lymphocytes (IELs), the largest component of intestinal TRM, were analyzed in DSS-induced chronic colitis. Based on it, we examined the function of rectal insulin instillation in a dextran sodium sulfate (DSS) induced chronic colitis. Furthermore, we investigated the downstream-target of the insulin pathway-EZH2 and the crucial role of EZH2 in intestinal tissue resident memory T cell differentiation by utilizing EZH2fl/flCD4cre mice. RESULTS Insulin receptor (INSR) expression was found to be significantly reduced. Activation of mucosal insulin pathway by rectal insulin instillation exacerbated colitis by disrupting IELs subgroups and up-regulating TNF-ɑ and IL-17 expression. Rectal insulin instillation promoted EZH2 expression and EZH2 inhibition alleviated chronic colitis. EZH2fl/flCD4cre mice restored the normal IEL subgroups and suppressed TNF-ɑ and IL-17 expression, exhibiting alleviated colitis. IELs from EZH2fl/flCD4cre mice exhibit significant changes in TRM related phenotype. CD4+TRM was significantly increased in chronic colitis and decreased in EZH2fl/flCD4cre mice. CONCLUSION Insulin receptor of intestinal mucosal T-cells could promote intestinal TRM differentiation via EZH2. Our discoveries suggest that therapies targeting colonic INSR and EZH2 could be potential treatment for IBD based on its regulatory effects on TRM. Insulin receptor inhibitors rather than insulin should be applied during colitis-active phase. In addition, EZH2 shows to be a downstream signal of the insulin pathway and EZH2 inhibitor could alleviating intestinal inflammation. However, the critical role of EZH2 in TRM differentiation restricts the anti-tumor effects of EZH2 inhibitor in vivo.
Collapse
Affiliation(s)
- Teming Li
- Department of General Surgery, Xinqiao Hospital, Army Medical University, Chongqing, 400037, China
- Department of General Surgery, Army 953 Hospital, Shigatse Branch of Xinqiao Hospital, Army Medical University, Shigatse, 857000, China
| | - Ben Han
- Department of General Surgery, Xinqiao Hospital, Army Medical University, Chongqing, 400037, China
| | - Liucan Wang
- Department of General Surgery, Xinqiao Hospital, Army Medical University, Chongqing, 400037, China
| | - Lihua Sun
- Department of General Surgery, Xinqiao Hospital, Army Medical University, Chongqing, 400037, China
| | - Yujiao Cai
- Department of General Surgery, Xinqiao Hospital, Army Medical University, Chongqing, 400037, China
| | - Min Yu
- Department of General Surgery, Chongqing General Hospital, Chongqing, 401147, China.
| | - Weidong Xiao
- Department of General Surgery, Xinqiao Hospital, Army Medical University, Chongqing, 400037, China.
| | - Hua Yang
- Department of General Surgery, Xinqiao Hospital, Army Medical University, Chongqing, 400037, China.
- Department of General Surgery, Chongqing General Hospital, Chongqing, 401147, China.
| |
Collapse
|
11
|
Kos FJ, Frankel P, Cristea M, Eng M, Tinsley R, Dempsey S, Ruel N, Stewart D, Dellinger TH, Diamond DJ. Immunologic Signatures of Peripheral Blood T Cells Reveal the Outcome of p53MVA Vaccine and Pembrolizumab Treatment in Patients with Advanced Ovarian Cancer. CANCER RESEARCH COMMUNICATIONS 2023; 3:2585-2595. [PMID: 38032111 PMCID: PMC10732002 DOI: 10.1158/2767-9764.crc-23-0394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 11/28/2023] [Accepted: 11/28/2023] [Indexed: 12/01/2023]
Abstract
PURPOSE Our previous studies indicated that p53-reactive T cells were associated with clinical benefit in patients with advanced ovarian cancer who were treated with p53-expressing modified vaccinia Ankara (p53MVA) vaccine and gemcitabine chemotherapy. To replace chemotherapy with an approach that will enhance vaccine efficacy and antitumor immunity, we treated patients with p53MVA in combination with PD-1 checkpoint blocker, pembrolizumab. We also attempted to further characterize the activation status of T cells prior to vaccination and during treatment. EXPERIMENTAL DESIGN Patients received up to three triweekly vaccinations concurrent with pembrolizumab, followed by pembrolizumab monotherapy at 3-week intervals. Correlative studies analyzed peripheral blood T-cell phenotypes and profiles of immune function gene expression. RESULTS We observed 6/28 (21%) patients with a clinical benefit to therapy, including 3 partial responses (PR) and 3 patients with stable disease (SD) for 6+ months. The median progression-free survival was 1.8 months (95% confidence interval: 1.7-3.8) and median overall survival was 15.1 months (9.4-30.4). Two patients remain progression-free at 28 and 33 months. Of the 18 patients evaluable in correlative studies, 6 were immunologic responders of whom 5 had clinical benefit (3 PR, 2 SD). Immunologic non-responders expressed in pretreatment peripheral blood mononuclear cell samples high levels of mRNA for multiple molecules associated with terminally differentiated T cells. CONCLUSIONS p53MVA/pembrolizumab immunotherapy showed promising antitumor activity in patients who demonstrated functionally competent peripheral blood T cells. Detection of markers of terminally differentiated T cells before treatment may identify patients unlikely to respond to p53MVA/pembrolizumab. SIGNIFICANCE The activity of a combination immunotherapy of p53 vaccine and PD-1 checkpoint blockade in patients with platinum-resistant ovarian cancer was evaluated in a phase II trial. Clinical benefit was correlated with the responsive immune status of patients before and during the treatment, defining potential predictive markers for immune therapy.
Collapse
Affiliation(s)
- Ferdynand J. Kos
- Department of Hematology and Hematopoietic Cell Transplantation, Beckman Research Institute, City of Hope, Duarte, California
| | - Paul Frankel
- Department of Computational and Quantitative Medicine, Beckman Research Institute, Duarte, California
| | - Mihaela Cristea
- Department of Medical Oncology, City of Hope National Medical Center, Duarte, California
| | - Melissa Eng
- Clinical Trials Office, City of Hope National Medical Center, Duarte, California
| | - Raechelle Tinsley
- Clinical Trials Office, City of Hope National Medical Center, Duarte, California
| | - Shannon Dempsey
- Department of Hematology and Hematopoietic Cell Transplantation, Beckman Research Institute, City of Hope, Duarte, California
| | - Nora Ruel
- Department of Computational and Quantitative Medicine, Beckman Research Institute, Duarte, California
| | - Daphne Stewart
- Department of Medical Oncology, City of Hope National Medical Center, Duarte, California
| | - Thanh H. Dellinger
- Department of Surgery, City of Hope National Medical Center, Duarte, California
| | - Don J. Diamond
- Department of Hematology and Hematopoietic Cell Transplantation, Beckman Research Institute, City of Hope, Duarte, California
| |
Collapse
|
12
|
Astorga-Gamaza A, Perea D, Sanchez-Gaona N, Calvet-Mirabent M, Gallego-Cortés A, Grau-Expósito J, Sanchez-Cerrillo I, Rey J, Castellví J, Curran A, Burgos J, Navarro J, Suanzes P, Falcó V, Genescà M, Martín-Gayo E, Buzon MJ. KLRG1 expression on natural killer cells is associated with HIV persistence, and its targeting promotes the reduction of the viral reservoir. Cell Rep Med 2023; 4:101202. [PMID: 37741278 PMCID: PMC10591043 DOI: 10.1016/j.xcrm.2023.101202] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 05/25/2023] [Accepted: 08/29/2023] [Indexed: 09/25/2023]
Abstract
Human immunodeficiency virus (HIV) infection induces immunological dysfunction, which limits the elimination of HIV-infected cells during treated infection. Identifying and targeting dysfunctional immune cells might help accelerate the purging of the persistent viral reservoir. Here, we show that chronic HIV infection increases natural killer (NK) cell populations expressing the negative immune regulator KLRG1, both in peripheral blood and lymph nodes. Antiretroviral treatment (ART) does not reestablish these functionally impaired NK populations, and the expression of KLRG1 correlates with active HIV transcription. Targeting KLRG1 with specific antibodies significantly restores the capacity of NK cells to kill HIV-infected cells, reactivates latent HIV present in CD4+ T cells co-expressing KLRG1, and reduces the intact HIV genomes in samples from ART-treated individuals. Our data support the potential use of immunotherapy against the KLRG1 receptor to impact the viral reservoir during HIV persistence.
Collapse
Affiliation(s)
- Antonio Astorga-Gamaza
- Infectious Diseases Department, Hospital Universitari Vall d'Hebron, Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, 08035 Barcelona, Spain
| | - David Perea
- Infectious Diseases Department, Hospital Universitari Vall d'Hebron, Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, 08035 Barcelona, Spain
| | - Nerea Sanchez-Gaona
- Infectious Diseases Department, Hospital Universitari Vall d'Hebron, Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, 08035 Barcelona, Spain
| | - Marta Calvet-Mirabent
- Universidad Autónoma de Madrid, 28049 Madrid, Spain; Immunology Unit from Hospital Universitario de La Princesa and Instituto de Investigación Sanitaria Princesa, 28006 Madrid, Spain
| | - Ana Gallego-Cortés
- Infectious Diseases Department, Hospital Universitari Vall d'Hebron, Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, 08035 Barcelona, Spain
| | - Judith Grau-Expósito
- Infectious Diseases Department, Hospital Universitari Vall d'Hebron, Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, 08035 Barcelona, Spain
| | - Ildefonso Sanchez-Cerrillo
- Universidad Autónoma de Madrid, 28049 Madrid, Spain; Immunology Unit from Hospital Universitario de La Princesa and Instituto de Investigación Sanitaria Princesa, 28006 Madrid, Spain
| | - Joan Rey
- Infectious Diseases Department, Hospital Universitari Vall d'Hebron, Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, 08035 Barcelona, Spain
| | - Josep Castellví
- Department of Pathology, Hospital Vall d'Hebron, Universitat Autònoma de Barcelona, 08035 Barcelona, Spain
| | - Adrian Curran
- Infectious Diseases Department, Hospital Universitari Vall d'Hebron, Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, 08035 Barcelona, Spain
| | - Joaquin Burgos
- Infectious Diseases Department, Hospital Universitari Vall d'Hebron, Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, 08035 Barcelona, Spain
| | - Jordi Navarro
- Infectious Diseases Department, Hospital Universitari Vall d'Hebron, Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, 08035 Barcelona, Spain
| | - Paula Suanzes
- Infectious Diseases Department, Hospital Universitari Vall d'Hebron, Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, 08035 Barcelona, Spain
| | - Vicenç Falcó
- Infectious Diseases Department, Hospital Universitari Vall d'Hebron, Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, 08035 Barcelona, Spain
| | - Meritxell Genescà
- Infectious Diseases Department, Hospital Universitari Vall d'Hebron, Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, 08035 Barcelona, Spain
| | - Enrique Martín-Gayo
- Universidad Autónoma de Madrid, 28049 Madrid, Spain; Immunology Unit from Hospital Universitario de La Princesa and Instituto de Investigación Sanitaria Princesa, 28006 Madrid, Spain; Infectious Diseases CIBER (CIBERINFECC), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Maria J Buzon
- Infectious Diseases Department, Hospital Universitari Vall d'Hebron, Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, 08035 Barcelona, Spain.
| |
Collapse
|
13
|
Jia H, Wan H, Zhang D. Innate lymphoid cells: a new key player in atopic dermatitis. Front Immunol 2023; 14:1277120. [PMID: 37908364 PMCID: PMC10613734 DOI: 10.3389/fimmu.2023.1277120] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Accepted: 09/25/2023] [Indexed: 11/02/2023] Open
Abstract
Atopic dermatitis (AD) is a common allergic inflammatory skin condition mainly caused by gene variants, immune disorders, and environmental risk factors. The T helper (Th) 2 immune response mediated by interleukin (IL)-4/13 is generally believed to be central in the pathogenesis of AD. It has been shown that innate lymphoid cells (ILCs) play a major effector cell role in the immune response in tissue homeostasis and inflammation and fascinating details about the interaction between innate and adaptive immunity. Changes in ILCs may contribute to the onset and progression of AD, and ILC2s especially have gained much attention. However, the role of ILCs in AD still needs to be further elucidated. This review summarizes the role of ILCs in skin homeostasis and highlights the signaling pathways in which ILCs may be involved in AD, thus providing valuable insights into the behavior of ILCs in skin homeostasis and inflammation, as well as new approaches to treating AD.
Collapse
Affiliation(s)
- Haiping Jia
- Institute of Basic Medicine and Forensic Medicine, North Sichuan Medical College, Nanchong, China
| | - Huiying Wan
- Department of Dermatology, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Dingding Zhang
- Sichuan Provincial Key Laboratory for Genetic Disease, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
14
|
Wang N, Scott TA, Kupz A, Shreenivas MM, Peres NG, Hocking DM, Yang C, Jebeli L, Beattie L, Groom JR, Pierce TP, Wakim LM, Bedoui S, Strugnell RA. Vaccine-induced inflammation and inflammatory monocytes promote CD4+ T cell-dependent immunity against murine salmonellosis. PLoS Pathog 2023; 19:e1011666. [PMID: 37733817 PMCID: PMC10547166 DOI: 10.1371/journal.ppat.1011666] [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: 12/21/2022] [Revised: 10/03/2023] [Accepted: 09/07/2023] [Indexed: 09/23/2023] Open
Abstract
Prior infection can generate protective immunity against subsequent infection, although the efficacy of such immunity can vary considerably. Live-attenuated vaccines (LAVs) are one of the most effective methods for mimicking this natural process, and analysis of their efficacy has proven instrumental in the identification of protective immune mechanisms. Here, we address the question of what makes a LAV efficacious by characterising immune responses to a LAV, termed TAS2010, which is highly protective (80-90%) against lethal murine salmonellosis, in comparison with a moderately protective (40-50%) LAV, BRD509. Mice vaccinated with TAS2010 developed immunity systemically and were protected against gut-associated virulent infection in a CD4+ T cell-dependent manner. TAS2010-vaccinated mice showed increased activation of Th1 responses compared with their BRD509-vaccinated counterparts, leading to increased Th1 memory populations in both lymphoid and non-lymphoid organs. The optimal development of Th1-driven immunity was closely correlated with the activation of CD11b+Ly6GnegLy6Chi inflammatory monocytes (IMs), the activation of which can be modulated proportionally by bacterial load in vivo. Upon vaccination with the LAV, IMs expressed T cell chemoattractant CXCL9 that attracted CD4+ T cells to the foci of infection, where IMs also served as a potent source of antigen presentation and Th1-promoting cytokine IL-12. The expression of MHC-II in IMs was rapidly upregulated following vaccination and then maintained at an elevated level in immune mice, suggesting IMs may have a role in sustained antigen stimulation. Our findings present a longitudinal analysis of CD4+ T cell development post-vaccination with an intracellular bacterial LAV, and highlight the benefit of inflammation in the development of Th1 immunity. Future studies focusing on the induction of IMs may reveal key strategies for improving vaccine-induced T cell immunity.
Collapse
Affiliation(s)
- Nancy Wang
- Department of Microbiology and Immunology, The University of Melbourne, at Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Timothy A. Scott
- Department of Microbiology and Immunology, The University of Melbourne, at Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Andreas Kupz
- Department of Microbiology and Immunology, The University of Melbourne, at Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Meghanashree M. Shreenivas
- Department of Microbiology and Immunology, The University of Melbourne, at Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Newton G. Peres
- Department of Microbiology and Immunology, The University of Melbourne, at Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Dianna M. Hocking
- Department of Microbiology and Immunology, The University of Melbourne, at Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Chenying Yang
- Department of Microbiology and Immunology, The University of Melbourne, at Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Leila Jebeli
- Department of Microbiology and Immunology, The University of Melbourne, at Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Lynette Beattie
- Department of Microbiology and Immunology, The University of Melbourne, at Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Joanna R. Groom
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia
| | - Thomas P. Pierce
- Ludwig Institute for Cancer Research, Melbourne-Parkville Branch, Parkville, Victoria, Australia
| | - Linda M. Wakim
- Department of Microbiology and Immunology, The University of Melbourne, at Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Sammy Bedoui
- Department of Microbiology and Immunology, The University of Melbourne, at Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Richard A. Strugnell
- Department of Microbiology and Immunology, The University of Melbourne, at Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| |
Collapse
|
15
|
Ager CR, Zhang M, Chaimowitz M, Bansal S, Tagore S, Obradovic A, Jugler C, Rogava M, Melms JC, McCann P, Spina C, Drake CG, Dallos MC, Izar B. KLRG1 marks tumor-infiltrating CD4 T cell subsets associated with tumor progression and immunotherapy response. J Immunother Cancer 2023; 11:e006782. [PMID: 37657842 PMCID: PMC10476134 DOI: 10.1136/jitc-2023-006782] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/08/2023] [Indexed: 09/03/2023] Open
Abstract
Current methods for biomarker discovery and target identification in immuno-oncology rely on static snapshots of tumor immunity. To thoroughly characterize the temporal nature of antitumor immune responses, we developed a 34-parameter spectral flow cytometry panel and performed high-throughput analyses in critical contexts. We leveraged two distinct preclinical models that recapitulate cancer immunoediting (NPK-C1) and immune checkpoint blockade (ICB) response (MC38), respectively, and profiled multiple relevant tissues at and around key inflection points of immune surveillance and escape and/or ICB response. Machine learning-driven data analysis revealed a pattern of KLRG1 expression that uniquely identified intratumoral effector CD4 T cell populations that constitutively associate with tumor burden across tumor models, and are lost in tumors undergoing regression in response to ICB. Similarly, a Helios-KLRG1+ subset of tumor-infiltrating regulatory T cells was associated with tumor progression from immune equilibrium to escape and was also lost in tumors responding to ICB. Validation studies confirmed KLRG1 signatures in human tumor-infiltrating CD4 T cells associate with disease progression in renal cancer. These findings nominate KLRG1+ CD4 T cell populations as subsets for further investigation in cancer immunity and demonstrate the utility of longitudinal spectral flow profiling as an engine of dynamic biomarker discovery.
Collapse
Affiliation(s)
- Casey R Ager
- Department of Medicine, Division of Hematology and Oncology, Columbia University Irving Medical Center, New York, New York, USA
- Department of Immunology, Mayo Clinic Arizona, Scottsdale, Arizona, USA
- Columbia Center for Translational Immunology, Columbia University Irving Medical Center, New York, New York, USA
- Department of Urology, Mayo Clinic Arizona, Scottsdale, Arizona, USA
| | - Mingxuan Zhang
- Department of Medicine, Division of Hematology and Oncology, Columbia University Irving Medical Center, New York, New York, USA
- Department of Molecular Pathology and Therapeutics, Columbia University Irving Medical Center, New York, New York, USA
| | - Matthew Chaimowitz
- Columbia Center for Translational Immunology, Columbia University Irving Medical Center, New York, New York, USA
- Department of Radiation Oncology, Columbia University Irving Medical Center, New York, New York, USA
| | - Shruti Bansal
- Columbia Center for Translational Immunology, Columbia University Irving Medical Center, New York, New York, USA
- Department of Radiation Oncology, Columbia University Irving Medical Center, New York, New York, USA
| | - Somnath Tagore
- Department of Medicine, Division of Hematology and Oncology, Columbia University Irving Medical Center, New York, New York, USA
- Department of Systems Biology, Columbia University Irving Medical Center, New York, New York, USA
| | - Aleksandar Obradovic
- Department of Medicine, Division of Hematology and Oncology, Columbia University Irving Medical Center, New York, New York, USA
- Department of Systems Biology, Columbia University Irving Medical Center, New York, New York, USA
| | - Collin Jugler
- Department of Immunology, Mayo Clinic Arizona, Scottsdale, Arizona, USA
| | - Meri Rogava
- Department of Medicine, Division of Hematology and Oncology, Columbia University Irving Medical Center, New York, New York, USA
- Columbia Center for Translational Immunology, Columbia University Irving Medical Center, New York, New York, USA
| | - Johannes C Melms
- Department of Medicine, Division of Hematology and Oncology, Columbia University Irving Medical Center, New York, New York, USA
- Columbia Center for Translational Immunology, Columbia University Irving Medical Center, New York, New York, USA
| | - Patrick McCann
- Columbia Center for Translational Immunology, Columbia University Irving Medical Center, New York, New York, USA
- Department of Radiation Oncology, Columbia University Irving Medical Center, New York, New York, USA
| | - Catherine Spina
- Columbia Center for Translational Immunology, Columbia University Irving Medical Center, New York, New York, USA
- Department of Radiation Oncology, Columbia University Irving Medical Center, New York, New York, USA
| | - Charles G Drake
- Department of Medicine, Division of Hematology and Oncology, Columbia University Irving Medical Center, New York, New York, USA
- Columbia Center for Translational Immunology, Columbia University Irving Medical Center, New York, New York, USA
- Department of Urology, Mayo Clinic Arizona, Scottsdale, Arizona, USA
- Janssen Research and Development, Janssen Pharmaceuticals, Spring House, Pennsylvania, USA
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, New York, USA
| | - Matthew C Dallos
- Department of Medicine, Division of Hematology and Oncology, Columbia University Irving Medical Center, New York, New York, USA
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, New York, USA
- Department of Urology, Columbia University Irving Medical Center, New York, New York, USA
| | - Benjamin Izar
- Department of Medicine, Division of Hematology and Oncology, Columbia University Irving Medical Center, New York, New York, USA
- Columbia Center for Translational Immunology, Columbia University Irving Medical Center, New York, New York, USA
- Department of Systems Biology, Columbia University Irving Medical Center, New York, New York, USA
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, New York, USA
- Columbia University Vagelos College of Physicians and Surgeons, New York, New York, USA
| |
Collapse
|
16
|
Evangelou K, Belogiannis K, Papaspyropoulos A, Petty R, Gorgoulis VG. Escape from senescence: molecular basis and therapeutic ramifications. J Pathol 2023; 260:649-665. [PMID: 37550877 DOI: 10.1002/path.6164] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 06/16/2023] [Accepted: 06/19/2023] [Indexed: 08/09/2023]
Abstract
Cellular senescence constitutes a stress response mechanism in reaction to a plethora of stimuli. Senescent cells exhibit cell-cycle arrest and altered function. While cell-cycle withdrawal has been perceived as permanent, recent evidence in cancer research introduced the so-called escape-from-senescence concept. In particular, under certain conditions, senescent cells may resume proliferation, acquiring highly aggressive features. As such, they have been associated with tumour relapse, rendering senescence less effective in inhibiting cancer progression. Thus, conventional cancer treatments, incapable of eliminating senescence, may benefit if revisited to include senolytic agents. To this end, it is anticipated that the assessment of the senescence burden in everyday clinical material by pathologists will play a crucial role in the near future, laying the foundation for more personalised approaches. Here, we provide an overview of the investigations that introduced the escape-from-senescence phenomenon, the identified mechanisms, as well as the major implications for pathology and therapy. © 2023 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Konstantinos Evangelou
- Molecular Carcinogenesis Group, Department of Histology and Embryology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Konstantinos Belogiannis
- Molecular Carcinogenesis Group, Department of Histology and Embryology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Angelos Papaspyropoulos
- Molecular Carcinogenesis Group, Department of Histology and Embryology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
- Biomedical Research Foundation, Academy of Athens, Athens, Greece
| | - Russell Petty
- Ninewells Hospital and Medical School, University of Dundee, Dundee, UK
| | - Vassilis G Gorgoulis
- Molecular Carcinogenesis Group, Department of Histology and Embryology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
- Biomedical Research Foundation, Academy of Athens, Athens, Greece
- Ninewells Hospital and Medical School, University of Dundee, Dundee, UK
- Faculty Institute for Cancer Sciences, Manchester Academic Health Sciences Centre, University of Manchester, Manchester, UK
- Center for New Biotechnologies and Precision Medicine, Medical School, National and Kapodistrian University of Athens, Athens, Greece
- Faculty of Health and Medical Sciences, University of Surrey, Guildford, UK
| |
Collapse
|
17
|
Stravokefalou V, Stellas D, Karaliota S, Nagy BA, Valentin A, Bergamaschi C, Dimas K, Pavlakis GN. Heterodimeric IL-15 (hetIL-15) reduces circulating tumor cells and metastasis formation improving chemotherapy and surgery in 4T1 mouse model of TNBC. Front Immunol 2023; 13:1014802. [PMID: 36713398 PMCID: PMC9880212 DOI: 10.3389/fimmu.2022.1014802] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 12/23/2022] [Indexed: 01/15/2023] Open
Abstract
Immunotherapy has emerged as a viable approach in cancer therapy, with cytokines being of great interest. Interleukin IL-15 (IL-15), a cytokine that supports cytotoxic immune cells, has been successfully tested as an anti-cancer and anti-metastatic agent, but combinations with conventional chemotherapy and surgery protocols have not been extensively studied. We have produced heterodimeric IL-15 (hetIL-15), which has shown anti-tumor efficacy in several murine cancer models and is being evaluated in clinical trials for metastatic cancers. In this study, we examined the therapeutic effects of hetIL-15 in combination with chemotherapy and surgery in the 4T1 mouse model of metastatic triple negative breast cancer (TNBC). hetIL-15 monotherapy exhibited potent anti-metastatic effects by diminishing the number of circulating tumor cells (CTCs) and by controlling tumor cells colonization of the lungs. hetIL-15 treatment in combination with doxorubicin resulted in enhanced anti-metastatic activity and extended animal survival. Systemic immune phenotype analysis showed that the chemoimmunotherapeutic regimen shifted the tumor-induced imbalance of polymorphonuclear myeloid-derived suppressor cells (PMN-MDSCs) in favor of cytotoxic effector cells, by simultaneously decreasing PMN-MDSCs and increasing the frequency and activation of effector (CD8+T and NK) cells. Tumor resection supported by neoadjuvant and adjuvant administration of hetIL-15, either alone or in combination with doxorubicin, resulted in the cure of approximately half of the treated animals and the development of anti-4T1 tumor immunity. Our findings demonstrate a significant anti-metastatic potential of hetIL-15 in combination with chemotherapy and surgery and suggest exploring the use of this regimen for the treatment of TNBC.
Collapse
Affiliation(s)
- Vasiliki Stravokefalou
- Human Retrovirus Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD, United States,Department of Pharmacology, Faculty of Medicine, University of Thessaly, Larissa, Greece
| | - Dimitris Stellas
- Human Retrovirus Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD, United States,Department of Chemical Biology, National Hellenic Research Foundation, Athens, Greece
| | - Sevasti Karaliota
- Human Retrovirus Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD, United States,Basic Science Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Frederick, MD, United States
| | - Bethany A. Nagy
- Laboratory Animal Sciences Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Frederick, MD, United States
| | - Antonio Valentin
- Human Retrovirus Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD, United States
| | - Cristina Bergamaschi
- Human Retrovirus Pathogenesis Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD, United States
| | - Konstantinos Dimas
- Department of Pharmacology, Faculty of Medicine, University of Thessaly, Larissa, Greece,*Correspondence: Konstantinos Dimas, ; George N. Pavlakis,
| | - George N. Pavlakis
- Human Retrovirus Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD, United States,*Correspondence: Konstantinos Dimas, ; George N. Pavlakis,
| |
Collapse
|
18
|
Ager CR, Zhang M, Chaimowitz M, Bansal S, Obradovic A, Rogava M, Melms JC, McCann P, Spina C, Drake CG, Dallos MC, Izar B. KLRG1 marks tumor-infiltrating CD4 T cell subsets associated with tumor progression and immunotherapy response. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.01.522340. [PMID: 36711647 PMCID: PMC9881861 DOI: 10.1101/2023.01.01.522340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Current methods for biomarker discovery and target identification in immuno-oncology rely on static snapshots of tumor immunity. To thoroughly characterize the temporal nature of antitumor immune responses, we developed a 34-parameter spectral flow cytometry panel and performed high-throughput analyses in critical contexts. We leveraged two distinct preclinical models that recapitulate cancer immunoediting (NPK-C1) and immune checkpoint blockade (ICB) response (MC38), respectively, and profiled multiple relevant tissues at and around key inflection points of immune surveillance and escape and/or ICB response. Machine learning-driven data analysis revealed a pattern of KLRG1 expression that uniquely identified intratumoral effector CD4 T cell populations that constitutively associate with tumor burden across tumor models, and are lost in tumors undergoing regression in response to ICB. Similarly, a Helios - KLRG1 + subset of tumor-infiltrating regulatory T cells (Tregs) was associated with tumor progression from immune equilibrium to escape, and were also lost in tumors responding to ICB. Validation studies confirmed KLRG1 signatures in human tumorinfiltrating CD4 T cells associate with disease progression in renal cancer. These findings nominate KLRG1 + CD4 T cell populations as subsets for further investigation in cancer immunity and demonstrate the utility of longitudinal spectral flow profiling as an engine of dynamic biomarker and/or target discovery.
Collapse
|
19
|
Roy G, Chakraborty A, Swami B, Pal L, Ahuja C, Basak S, Bhaskar S. Type 1 interferon mediated signaling is indispensable for eliciting anti-tumor responses by Mycobacterium indicus pranii. Front Immunol 2023; 14:1104711. [PMID: 37122749 PMCID: PMC10140407 DOI: 10.3389/fimmu.2023.1104711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 03/23/2023] [Indexed: 05/02/2023] Open
Abstract
Introduction The evolving tumor secretes various immunosuppressive factors that reprogram the tumor microenvironment (TME) to become immunologically cold. Consequently, various immunosuppressive cells like Tregs are recruited into the TME which in turn subverts the anti-tumor response of dendritic cells and T cells.Tumor immunotherapy is a popular means to rejuvenate the immunologically cold TME into hot. Mycobacterium indicus pranii (MIP) has shown strong immunomodulatory activity in different animal and human tumor models and has been approved for treatment of lung cancer (NSCLC) patients as an adjunct therapy. Previously, MIP has shown TLR2/9 mediated activation of antigen presenting cells/Th1 cells and their enhanced infiltration in mouse melanoma but the underlying mechanism by which it is modulating these immune cells is not yet known. Results This study reports for the first time that MIP immunotherapy involves type 1 interferon (IFN) signaling as one of the major signaling pathways to mediate the antitumor responses. Further, it was observed that MIP therapy significantly influenced frequency and activation of different subsets of T cells like regulatory T cells (Tregs) and CD8+ T cells in the TME. It reduces the migration of Tregs into the TME by suppressing the expression of CCL22, a Treg recruiting chemokine on DCs and this process is dependent on type 1 IFN. Simultaneously, in a type 1 IFN dependent pathway, it enhances the activation and effector function of the immunosuppressive tumor resident DCs which in turn effectively induce the proliferation and effector function of the CD8+ T cells. Conclusion This study also provides evidence that MIP induced pro-inflammatory responses including induction of effector function of conventional dendritic cells and CD8+ T cells along with reduction of intratumoral Treg frequency are essentially mediated in a type 1 IFN-dependent pathway.
Collapse
Affiliation(s)
- Gargi Roy
- Product Development Cell, National Institute of Immunology, New Delhi, India
| | - Anush Chakraborty
- Product Development Cell, National Institute of Immunology, New Delhi, India
| | - Bharati Swami
- Product Development Cell, National Institute of Immunology, New Delhi, India
| | - Lalit Pal
- Product Development Cell, National Institute of Immunology, New Delhi, India
| | - Charvi Ahuja
- Product Development Cell, National Institute of Immunology, New Delhi, India
| | - Soumen Basak
- Systems Immunology Lab, National Institute of Immunology, New Delhi, India
| | - Sangeeta Bhaskar
- Product Development Cell, National Institute of Immunology, New Delhi, India
- *Correspondence: Sangeeta Bhaskar,
| |
Collapse
|
20
|
Axisa PP, Yoshida TM, Lucca LE, Kasler HG, Lincoln MR, Pham GH, Del Priore D, Carpier JM, Lucas CL, Verdin E, Sumida TS, Hafler DA. A multiple sclerosis-protective coding variant reveals an essential role for HDAC7 in regulatory T cells. Sci Transl Med 2022; 14:eabl3651. [PMID: 36516268 DOI: 10.1126/scitranslmed.abl3651] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Genome-wide association studies identifying hundreds of susceptibility loci for autoimmune diseases indicate that genes active in immune cells predominantly mediate risk. However, identification and functional characterization of causal variants remain challenging. Here, we focused on the immunomodulatory role of a protective variant of histone deacetylase 7 (HDAC7). This variant (rs148755202, HDAC7.p.R166H) was identified in a study of low-frequency coding variation in multiple sclerosis (MS). Through transcriptomic analyses, we demonstrate that wild-type HDAC7 regulates genes essential for the function of Foxp3+ regulatory T cells (Tregs), an immunosuppressive subset of CD4 T cells that is generally dysfunctional in patients with MS. Moreover, Treg-specific conditional hemizygous deletion of HDAC7 increased the severity of experimental autoimmune encephalitis (EAE), a mouse model of neuroinflammation. In contrast, Tregs transduced with the protective HDAC7 R166H variant exhibited higher suppressive capacity in an in vitro functional assay, mirroring phenotypes previously observed in patient samples. In vivo modeling of the human HDAC7 R166H variant by generation of a knock-in mouse model bearing an orthologous R150H substitution demonstrated decreased EAE severity linked to transcriptomic alterations of brain-infiltrating Tregs, as assessed by single-cell RNA sequencing. Our data suggest that dysregulation of epigenetic modifiers, a distinct molecular class associated with disease risk, may influence disease onset. Last, our approach provides a template for the translation of genetic susceptibility loci to detailed functional characterization, using in vitro and in vivo modeling.
Collapse
Affiliation(s)
- Pierre-Paul Axisa
- Department of Neurology, Yale School of Medicine, New Haven, CT 06510, USA
| | - Tomomi M Yoshida
- Department of Immunobiology, Yale School of Medicine, New Haven, CT 06510, USA
| | - Liliana E Lucca
- Department of Neurology, Yale School of Medicine, New Haven, CT 06510, USA
| | | | - Matthew R Lincoln
- Department of Neurology, Yale School of Medicine, New Haven, CT 06510, USA.,Broad Institute of MIT and Harvard University, Cambridge, MA 02142, USA
| | - Giang H Pham
- Department of Neurology, Yale School of Medicine, New Haven, CT 06510, USA
| | - Dante Del Priore
- Department of Neurology, Yale School of Medicine, New Haven, CT 06510, USA
| | - Jean-Marie Carpier
- Department of Immunobiology, Yale School of Medicine, New Haven, CT 06510, USA
| | - Carrie L Lucas
- Department of Immunobiology, Yale School of Medicine, New Haven, CT 06510, USA
| | - Eric Verdin
- Buck Institute for Research on Aging, Novato, CA 94945, USA
| | - Tomokazu S Sumida
- Department of Neurology, Yale School of Medicine, New Haven, CT 06510, USA.,Broad Institute of MIT and Harvard University, Cambridge, MA 02142, USA
| | - David A Hafler
- Department of Neurology, Yale School of Medicine, New Haven, CT 06510, USA.,Department of Immunobiology, Yale School of Medicine, New Haven, CT 06510, USA.,Broad Institute of MIT and Harvard University, Cambridge, MA 02142, USA
| |
Collapse
|