1
|
Kewang L, Wei Y, Meiye L, Tianyong H, Baohui C. The inhibitory effects of modified HSJZ decoction on NSCLC by regulating regulatory T cells via downregulation of EZH2 and PI3K/AKT pathway. JOURNAL OF ETHNOPHARMACOLOGY 2025; 348:119802. [PMID: 40245962 DOI: 10.1016/j.jep.2025.119802] [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: 01/11/2025] [Revised: 03/29/2025] [Accepted: 04/11/2025] [Indexed: 04/19/2025]
Abstract
ETHNOPHARMACOLOGY REVELANCE Huangqi Si Jun Zi decoction (HSJZ), a modified traditional Chinese medicine formula, is known to enhance immunity. However, its immunomodulatory effects in non-small cell lung cancer (NSCLC) remain poorly understood. AIM OF STUDY This study aimed to investigate the anti-tumor potential of HSJZ in NSCLC and elucidate its mechanisms of action. MATERIAL AND METHODS In vitro studies assessed HSJZ cytotoxicity in 4 tumor cell lines and its ability to activate immune cells from NSCLC patients, followed by evaluating the cytotoxicity of these activated immune cells against NSCLC cell lines. An orthotopic lung cancer model in mice evaluated in vivo anti-tumor activity. Flow cytometry, immunohistochemistry, and Western blot analysis were conducted to analyze immune cell populations, cytokine production, and protein expression levels, including Tregs, CD8+ T cells, EZH2, and PI3K/AKT signaling pathways. RESULTS While HSJZ alone exhibited no direct cytotoxicity on NSCLC cells in vitro, it significantly enhanced immune cell-mediated killing when co-cultured with patient-derived peripheral blood mononuclear cells (PBMCs), accompanied with increased IFN-γ and TNF-α production. In vivo, HSJZ (1 g/kg) treatment in a murine orthotopic lung cancer model resulted in a 54.2 % reduction in tumor burden, as assessed by IVIS spectrum imaging. Mechanistically, HSJZ suppressed regulatory T cells (Tregs) both in vivo and in vitro, as demonstrated by decreased Treg frequency and downregulation of phosphorylated PI3K/AKT signaling in mice bearing Lewis tumor. Moreover, HSJZ significantly inhibited the expression of EZH2, an epigenetic regulator crucial for Treg differentiation. CONCLUSION These findings provide compelling evidence that HSJZ exerts anti-tumor effects in NSCLC by modulating the immune microenvironment, particularly through the inhibition of Tregs activity, as assessed by in vitro co-culture system, in vivo mouse orthotopic Lewis cancer model, immunohistochemistry, etc. Our results support the potential clinical application of HSJZ as an adjunct therapy for NSCLC patients.
Collapse
MESH Headings
- Enhancer of Zeste Homolog 2 Protein/metabolism
- Animals
- Carcinoma, Non-Small-Cell Lung/drug therapy
- Carcinoma, Non-Small-Cell Lung/immunology
- Carcinoma, Non-Small-Cell Lung/pathology
- Carcinoma, Non-Small-Cell Lung/metabolism
- T-Lymphocytes, Regulatory/drug effects
- T-Lymphocytes, Regulatory/immunology
- Humans
- Lung Neoplasms/drug therapy
- Lung Neoplasms/immunology
- Lung Neoplasms/pathology
- Proto-Oncogene Proteins c-akt/metabolism
- Drugs, Chinese Herbal/pharmacology
- Drugs, Chinese Herbal/therapeutic use
- Down-Regulation/drug effects
- Signal Transduction/drug effects
- Phosphatidylinositol 3-Kinases/metabolism
- Mice
- Cell Line, Tumor
- Male
- Antineoplastic Agents, Phytogenic/pharmacology
- Female
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Luo Kewang
- People's Hospital of Longhua, The Affiliated Hospital of Southern Medical University, Shenzhen, China; School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China.
| | - Ye Wei
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Li Meiye
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Hu Tianyong
- Department of Otolaryngology, Shenzhen Longgang Otolaryngology Hospital & Shenzhen Otolaryngology Research Institute, China
| | - Cheng Baohui
- Department of Otolaryngology, Shenzhen Longgang Otolaryngology Hospital & Shenzhen Otolaryngology Research Institute, China.
| |
Collapse
|
2
|
Zheng J, Yi Y, Tian T, Luo S, Liang X, Bai Y. ICI-induced cardiovascular toxicity: mechanisms and immune reprogramming therapeutic strategies. Front Immunol 2025; 16:1550400. [PMID: 40356915 PMCID: PMC12066601 DOI: 10.3389/fimmu.2025.1550400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Accepted: 04/07/2025] [Indexed: 05/15/2025] Open
Abstract
The advent of immune checkpoint inhibitors (ICIs) has revolutionized cancer treatment, offering life-saving benefits to tumor patients. However, the utilize of ICI agents is often accompanied by immune-related adverse events (irAEs), among which cardiovascular toxicities have attracted more and more attention. ICI induced cardiovascular toxicities predominantly present as acute myocarditis and chronic atherosclerosis, both of which are driven by excessive immune activation. Reprogramming of T cells and macrophages has been demonstrated as a pivotal factor in the pathogenesis of these complications. Therapeutic strategies targeting glycolysis, fatty acid oxidation, reactive oxygen species (ROS) production and some other key signaling have shown promise in mitigating immune hyperactivation and inflammation. In this review, we explored the intricate mechanisms underlying ICI-induced cardiovascular toxicities and highlighted the protective potential of immune reprogramming. We emphasize the roles of T cell and macrophage reprogramming in the heart and vasculature, showcasing their contributions to both short-term and long-term regulation of cardiovascular health. Ultimately, a deeper understanding of these processes will not only enhance the safety of ICIs but also pave the way for innovative strategies to manage immune-related toxicities in cancers therapy.
Collapse
Affiliation(s)
| | | | | | | | | | - Yu Bai
- Department of Reproductive Medicine, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second Hospital, West China School of Medicine, West China School of Pharmacy, Sichuan University, Chengdu, China
| |
Collapse
|
3
|
Albini A, Di Paola L, Mei G, Baci D, Fusco N, Corso G, Noonan D. Inflammation and cancer cell survival: TRAF2 as a key player. Cell Death Dis 2025; 16:292. [PMID: 40229245 PMCID: PMC11997178 DOI: 10.1038/s41419-025-07609-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: 12/05/2024] [Revised: 03/25/2025] [Accepted: 03/31/2025] [Indexed: 04/16/2025]
Abstract
TNF receptor-associated factor 2 (TRAF2) plays a crucial role in both physiological and pathological processes. It takes part in the regulation of cell survival and death, tissue regeneration, development, endoplasmic reticulum stress response, autophagy, homeostasis of the epithelial barrier and regulation of adaptive and innate immunity. Initially identified for its interaction with TNF receptor 2 (TNFR2), TRAF2 contains a TRAF domain that enables homo- and hetero-oligomerization, allowing it to interact with multiple receptors and signaling molecules. While best known for mediating TNFR1 and TNFR2 signaling, TRAF2 also modulates other receptor pathways, including MAPK, NF-κB, and Wnt/β-catenin cascades. By regulating NF-κB-inducing kinase (NIK), TRAF2 is a key activator of the alternative NF-κB pathway, linking it to inflammatory diseases, immune dysfunction, and tumorigenesis. In the innate immune system, TRAF2 influences macrophage differentiation, activation, and survival and stimulates natural killer cell cytotoxicity. In the adaptive immune system, it represses effector B- and T-cell activity while sustaining regulatory T-cell function, thus promoting immune suppression. The lack of fine-tuning of TRAF2 activity leads to excessive NF-kB activation, driving chronic inflammation and autoimmunity. Although TRAF2 can act as a tumor suppressor, it is predominantly described as a tumor promoter, as its expression has been correlated with increased metastatic potential and poorer prognosis in several types of cancer. Targeting TRAF2 or TRAF2-dependent signaling pathways might represent a promising anti-cancer therapeutic strategy.
Collapse
Grants
- The work was also supported by the Italian Ministry of Health Ricerca Corrente to IRCCS IEO, European Institute of Oncology, and IRCCS MultiMedica, Italy.
- PRIN 2022, grant 2022PJKF88 The work was also supported by the Italian Ministry of Health Ricerca Corrente to IRCCS IEO, European Institute of Oncology, and IRCCS MultiMedica, Italy.
- PRIN 2022 The work was also supported by the Italian Ministry of Health Ricerca Corrente to IRCCS IEO, European Institute of Oncology, and IRCCS MultiMedica, Italy.
- "Umberto Veronesi" Foundation project: "Massive CDH1 genetic screening in the so-called hereditary breast-gastric cancer syndrome". The work was also supported by the Italian Ministry of Health Ricerca Corrente to IRCCS IEO, European Institute of Oncology, and IRCCS MultiMedica, Italy.
Collapse
Affiliation(s)
- Adriana Albini
- European Institute of Oncology (IEO), Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Milan, Italy.
| | - Luisa Di Paola
- Unit of Chemical-Physics Fundamentals in Chemical Engineering, Faculty Department of Science and Technology for Sustainable Development and One Health, Università Campus Bio-Medico, Rome, Italy
| | - Giampiero Mei
- Department of Experimental Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Denisa Baci
- Department of Biotechnology and Life Sciences, University of Insubria, Varese, Italy
- Molecular Cardiology Laboratory, IRCCS-Policlinico San Donato, Milan, Italy
| | - Nicola Fusco
- European Institute of Oncology (IEO), Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Milan, Italy
| | - Giovanni Corso
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy.
- Division of Breast Surgery, European Institute of Oncology (IEO), Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Milan, Italy.
| | - Douglas Noonan
- Department of Biotechnology and Life Sciences, University of Insubria, Varese, Italy
- IRCCS MultiMedica, Milan, Italy
| |
Collapse
|
4
|
White TLA, Henry DJ, Roberts SDA, Jin Y, Gong YN, Morel PA. HnRNP L is essential for peripheral T cell proliferation and survival. Front Immunol 2025; 16:1543145. [PMID: 40276511 PMCID: PMC12018431 DOI: 10.3389/fimmu.2025.1543145] [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: 12/10/2024] [Accepted: 03/14/2025] [Indexed: 04/26/2025] Open
Abstract
Introduction During T cell development, heterogeneous nuclear ribonucleoprotein (hnRNP) L is known to regulate CD4 T helper subset differentiation, the proliferation and migration of thymocytes, as loss of hnRNP L in early T cell development results in a failure of T cells to reach the periphery. Methods To better understand the role of hnRNP L in modulating peripheral T cell trafficking and function, we analyzed T survival and activation in newly generated CD4Cre x hnRNP LFl/Fl (KO) mice. In vitro and in vivo analyses of CD4 T cell differentiation, T cell proliferation and death post activation were performed. Results Our initial study of the steady state profile of the KO mice showed normal migration of T cells from the thymus, but peripheral T cell numbers were reduced. Analysis of TCR-mediated signaling pathways revealed normal early T cell activation. However, T cells lacking hnRNP L had marked defects in their ability to differentiate into T helper cell subsets due to reduced proliferation and increased death. In vivo, using immunization studies, KO CD4 T cells failed to fully differentiate into T follicular helper (Tfh) cells and were unable to support the formation of germinal center B cells. Death of activated hnRNP L KO cells could be reversed by treating the cells with zVAD, a pan-caspase inhibitor. In addition, hnRNP L KO cells failed to upregulate the anti-apoptotic protein Bcl-XL following activation. Discussion These studies suggest that hnRNP L plays an important role in T cell activation and survival. Our studies suggest that hnRNP L plays a critical pro-survival role in activated T cells and that alternative splicing of factors that prevent apoptosis may be an important mechanism by which this is achieved.
Collapse
Affiliation(s)
| | | | | | | | | | - Penelope A. Morel
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| |
Collapse
|
5
|
Lu Z, Zhang C, Zhang J, Su W, Wang G, Wang Z. The Kynurenine Pathway and Indole Pathway in Tryptophan Metabolism Influence Tumor Progression. Cancer Med 2025; 14:e70703. [PMID: 40103267 PMCID: PMC11919716 DOI: 10.1002/cam4.70703] [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: 09/19/2024] [Revised: 01/22/2025] [Accepted: 02/04/2025] [Indexed: 03/20/2025] Open
Abstract
Tryptophan (Trp), an essential amino acid, is solely acquired through dietary intake. It is vital for protein biosynthesis and acts as a precursor for numerous key bioactive compounds. The Kynurenine Pathway and the Indole Pathway are the main metabolic routes and are extensively involved in the occurrence and progression of diseases in the digestive, nervous, and urinary systems. In the Kynurenine Pathway, enzymes crucial to tryptophan metabolism, indoleamine-2,3-dioxygenase 1 (IDO1), IDO2, and Trp-2,3-dioxygenase (TDO), trigger tumor immune resistance within the tumor microenvironment and nearby lymph nodes by depleting Trp or by activating the Aromatic Hydrocarbon Receptor (AhR) through its metabolites. Furthermore, IDO1 can influence immune responses via non-enzymatic pathways. The Kynurenine Pathway exerts its effects on tumor growth through various mechanisms, including NAD+ regulation, angiogenesis promotion, tumor metastasis enhancement, and the inhibition of tumor ferroptosis. In the Indole Pathway, indole and its related metabolites are involved in gastrointestinal homeostasis, tumor immunity, and drug resistance. The gut microbiota related to indole metabolism plays a critical role in determining the effectiveness of tumor treatment strategies and can influence the efficacy of immunochemotherapy. It is worth noting that there are conflicting effects of the Kynurenine Pathway and the Indole Pathway on the same tumor phenotype. For example, different tryptophan metabolites affect the cell cycle differently, and indole metabolism has inconsistent protective effects on tumors in different regions. These differences may hold potential for enhancing therapeutic efficacy.
Collapse
Affiliation(s)
- Zhanhui Lu
- Department of Medical Oncology, Longhua HospitalShanghai University of Traditional Chinese MedicineShanghaiChina
- Shanghai University of Traditional Chinese MedicineShanghaiChina
- Cancer Institute, Longhua HospitalShanghai University of Traditional Chinese MedicineShanghaiChina
| | - Chengcheng Zhang
- Department of Medical Oncology, Longhua HospitalShanghai University of Traditional Chinese MedicineShanghaiChina
- Shanghai University of Traditional Chinese MedicineShanghaiChina
- Cancer Institute, Longhua HospitalShanghai University of Traditional Chinese MedicineShanghaiChina
| | - Jia Zhang
- Provincial Hospital Affiliated to Shandong First Medical UniversityJinanShandongChina
| | - Wan Su
- Department of Medical Oncology, Longhua HospitalShanghai University of Traditional Chinese MedicineShanghaiChina
| | - Guoying Wang
- Department of Critical Care MedicineThe Second People's Hospital of DongyingDongyingShandongChina
| | - Zhongqi Wang
- Department of Medical Oncology, Longhua HospitalShanghai University of Traditional Chinese MedicineShanghaiChina
| |
Collapse
|
6
|
Gan M, Liu N, Li W, Chen M, Bai Z, Liu D, Liu S. Metabolic targeting of regulatory T cells in oral squamous cell carcinoma: new horizons in immunotherapy. Mol Cancer 2024; 23:273. [PMID: 39696340 DOI: 10.1186/s12943-024-02193-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Accepted: 12/03/2024] [Indexed: 12/20/2024] Open
Abstract
Oral squamous cell carcinoma (OSCC) is a prevalent oral malignancy, which poses significant health risks with a high mortality rate. Regulatory T cells (Tregs), characterized by their immunosuppressive capabilities, are intricately linked to OSCC progression and patient outcomes. The metabolic reprogramming of Tregs within the OSCC tumor microenvironment (TME) underpins their function, with key pathways such as the tryptophan-kynurenine-aryl hydrocarbon receptor, PI3K-Akt-mTOR and nucleotide metabolism significantly contributing to their suppressive activities. Targeting these metabolic pathways offers a novel therapeutic approach to reduce Treg-mediated immunosuppression and enhance anti-tumor responses. This review explores the metabolic dependencies and pathways that sustain Treg function in OSCC, highlighting key metabolic adaptations such as glycolysis, fatty acid oxidation, amino acid metabolism and PI3K-Akt-mTOR signaling pathway that enable Tregs to thrive in the challenging conditions of the TME. Additionally, the review discusses the influence of the oral microbiome on Treg metabolism and evaluates potential therapeutic strategies targeting these metabolic pathways. Despite the promising potential of these interventions, challenges such as selectivity, toxicity, tumor heterogeneity, and resistance mechanisms remain. The review concludes with perspectives on personalized medicine and integrative approaches, emphasizing the need for continued research to translate these findings into effective clinical applications for OSCC treatment.
Collapse
Affiliation(s)
- Menglai Gan
- Department of Dental Materials, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, No. 117 Nanjing North Street, Heping District, Shenyang, 110002, Liaoning, China
| | - Nanshu Liu
- Department of Emergency and Oral Medicine, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, No. 117 Nanjing North Street, Heping District, Shenyang, 110002, Liaoning, China
| | - Wenting Li
- Department of Dental Materials, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, No. 117 Nanjing North Street, Heping District, Shenyang, 110002, Liaoning, China
| | - Mingwei Chen
- Department of Dental Materials, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, No. 117 Nanjing North Street, Heping District, Shenyang, 110002, Liaoning, China
| | - Zhongyu Bai
- Department of Emergency and Oral Medicine, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, No. 117 Nanjing North Street, Heping District, Shenyang, 110002, Liaoning, China
| | - Dongjuan Liu
- Department of Emergency and Oral Medicine, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, No. 117 Nanjing North Street, Heping District, Shenyang, 110002, Liaoning, China.
| | - Sai Liu
- Department of Dental Materials, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, No. 117 Nanjing North Street, Heping District, Shenyang, 110002, Liaoning, China.
| |
Collapse
|
7
|
Hoelting K, Madlmayr A, Hoeger B, Lewitz D, Weng M, Haider T, Duggan M, Ross R, Horgen FD, Sperandio M, Dietrich A, Gudermann T, Zierler S. TRPM7 activity drives human CD4 T-cell activation and differentiation in a magnesium dependent manner. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.04.626765. [PMID: 39677770 PMCID: PMC11643021 DOI: 10.1101/2024.12.04.626765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
T lymphocyte activation is a crucial process in the regulation of innate and adaptive immune responses. The ion channel-kinase TRPM7 has previously been implicated in cellular Mg2+ homeostasis, proliferation, and immune cell modulation. Here, we show that pharmacological and genetic silencing of TRPM7 leads to diminished human CD4 T-cell activation and proliferation following TCR mediated stimulation. In both primary human CD4 T cells and CRISPR/Cas-9 engineered Jurkat T cells, loss of TRPM7 led to altered Mg2+ homeostasis, Ca2+ signaling, reduced NFAT translocation, decreased IL-2 secretion and ultimately diminished proliferation and differentiation. While the activation of primary human CD4 T cells was dependent on TRPM7, polarization of naïve CD4 T cells into regulatory T cells (Treg) was not. Taken together, these results highlight TRPM7 as a key protein of cellular Mg2+ homeostasis and CD4 T-cell activation. Its role in lymphocyte activation suggests therapeutic potential for TRPM7 in numerous T-cell mediated diseases.
Collapse
Affiliation(s)
- Kilian Hoelting
- Walther Straub Institute of Pharmacology and Toxicology, Ludwig-Maximilians-Universität München, Goethestr.33, 80336 Munich, Germany
| | - Anna Madlmayr
- Institute of Pharmacology, Johannes Kepler University Linz, Altenbergerstr. 69, 4040 Linz, Austria
| | - Birgit Hoeger
- Institute of Pharmacology, Johannes Kepler University Linz, Altenbergerstr. 69, 4040 Linz, Austria
| | - Dorothea Lewitz
- Walther Straub Institute of Pharmacology and Toxicology, Ludwig-Maximilians-Universität München, Goethestr.33, 80336 Munich, Germany
| | - Marius Weng
- Institute of Pharmacology, Johannes Kepler University Linz, Altenbergerstr. 69, 4040 Linz, Austria
| | - Tanja Haider
- Institute of Pharmacology, Johannes Kepler University Linz, Altenbergerstr. 69, 4040 Linz, Austria
| | - Michelle Duggan
- Institute of Pharmacology, Johannes Kepler University Linz, Altenbergerstr. 69, 4040 Linz, Austria
| | - Rylee Ross
- Laboratory of Marine Biological Chemistry, Hawai’i Pacific University, 1042 Fort Street Mall, Honolulu, HI 96813, USA
| | - F. David Horgen
- Laboratory of Marine Biological Chemistry, Hawai’i Pacific University, 1042 Fort Street Mall, Honolulu, HI 96813, USA
| | - Markus Sperandio
- Institute of Cardiovascular Physiology and Pathophysiology, Biomedical Center, Ludwig-Maximilians-Universität München, Großharderner Str. 9, 82152 Planegg-Martinsried, Germany
| | - Alexander Dietrich
- Walther Straub Institute of Pharmacology and Toxicology, Ludwig-Maximilians-Universität München, Goethestr.33, 80336 Munich, Germany
| | - Thomas Gudermann
- Walther Straub Institute of Pharmacology and Toxicology, Ludwig-Maximilians-Universität München, Goethestr.33, 80336 Munich, Germany
| | - Susanna Zierler
- Walther Straub Institute of Pharmacology and Toxicology, Ludwig-Maximilians-Universität München, Goethestr.33, 80336 Munich, Germany
- Institute of Pharmacology, Johannes Kepler University Linz, Altenbergerstr. 69, 4040 Linz, Austria
| |
Collapse
|
8
|
Costaguta A, Costaguta G, Álvarez F. Autoimmune hepatitis: Towards a personalized treatment. World J Hepatol 2024; 16:1225-1242. [PMID: 39606175 PMCID: PMC11586748 DOI: 10.4254/wjh.v16.i11.1225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 09/02/2024] [Accepted: 10/11/2024] [Indexed: 11/06/2024] Open
Abstract
Autoimmune hepatitis is an uncommon condition that affects both adults and children and is characterized by chronic and recurrent inflammatory activity in the liver. This inflammation is accompanied by elevated IgG and autoantibody levels. Historically, treatment consists of steroids with the addition of azathioprine, which results in remission in approximately 80% of patients. Despite significant advancements in our understanding of the immune system over the past two decades, few modifications have been made to treatment algorithms, which have remained largely unchanged since they were first proposed more than 40 years ago. This review summarized the various treatment options currently available as well as our experiences using them. Although steroids are the standard treatment for induction therapy, other medications may be considered. Cyclosporin A, a calcineurin inhibitor that decreases T cell activation, has proven effective for induction of remission, but its long-term side effects limit its appeal for maintenance. Tacrolimus, a drug belonging to the same family, has been used in patients with refractory diseases with fewer side effects. Sirolimus and everolimus have interesting effects on regulatory T cell populations and may become viable options in the future. Mycophenolate mofetil is not effective for induction but is a valid alternative for patients who are intolerant to azathioprine. B cell-depleting drugs, such as rituximab and belimumab, have been successfully used in refractory cases and are useful in both the short and long term. Other promising treatments include anti-tumor necrosis factors, Janus kinases inhibitors, and chimeric antigen receptor T cell therapy. This growing armamentarium allows us to imagine a more tailored approach to the treatment of autoimmune hepatitis in the near future.
Collapse
Affiliation(s)
- Alejandro Costaguta
- Department of Hepatology and Liver Transplant Unit, Sanatorio de Niños de Rosario, Rosario 2000, Santa Fe, Argentina.
| | - Guillermo Costaguta
- Department of Gastroenterology, Hepatology, and Nutrition, CHU Sainte-Justine, Montreal H3T 1C5, Quebec, Canada
| | - Fernando Álvarez
- Department of Pediatrics, CHU Sainte-Justine, Montreal H3T 1C5, Quebec, Canada
| |
Collapse
|
9
|
Singer M, Elsayed AM, Husseiny MI. Regulatory T-cells: The Face-off of the Immune Balance. FRONT BIOSCI-LANDMRK 2024; 29:377. [PMID: 39614434 DOI: 10.31083/j.fbl2911377] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 07/29/2024] [Accepted: 08/13/2024] [Indexed: 12/01/2024]
Abstract
Regulatory T-cells (Tregs) play a crucial role in maintaining immune homeostasis, ensuring a balanced immune response. Tregs primarily operate in an antigen-specific fashion, facilitated by their distinct distribution within discrete niches. Tregs have been studied extensively, from their point of origin in the thymus origin to their fate in the periphery or organs. Signals received from antigen-presenting cells (APCs) stimulate Tregs to dampen inflammation. Almost all tumors are characterized by a pathological abundance of immune suppression in their microenvironment. Conversely, the lack thereof proves detrimental to immunological disorders. Achieving a balanced expression of Tregs in relation to other immune compartments is important in establishing an effective and adaptable immune tolerance towards cancer cells and autoantigens. In the context of cancer, it is essential to decrease the frequency of Tregs to overcome tumor suppression. A lower survival rate is associated with the presence of excessive exhausted effector immune cells and an increased frequency of regulatory cells. However, when it comes to treating graft rejection and autoimmune diseases, the focus lies on immune tolerance and the transfer of Tregs. Here, we explore the complex mechanisms that Tregs use in human disease to balance effector immune cells.
Collapse
Affiliation(s)
- Mahmoud Singer
- School of Medicine, University of California Irvine, Irvine, CA 92617, USA
| | - Ahmed M Elsayed
- Division of Infectious Diseases, The Lundquist Institute at Harbor-UCLA Medical Center, Torrance, CA 90502, USA
| | - Mohamed I Husseiny
- Department of Translational Research & Cellular Therapeutics, Arthur Riggs Diabetes & Metabolism Research Institute, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA 91010, USA
| |
Collapse
|
10
|
Son B, Lee W, Kim H, Shin H, Park HH. Targeted therapy of cancer stem cells: inhibition of mTOR in pre-clinical and clinical research. Cell Death Dis 2024; 15:696. [PMID: 39349424 PMCID: PMC11442590 DOI: 10.1038/s41419-024-07077-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 09/10/2024] [Accepted: 09/13/2024] [Indexed: 10/02/2024]
Abstract
Cancer stem cells (CSCs) are a type of stem cell that possesses not only the intrinsic abilities of stem cells but also the properties of cancer cells. Therefore, CSCs are known to have self-renewal and outstanding proliferation capacity, along with the potential to differentiate into specific types of tumor cells. Cancers typically originate from CSCs, making them a significant target for tumor treatment. Among the related cascades of the CSCs, mammalian target of rapamycin (mTOR) pathway is regarded as one of the most important signaling pathways because of its association with significant upstream signaling: phosphatidylinositol 3‑kinase/protein kinase B (PI3K/AKT) pathway and mitogen‑activated protein kinase (MAPK) cascade, which influence various activities of stem cells, including CSCs. Recent studies have shown that the mTOR pathway not only affects generation of CSCs but also the maintenance of their pluripotency. Furthermore, the maintenance of pluripotency or differentiation into specific types of cancer cells depends on the regulation of the mTOR signal in CSCs. Consequently, the clinical potential and importance of mTOR in effective cancer therapy are increasing. In this review, we demonstrate the association between the mTOR pathway and cancer, including CSCs. Additionally, we discuss a new concept for anti-cancer drug development aimed at overcoming existing drawbacks, such as drug resistance, by targeting CSCs through mTOR inhibition.
Collapse
Affiliation(s)
- Boram Son
- Department of Bioengineering, Hanyang University, Seoul, 04763, Republic of Korea
- Department of Bio and Fermentation Convergence Technology, Kookmin University, Seoul, 02707, Republic of Korea
| | - Wonhwa Lee
- Department of Chemistry, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Hyeonjeong Kim
- Department of Bioengineering, Hanyang University, Seoul, 04763, Republic of Korea
| | - Heungsoo Shin
- Department of Bioengineering, Hanyang University, Seoul, 04763, Republic of Korea.
| | - Hee Ho Park
- Department of Bioengineering, Hanyang University, Seoul, 04763, Republic of Korea.
- Research Institute for Convergence of Basic Science, Hanyang University, Seoul, 04763, Republic of Korea.
| |
Collapse
|
11
|
Ma W, Luo H, Lv J, Wen P, Liu G, Yu Z, Yang Z, Huang W. Immunoregulatory Engineering of Semiconducting Charge-Reversal Nanoantioxidant for Ameliorating Cancer Radioimmunotheranostics. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2402929. [PMID: 38847976 DOI: 10.1002/adma.202402929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 05/28/2024] [Indexed: 06/18/2024]
Abstract
Radiotherapy (RT) is a crucial clinical modality for cancer. However, nonselectivity, toxicity to normal tissues, and radio-resistance severely limit RT applications. This study develops a versatile X-ray theranostic nano-antioxidant (XTN) to prevent normal tissues from oxidative damage and induce systematic and robust anticancer immunity. XTN owns NIR-II photoacoustic (PA) imaging properties for precise discrimination of the tumor margin through, thereby improving the accuracy of RT. Additionally, XTN is a nano-antioxidant to enhance the cell viability of normal cells after irradiation. Most importantly, XTN scavenges reactive oxygen species (ROS) in the TME to preserve the stimulatory activity of released high mobility group protein B1 to dendritic cells (DCs) and recover T cells' immune function. Meanwhile, XTN achieves charge-reversal specifically releasing an immunomodulator (demethylcantharidin, DMC) in the acidic TME. Moreover, the specifically released DMC inhibits protein phosphatase-2A activity and reduces regulatory T cell (Treg) differentiation. In the bilateral 4T1 tumor model, XTN-mediated radioimmunotherapy remarkably boosts a systemic antitumor immune response and induces durable immunological memory against tumor growth.
Collapse
Affiliation(s)
- Wen Ma
- Strait Laboratory of Flexible Electronics (SLoFE), Fujian Key Laboratory of Flexible Electronics, Strait Institute of Flexible Electronics (SIFE, Future Technologies), Fujian Normal University, Fuzhou, Fujian, 350117, China
| | - Haifen Luo
- Strait Laboratory of Flexible Electronics (SLoFE), Fujian Key Laboratory of Flexible Electronics, Strait Institute of Flexible Electronics (SIFE, Future Technologies), Fujian Normal University, Fuzhou, Fujian, 350117, China
| | - Jingqi Lv
- Strait Laboratory of Flexible Electronics (SLoFE), Fujian Key Laboratory of Flexible Electronics, Strait Institute of Flexible Electronics (SIFE, Future Technologies), Fujian Normal University, Fuzhou, Fujian, 350117, China
| | - Peiye Wen
- Strait Laboratory of Flexible Electronics (SLoFE), Fujian Key Laboratory of Flexible Electronics, Strait Institute of Flexible Electronics (SIFE, Future Technologies), Fujian Normal University, Fuzhou, Fujian, 350117, China
| | - Gang Liu
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang an Biomedicine Laboratory, Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, 361102, China
| | - Zhiqiang Yu
- Department of Laboratory Medicine, Dongguan Institute of Clinical Cancer Research, Affiliated Dongguan Hospital, Southern Medical University, Dongguan, Guangdong, 523018, China
| | - Zhen Yang
- Strait Laboratory of Flexible Electronics (SLoFE), Fujian Key Laboratory of Flexible Electronics, Strait Institute of Flexible Electronics (SIFE, Future Technologies), Fujian Normal University, Fuzhou, Fujian, 350117, China
| | - Wei Huang
- Strait Laboratory of Flexible Electronics (SLoFE), Fujian Key Laboratory of Flexible Electronics, Strait Institute of Flexible Electronics (SIFE, Future Technologies), Fujian Normal University, Fuzhou, Fujian, 350117, China
- Frontiers Science Center for Flexible Electronics, Xi'an Institute of Flexible Electronics (IFE), Northwestern Polytechnical University, Xi'an, 710072, China
| |
Collapse
|
12
|
Lepore MT, Bruzzaniti S, La Rocca C, Fusco C, Carbone F, Mottola M, Zuccarelli B, Lanzillo R, Brescia Morra V, Maniscalco GT, De Simone S, Procaccini C, Porcellini A, De Rosa V, Galgani M, Cassano S, Matarese G. Deciphering the role of protein kinase A in the control of FoxP3 expression in regulatory T cells in health and autoimmunity. Sci Rep 2024; 14:17571. [PMID: 39080325 PMCID: PMC11289137 DOI: 10.1038/s41598-024-68098-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 07/18/2024] [Indexed: 08/02/2024] Open
Abstract
The molecular mechanisms that govern differential T cell development from CD4+CD25-conventional T (Tconv) into CD4+CD25+ forkhead-box-P3+ (FoxP3+) inducible regulatory T (iTreg) cells remain unclear. Herein, we investigated the relative contribution of protein kinase A (PKA) in this process. Mechanistically, we found that PKA controlled the efficiency of human iTreg cell generation through the expression of different FoxP3 splicing variants containing or not the exon 2. We found that transient PKA inhibition reduced the recruitment of cAMP-responsive element-binding protein (CREB) on regulatory regions of the FoxP3 gene, a condition that is associated with an impaired acquisition of their suppressive capacity in vitro. To corroborate our findings in a human model of autoimmunity, we measured CREB phosphorylation and FoxP3 levels in iTreg cells from treatment-naïve relapsing-remitting (RR)-multiple sclerosis (MS) subjects. Interestingly, both phospho-CREB and FoxP3 induction directly correlated and were significantly reduced in RR-MS patients, suggesting a previously unknown mechanism involved in the induction and function of human iTreg cells.
Collapse
Affiliation(s)
- Maria Teresa Lepore
- Laboratorio di Immunologia, Istituto per l'Endocrinologia e l'Oncologia Sperimentale "G. Salvatore", Consiglio Nazionale delle Ricerche, Naples, Italy
| | - Sara Bruzzaniti
- Laboratorio di Immunologia, Istituto per l'Endocrinologia e l'Oncologia Sperimentale "G. Salvatore", Consiglio Nazionale delle Ricerche, Naples, Italy
| | - Claudia La Rocca
- Laboratorio di Immunologia, Istituto per l'Endocrinologia e l'Oncologia Sperimentale "G. Salvatore", Consiglio Nazionale delle Ricerche, Naples, Italy
| | - Clorinda Fusco
- Laboratorio di Immunologia, Istituto per l'Endocrinologia e l'Oncologia Sperimentale "G. Salvatore", Consiglio Nazionale delle Ricerche, Naples, Italy
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli "Federico II", Naples, Italy
| | - Fortunata Carbone
- Laboratorio di Immunologia, Istituto per l'Endocrinologia e l'Oncologia Sperimentale "G. Salvatore", Consiglio Nazionale delle Ricerche, Naples, Italy
- Unità di Neuroimmunologia, IRCCS Fondazione Santa Lucia, Rome, Italy
| | - Maria Mottola
- UOC di Medicina Trasfusionale, AORN Ospedale dei Colli, Ospedale Monaldi, Naples, Italy
| | - Bruno Zuccarelli
- UOC di Medicina Trasfusionale, AORN Ospedale dei Colli, Ospedale Monaldi, Naples, Italy
| | - Roberta Lanzillo
- Dipartimento di Neuroscienze, Scienze Riproduttive ed Odontostomatologiche, Università degli Studi di Napoli "Federico II", Naples, Italy
| | - Vincenzo Brescia Morra
- Dipartimento di Neuroscienze, Scienze Riproduttive ed Odontostomatologiche, Università degli Studi di Napoli "Federico II", Naples, Italy
| | - Giorgia Teresa Maniscalco
- Dipartimento di Neurologia, Centro Regionale Sclerosi Multipla, Azienda Ospedaliera "A. Cardarelli", Naples, Italy
| | - Salvatore De Simone
- Laboratorio di Immunologia, Istituto per l'Endocrinologia e l'Oncologia Sperimentale "G. Salvatore", Consiglio Nazionale delle Ricerche, Naples, Italy
| | - Claudio Procaccini
- Laboratorio di Immunologia, Istituto per l'Endocrinologia e l'Oncologia Sperimentale "G. Salvatore", Consiglio Nazionale delle Ricerche, Naples, Italy
- Unità di Neuroimmunologia, IRCCS Fondazione Santa Lucia, Rome, Italy
| | - Antonio Porcellini
- Dipartimento di Biologia, Università degli Studi di Napoli "Federico II", Naples, Italy
| | - Veronica De Rosa
- Laboratorio di Immunologia, Istituto per l'Endocrinologia e l'Oncologia Sperimentale "G. Salvatore", Consiglio Nazionale delle Ricerche, Naples, Italy
| | - Mario Galgani
- Laboratorio di Immunologia, Istituto per l'Endocrinologia e l'Oncologia Sperimentale "G. Salvatore", Consiglio Nazionale delle Ricerche, Naples, Italy
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli "Federico II", Naples, Italy
| | - Silvana Cassano
- Laboratorio di Immunologia, Istituto per l'Endocrinologia e l'Oncologia Sperimentale "G. Salvatore", Consiglio Nazionale delle Ricerche, Naples, Italy
| | - Giuseppe Matarese
- Laboratorio di Immunologia, Istituto per l'Endocrinologia e l'Oncologia Sperimentale "G. Salvatore", Consiglio Nazionale delle Ricerche, Naples, Italy.
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli "Federico II", Naples, Italy.
| |
Collapse
|
13
|
Sumida TS, Cheru NT, Hafler DA. The regulation and differentiation of regulatory T cells and their dysfunction in autoimmune diseases. Nat Rev Immunol 2024; 24:503-517. [PMID: 38374298 PMCID: PMC11216899 DOI: 10.1038/s41577-024-00994-x] [Citation(s) in RCA: 37] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/15/2024] [Indexed: 02/21/2024]
Abstract
The discovery of FOXP3+ regulatory T (Treg) cells as a distinct cell lineage with a central role in regulating immune responses provided a deeper understanding of self-tolerance. The transcription factor FOXP3 serves a key role in Treg cell lineage determination and maintenance, but is not sufficient to enable the full potential of Treg cell suppression, indicating that other factors orchestrate the fine-tuning of Treg cell function. Moreover, FOXP3-independent mechanisms have recently been shown to contribute to Treg cell dysfunction. FOXP3 mutations in humans cause lethal fulminant systemic autoinflammation (IPEX syndrome). However, it remains unclear to what degree Treg cell dysfunction is contributing to the pathophysiology of common autoimmune diseases. In this Review, we discuss the origins of Treg cells in the periphery and the multilayered mechanisms by which Treg cells are induced, as well as the FOXP3-dependent and FOXP3-independent cellular programmes that maintain the suppressive function of Treg cells in humans and mice. Further, we examine evidence for Treg cell dysfunction in the context of common autoimmune diseases such as multiple sclerosis, inflammatory bowel disease, systemic lupus erythematosus and rheumatoid arthritis.
Collapse
Affiliation(s)
- Tomokazu S Sumida
- Department of Neurology, Yale School of Medicine, New Haven, CT, USA.
| | - Nardos T Cheru
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA
| | - David A Hafler
- Department of Neurology, Yale School of Medicine, New Haven, CT, USA.
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA.
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| |
Collapse
|
14
|
Ramapriyan R, Vykunta VS, Vandecandelaere G, Richardson LGK, Sun J, Curry WT, Choi BD. Altered cancer metabolism and implications for next-generation CAR T-cell therapies. Pharmacol Ther 2024; 259:108667. [PMID: 38763321 DOI: 10.1016/j.pharmthera.2024.108667] [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: 02/16/2024] [Revised: 04/30/2024] [Accepted: 05/14/2024] [Indexed: 05/21/2024]
Abstract
This review critically examines the evolving landscape of chimeric antigen receptor (CAR) T-cell therapy in treating solid tumors, with a particular focus on the metabolic challenges within the tumor microenvironment. CAR T-cell therapy has demonstrated remarkable success in hematologic malignancies, yet its efficacy in solid tumors remains limited. A significant barrier is the hostile milieu of the tumor microenvironment, which impairs CAR T-cell survival and function. This review delves into the metabolic adaptations of cancer cells and their impact on immune cells, highlighting the competition for nutrients and the accumulation of immunosuppressive metabolites. It also explores emerging strategies to enhance CAR T-cell metabolic fitness and persistence, including genetic engineering and metabolic reprogramming. An integrated approach, combining metabolic interventions with CAR T-cell therapy, has the potential to overcome these constraints and improve therapeutic outcomes in solid tumors.
Collapse
Affiliation(s)
- Rishab Ramapriyan
- Brain Tumor Immunotherapy Laboratory, Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA.
| | - Vivasvan S Vykunta
- Department of Pathology, University of California, San Francisco, San Francisco, CA 94143, USA; ImmunoX Initiative, University of California, San Francisco, San Francisco, CA 94143, USA; Medical Scientist Training Program, School of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA; Biomedical Sciences Graduate Program, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Gust Vandecandelaere
- Brain Tumor Immunotherapy Laboratory, Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Leland G K Richardson
- Brain Tumor Immunotherapy Laboratory, Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Jing Sun
- Brain Tumor Immunotherapy Laboratory, Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - William T Curry
- Brain Tumor Immunotherapy Laboratory, Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Bryan D Choi
- Brain Tumor Immunotherapy Laboratory, Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA.
| |
Collapse
|
15
|
Moraly J, Kondo T, Benzaoui M, DuSold J, Talluri S, Pouzolles MC, Chien C, Dardalhon V, Taylor N. Metabolic dialogues: regulators of chimeric antigen receptor T cell function in the tumor microenvironment. Mol Oncol 2024; 18:1695-1718. [PMID: 38922759 PMCID: PMC11223614 DOI: 10.1002/1878-0261.13691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 02/23/2024] [Accepted: 06/11/2024] [Indexed: 06/28/2024] Open
Abstract
Tumor-infiltrating lymphocytes (TILs) and chimeric antigen receptor (CAR) T cells have demonstrated remarkable success in the treatment of relapsed/refractory melanoma and hematological malignancies, respectively. These treatments have marked a pivotal shift in cancer management. However, as "living drugs," their effectiveness is dependent on their ability to proliferate and persist in patients. Recent studies indicate that the mechanisms regulating these crucial functions, as well as the T cell's differentiation state, are conditioned by metabolic shifts and the distinct utilization of metabolic pathways. These metabolic shifts, conditioned by nutrient availability as well as cell surface expression of metabolite transporters, are coupled to signaling pathways and the epigenetic landscape of the cell, modulating transcriptional, translational, and post-translational profiles. In this review, we discuss the processes underlying the metabolic remodeling of activated T cells, the impact of a tumor metabolic environment on T cell function, and potential metabolic-based strategies to enhance T cell immunotherapy.
Collapse
Affiliation(s)
- Josquin Moraly
- Pediatric Oncology Branch, National Cancer InstituteNational Institutes of HealthBethesdaMDUSA
- Université Sorbonne Paris CitéParisFrance
| | - Taisuke Kondo
- Pediatric Oncology Branch, National Cancer InstituteNational Institutes of HealthBethesdaMDUSA
| | - Mehdi Benzaoui
- Pediatric Oncology Branch, National Cancer InstituteNational Institutes of HealthBethesdaMDUSA
- Université de Montpellier, Institut de Génétique Moléculaire de Montpellier, CNRSMontpellierFrance
| | - Justyn DuSold
- Pediatric Oncology Branch, National Cancer InstituteNational Institutes of HealthBethesdaMDUSA
| | - Sohan Talluri
- Pediatric Oncology Branch, National Cancer InstituteNational Institutes of HealthBethesdaMDUSA
| | - Marie C. Pouzolles
- Pediatric Oncology Branch, National Cancer InstituteNational Institutes of HealthBethesdaMDUSA
| | - Christopher Chien
- Pediatric Oncology Branch, National Cancer InstituteNational Institutes of HealthBethesdaMDUSA
| | - Valérie Dardalhon
- Université de Montpellier, Institut de Génétique Moléculaire de Montpellier, CNRSMontpellierFrance
| | - Naomi Taylor
- Pediatric Oncology Branch, National Cancer InstituteNational Institutes of HealthBethesdaMDUSA
- Université de Montpellier, Institut de Génétique Moléculaire de Montpellier, CNRSMontpellierFrance
| |
Collapse
|
16
|
Jia M, Yuan Z, Yu H, Feng S, Tan X, Long Z, Duan Y, Zhu W, Yan P. Rapamycin circumvents anti PD-1 therapy resistance in colorectal cancer by reducing PD-L1 expression and optimizing the tumor microenvironment. Biomed Pharmacother 2024; 176:116883. [PMID: 38876047 DOI: 10.1016/j.biopha.2024.116883] [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: 02/20/2024] [Revised: 05/23/2024] [Accepted: 06/03/2024] [Indexed: 06/16/2024] Open
Abstract
The unresectable or postoperative recurrence of advanced metastatic colorectal cancer (CRC) is the difficulty of its clinical management, and pharmacological therapy is the main source of benefit. Immune checkpoint inhibitors are therapeutic options but are effective in approximately 5 % of patients with deficient mismatch repair (MMR)/microsatellite instability CRC and are ineffective in patients with MMR-proficient (pMMR)/microsatellite stable (MSS) CRCs, which may be associated with the tumor microenvironment (TME). Here, we propose a new combination strategy and evaluate the efficacy of rapamycin (Rapa) combined with anti-PD-1 (αPD-1) in CT26 tumor-bearing mice, azoxymethane (AOM)/dextran sodium sulfate (DSS) inflammation-associated CRC mice, CT26-Luc tumor-bearing mice with postoperative recurrence, and CT26 liver metastasis mice. The results revealed that Rapa improved the therapeutic effect of αPD-1 and effectively inhibited colorectal carcinogenesis, postoperative recurrence, and liver metastasis. Mechanistically, Rapa improved the anticancer effect of αPD-1, associated with Rapa reprograming of the immunosuppressive TME. Rapa effectively depleted α-SMA+ cancer-associated fibroblasts and degraded collagen in the tumor tissue, increasing T lymphocyte infiltration into the tumor tissue. Rapa induced the downregulation of programed cell death 1 ligand 1 (PD-L1) protein and transcript levels in CT26 cells, which may be associated with the inhibition of the mTOR/P70S6K signaling axis. Furthermore, co-culture of tumor cells and CD8+ T lymphocytes demonstrated that Rapa-induced PD-L1 downregulation in tumor cells increased spleen-derived CD8+ T lymphocyte activation. Therefore, Rapa improves the anti-tumor effect of αPD-1 in CRCs, providing new ideas for its use to improve combinatorial strategies for anti-PD-1 immunotherapy.
Collapse
Affiliation(s)
- Menglei Jia
- Department of Pharmacy, Biomedicine Research Center, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou 510150, China
| | - Zhongwen Yuan
- Department of Pharmacy, Biomedicine Research Center, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou 510150, China
| | - Hang Yu
- Department of Pharmacy, Biomedicine Research Center, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou 510150, China
| | - Senling Feng
- Department of Pharmacy, Biomedicine Research Center, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou 510150, China
| | - Xiaoxiao Tan
- Department of Pharmacy, Biomedicine Research Center, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou 510150, China
| | - Zijing Long
- Department of Pharmacy, Biomedicine Research Center, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou 510150, China
| | - Yanrong Duan
- Department of Pharmacy, Biomedicine Research Center, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou 510150, China
| | - Wenting Zhu
- Department of Pharmacy, Biomedicine Research Center, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou 510150, China.
| | - Pengke Yan
- Department of Pharmacy, Biomedicine Research Center, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou 510150, China.
| |
Collapse
|
17
|
Qin Z, Hou P, Lin H, Chen M, Wang R, Xu T. Inhibition of Lck/Fyn kinase activity promotes the differentiation of induced Treg cells through AKT/mTOR pathway. Int Immunopharmacol 2024; 134:112237. [PMID: 38744170 DOI: 10.1016/j.intimp.2024.112237] [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: 02/28/2024] [Revised: 05/06/2024] [Accepted: 05/07/2024] [Indexed: 05/16/2024]
Abstract
Regulatory T (Treg) cells are indispensable in maintaining the immune homeostasis and preventing autoimmune diseases. Regulatory T (Treg) cells include thymus derived Treg cells (tTregs) and peripherally induced Treg cells (iTreg), which are differentiated from antigen stimulated CD4+ naïve T cells in presence of TGFβ. tTregs are quite stable, and more immune suppressive, while iTreg cells are less stable, and are prone to differentiate into inflammatory T cells. Therefore, identification of small molecules that could promote the differentiation of iTreg cells is an attractive strategy for autoimmune diseases. Inhibition of AKT/mTOR pathway promotes their differentiation. Whether inhibition of Lck/Fyn kinase activity (upstream of AKT/mTOR pathway) can be used to promote the differentiation of iTreg cells has not been determined. Here, we showed that Srci1, a small molecular inhibitor of Lck/Fyn, promoted the differentiation of FOXP3+ iTreg cells. Srci1 treatment resulted in inhibition of phosphorylation of key components of AKT/mTOR pathway, including mTOR, p70 S6K, 4EBP1, and promoted the expression of Foxp3 and its target genes, thereby promoted differentiation of in vitro iTreg cells. Srci1 treated iTreg cells showed more similar gene expression profile to that of tTreg cells. Our results thus suggest that inhibition of Lck/Fyn kinase activity can promote the differentiation of iTreg cells, and may have implication in autoimmune diseases.
Collapse
Affiliation(s)
- Zhen Qin
- Department of Immunology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Ping Hou
- Department of Immunology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Huizhen Lin
- Department of Immunology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Minghui Chen
- Department of Immunology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Ruining Wang
- Department of Immunology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Tao Xu
- Department of Immunology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China.
| |
Collapse
|
18
|
Santagata S, Rea G, Bello AM, Capiluongo A, Napolitano M, Desicato S, Fragale A, D'Alterio C, Trotta AM, Ieranò C, Portella L, Persico F, Di Napoli M, Di Maro S, Feroce F, Azzaro R, Gabriele L, Longo N, Pignata S, Perdonà S, Scala S. Targeting CXCR4 impaired T regulatory function through PTEN in renal cancer patients. Br J Cancer 2024; 130:2016-2026. [PMID: 38704478 PMCID: PMC11183124 DOI: 10.1038/s41416-024-02702-x] [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: 09/20/2023] [Revised: 04/12/2024] [Accepted: 04/19/2024] [Indexed: 05/06/2024] Open
Abstract
BACKGROUND Tregs trafficking is controlled by CXCR4. In Renal Cell Carcinoma (RCC), the effect of the new CXCR4 antagonist, R54, was explored in peripheral blood (PB)-Tregs isolated from primary RCC patients. METHODS PB-Tregs were isolated from 77 RCC patients and 38 healthy donors (HDs). CFSE-T effector-Tregs suppression assay, IL-35, IFN-γ, IL-10, TGF-β1 secretion, and Nrp-1+Tregs frequency were evaluated. Tregs were characterised for CTLA-4, PD-1, CD40L, PTEN, CD25, TGF-β1, FOXP3, DNMT1 transcriptional profile. PTEN-pAKT signalling was evaluated in the presence of R54 and/or triciribine (TCB), an AKT inhibitor. Methylation of TSDR (Treg-Specific-Demethylated-Region) was conducted. RESULTS R54 impaired PB-RCC-Tregs function, reduced Nrp-1+Tregs frequency, the release of IL-35, IL-10, and TGF-β1, while increased IFN-γ Teff-secretion. The CXCR4 ligand, CXCL12, recruited CD25+PTEN+Tregs in RCC while R54 significantly reduced it. IL-2/PMA activates Tregs reducing pAKT+Tregs while R54 increases it. The AKT inhibitor, TCB, prevented the increase in pAKT+Tregs R54-mediated. Moreover, R54 significantly reduced FOXP3-TSDR demethylation with DNMT1 and FOXP3 downregulation. CONCLUSION R54 impairs Tregs function in primary RCC patients targeting PTEN/PI3K/AKT pathway, reducing TSDR demethylation and FOXP3 and DNMT1 expression. Thus, CXCR4 targeting is a strategy to inhibit Tregs activity in the RCC tumour microenvironment.
Collapse
Affiliation(s)
- Sara Santagata
- Microenvironment Molecular Targets, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131, Naples, Italy
| | - Giuseppina Rea
- Microenvironment Molecular Targets, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131, Naples, Italy
| | - Anna Maria Bello
- Microenvironment Molecular Targets, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131, Naples, Italy
| | - Anna Capiluongo
- Microenvironment Molecular Targets, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131, Naples, Italy
| | - Maria Napolitano
- Microenvironment Molecular Targets, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131, Naples, Italy
| | - Sonia Desicato
- Urology, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131, Naples, Italy
| | - Alessandra Fragale
- Department of Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161, Rome, Italy
| | - Crescenzo D'Alterio
- Microenvironment Molecular Targets, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131, Naples, Italy
| | - Anna Maria Trotta
- Microenvironment Molecular Targets, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131, Naples, Italy
| | - Caterina Ieranò
- Microenvironment Molecular Targets, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131, Naples, Italy
| | - Luigi Portella
- Microenvironment Molecular Targets, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131, Naples, Italy
| | - Francesco Persico
- Department of Neurosciences, Reproductive Sciences and Odontostomatology, Urology Unit, University of Naples "Federico II", 80138, Napoli, Italy
| | - Marilena Di Napoli
- Uro-gynecological Oncology, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131, Naples, Italy
| | - Salvatore Di Maro
- Department of Environmental, Biological and Pharmaceutical Science and Technology, University of Campania "Luigi Vanvitelli", 81100, Caserta, Italy
| | - Florinda Feroce
- Pathology, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131, Naples, Italy
| | - Rosa Azzaro
- Transfusion Medicine Unit, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131, Naples, Italy
| | - Lucia Gabriele
- Department of Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161, Rome, Italy
| | - Nicola Longo
- Department of Neurosciences, Reproductive Sciences and Odontostomatology, Urology Unit, University of Naples "Federico II", 80138, Napoli, Italy
| | - Sandro Pignata
- Uro-gynecological Oncology, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131, Naples, Italy
| | - Sisto Perdonà
- Urology, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131, Naples, Italy
| | - Stefania Scala
- Microenvironment Molecular Targets, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131, Naples, Italy.
| |
Collapse
|
19
|
Joo V, Abdelhamid K, Noto A, Latifyan S, Martina F, Daoudlarian D, De Micheli R, Pruijm M, Peters S, Hullin R, Gaide O, Pantaleo G, Obeid M. Primary prophylaxis with mTOR inhibitor enhances T cell effector function and prevents heart transplant rejection during talimogene laherparepvec therapy of squamous cell carcinoma. Nat Commun 2024; 15:3664. [PMID: 38693123 PMCID: PMC11063183 DOI: 10.1038/s41467-024-47965-3] [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: 08/03/2023] [Accepted: 04/15/2024] [Indexed: 05/03/2024] Open
Abstract
The application of mammalian target of rapamycin inhibition (mTORi) as primary prophylactic therapy to optimize T cell effector function while preserving allograft tolerance remains challenging. Here, we present a comprehensive two-step therapeutic approach in a male patient with metastatic cutaneous squamous cell carcinoma and heart transplantation followed with concomitant longitudinal analysis of systemic immunologic changes. In the first step, calcineurin inhibitor/ mycophenolic acid is replaced by the mTORi everolimus to achieve an improved effector T cell status with increased cytotoxic activity (perforin, granzyme), enhanced proliferation (Ki67) and upregulated activation markers (CD38, CD69). In the second step, talimogene laherparepvec (T-VEC) injection further enhances effector function by switching CD4 and CD8 cells from central memory to effector memory profiles, enhancing Th1 responses, and boosting cytotoxic and proliferative activities. In addition, cytokine release (IL-6, IL-18, sCD25, CCL-2, CCL-4) is enhanced and the frequency of circulating regulatory T cells is increased. Notably, no histologic signs of allograft rejection are observed in consecutive end-myocardial biopsies. These findings provide valuable insights into the dynamics of T cell activation and differentiation and suggest that timely initiation of mTORi-based primary prophylaxis may provide a dual benefit of revitalizing T cell function while maintaining allograft tolerance.
Collapse
Affiliation(s)
- Victor Joo
- Centre Hospitalier Universitaire Vaudois (CHUV), University of Lausanne, Department of Medicine, Immunology and Allergy Division, Rue du Bugnon 46, CH-1011, Lausanne, Switzerland
| | - Karim Abdelhamid
- Centre Hospitalier Universitaire Vaudois (CHUV), University of Lausanne, Oncology Department, Rue du Bugnon 46, CH-1011, Lausanne, Switzerland
| | - Alessandra Noto
- Centre Hospitalier Universitaire Vaudois (CHUV), University of Lausanne, Department of Medicine, Immunology and Allergy Division, Rue du Bugnon 46, CH-1011, Lausanne, Switzerland
| | - Sofiya Latifyan
- Centre Hospitalier Universitaire Vaudois (CHUV), University of Lausanne, Oncology Department, Rue du Bugnon 46, CH-1011, Lausanne, Switzerland
| | - Federica Martina
- Centre Hospitalier Universitaire Vaudois (CHUV), University of Lausanne, Department of Medicine, Immunology and Allergy Division, Rue du Bugnon 46, CH-1011, Lausanne, Switzerland
| | - Douglas Daoudlarian
- Centre Hospitalier Universitaire Vaudois (CHUV), University of Lausanne, Department of Medicine, Immunology and Allergy Division, Rue du Bugnon 46, CH-1011, Lausanne, Switzerland
| | - Rita De Micheli
- Centre Hospitalier Universitaire Vaudois (CHUV), University of Lausanne, Oncology Department, Rue du Bugnon 46, CH-1011, Lausanne, Switzerland
| | - Menno Pruijm
- Centre Hospitalier Universitaire Vaudois (CHUV), University of Lausanne, Department of Medicine, Nephrology Division, Rue du Bugnon 17, CH-1011, Lausanne, Switzerland
| | - Solange Peters
- Centre Hospitalier Universitaire Vaudois (CHUV), University of Lausanne, Oncology Department, Rue du Bugnon 46, CH-1011, Lausanne, Switzerland
| | - Roger Hullin
- Centre Hospitalier Universitaire Vaudois (CHUV), University of Lausanne, Cardiology, Cardiovascular Department, Rue du Bugnon 46, CH-1011, Lausanne, Switzerland
| | - Olivier Gaide
- Centre Hospitalier Universitaire Vaudois (CHUV), University of Lausanne, Dermatology Division, Rue du Bugnon 46, CH-1011, Lausanne, Switzerland
| | - Giuseppe Pantaleo
- Centre Hospitalier Universitaire Vaudois (CHUV), University of Lausanne, Department of Medicine, Immunology and Allergy Division, Rue du Bugnon 46, CH-1011, Lausanne, Switzerland
| | - Michel Obeid
- Centre Hospitalier Universitaire Vaudois (CHUV), University of Lausanne, Department of Medicine, Immunology and Allergy Division, Rue du Bugnon 46, CH-1011, Lausanne, Switzerland.
| |
Collapse
|
20
|
Sekiya T, Hidano S, Takaki S. Tonic TCR and IL-1β signaling mediate phenotypic alterations of naive CD4 + T cells. Cell Rep 2024; 43:113954. [PMID: 38492221 DOI: 10.1016/j.celrep.2024.113954] [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: 05/30/2023] [Revised: 11/06/2023] [Accepted: 02/28/2024] [Indexed: 03/18/2024] Open
Abstract
Inert naive CD4+ T (TN) cells differentiate into functional T helper (Th) or regulatory T (Treg) cell subsets upon encountering antigens, mediating properly directed immune responses. Although all TN cells can differentiate into any of the Th and Treg cell subsets, heterogeneity exists among TN cells. By constructing reporter mice to detect ongoing T cell receptor (TCR) signaling, we identify that interleukin (IL)-1β signaling affects TN cell characteristics, independent of tonic TCR signaling, which also alters TN cell phenotypes. IL-1β reversibly attenuates the differentiation potential of TN cells toward Treg cells. IL-1β signaling is elevated in the splenic TN cells, consequently attenuating their differentiation potential toward Treg cells. Aberrant elevation of IL-1β signaling augments colitogenic activities of TN cells. TN cells in patients with colitis exhibited elevated IL-1β signaling. We demonstrate that phenotypic alteration in TN cells by IL-1β is an important mechanism in the regulation of immune responses.
Collapse
Affiliation(s)
- Takashi Sekiya
- Section of Immune Response Modification, The Research Center for Hepatitis and Immunology, National Center for Global Health and Medicine, 1-7-1 Kohnodai, Ichikawa, Chiba 272-8516, Japan; Department of Immune Regulation, The Research Center for Hepatitis and Immunology, National Center for Global Health and Medicine, 1-7-1 Kohnodai, Ichikawa, Chiba 272-8516, Japan.
| | - Shinya Hidano
- Department of Immune Regulation, The Research Center for Hepatitis and Immunology, National Center for Global Health and Medicine, 1-7-1 Kohnodai, Ichikawa, Chiba 272-8516, Japan
| | - Satoshi Takaki
- Department of Immune Regulation, The Research Center for Hepatitis and Immunology, National Center for Global Health and Medicine, 1-7-1 Kohnodai, Ichikawa, Chiba 272-8516, Japan
| |
Collapse
|
21
|
White TLA, Jin Y, Roberts SDA, Gable MJ, Morel PA. Phosphorylation of hnRNP A1-Serine 199 Is Not Required for T Cell Differentiation and Function. Immunohorizons 2024; 8:136-146. [PMID: 38334757 PMCID: PMC10916359 DOI: 10.4049/immunohorizons.2300074] [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: 09/11/2023] [Accepted: 01/05/2024] [Indexed: 02/10/2024] Open
Abstract
hnRNP A1 is an important RNA-binding protein that influences many stages of RNA processing, including transcription, alternative splicing, mRNA nuclear export, and RNA stability. However, the role of hnRNP A1 in immune cells, specifically CD4+ T cells, remains unclear. We previously showed that Akt phosphorylation of hnRNP A1 was dependent on TCR signal strength and was associated with Treg differentiation. To explore the impact of hnRNP A1 phosphorylation by Akt on CD4+ T cell differentiation, our laboratory generated a mutant mouse model, hnRNP A1-S199A (A1-MUT) in which the major Akt phosphorylation site on hnRNP A1 was mutated to alanine using CRISPR Cas9 technology. Immune profiling of A1-MUT mice revealed changes in the numbers of Tregs in the mesenteric lymph node. We found no significant differences in naive CD4+ T cell differentiation into Th1, Th2, Th17, or T regulatory cells (Tregs) in vitro. In vivo, Treg differentiation assays using OTII-A1-Mut CD4+ T cells exposed to OVA food revealed migration and homing defects in the A1-MUT but no change in Treg induction. A1-MUT mice were immunized with NP- keyhole limpet hemocyanin, and normal germinal center development, normal numbers of NP-specific B cells, and no change in Tfh numbers were observed. In conclusion, Akt phosphorylation of hnRNP A1 S199 does not play a role in CD4+ T cell fate or function in the models tested. This hnRNP A1-S199A mouse model should be a valuable tool to study the role of Akt phosphorylation of hnRNP A1-S199 in different cell types or other mouse models of human disease.
Collapse
Affiliation(s)
- Tristan L. A. White
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Ye Jin
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Sean D. A. Roberts
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Matthew J. Gable
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Penelope A. Morel
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA
| |
Collapse
|
22
|
Lee J, Park N, Nicosia M, Park JY, Pruett SB, Seo KS. Stimulation Strength Determined by Superantigen Dose Controls Subcellular Localization of FOXP3 Isoforms and Suppressive Function of CD4+CD25+FOXP3+ T Cells. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 212:421-432. [PMID: 38108423 PMCID: PMC10784726 DOI: 10.4049/jimmunol.2300019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Accepted: 11/20/2023] [Indexed: 12/19/2023]
Abstract
Staphylococcal superantigens induce massive activation of T cells and inflammation, leading to toxic shock syndrome. Paradoxically, increasing evidence indicates that superantigens can also induce immunosuppression by promoting regulatory T cell (Treg) development. In this study, we demonstrate that stimulation strength plays a critical role in superantigen-mediated induction of immunosuppressive human CD4+CD25+FOXP3+ T cells. Suboptimal stimulation by a low dose (1 ng/ml) of staphylococcal enterotoxin C1 (SEC1) led to de novo generation of Treg-like CD4+CD25+FOXP3+ T cells with strong suppressive activity. In contrast, CD4+CD25+ T cells induced by optimal stimulation with high-dose SEC1 (1 µg/ml) were not immunosuppressive, despite high FOXP3 expression. Signal transduction pathway analysis revealed differential activation of the PI3K signaling pathway and expression of PTEN in optimal and suboptimal stimulation with SEC1. Additionally, we identified that FOXP3 isoforms in Treg-like cells from the suboptimal condition were located in the nucleus, whereas FOXP3 in nonsuppressive cells from the optimal condition localized in cytoplasm. Sequencing analysis of FOXP3 isoform transcripts identified five isoforms, including a FOXP3 isoform lacking partial exon 3. Overexpression of FOXP3 isoforms confirmed that both an exon 2-lacking isoform and a partial exon 3-lacking isoform confer suppressive activity. Furthermore, blockade of PI3K in optimal stimulation conditions led to induction of suppressive Treg-like cells with nuclear translocation of FOXP3, suggesting that PI3K signaling impairs induction of Tregs in a SEC1 dose-dependent manner. Taken together, these data demonstrate that the strength of activation signals determined by superantigen dose regulates subcellular localization of FOXP3 isoforms, which confers suppressive functionality.
Collapse
Affiliation(s)
- Juyeun Lee
- Department of Comparative Biomedical Sciences, College of Veterinary Medicine, Mississippi State University, Mississippi State, MS
| | - Nogi Park
- Department of Comparative Biomedical Sciences, College of Veterinary Medicine, Mississippi State University, Mississippi State, MS
| | - Michael Nicosia
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH
| | - Joo Youn Park
- Department of Comparative Biomedical Sciences, College of Veterinary Medicine, Mississippi State University, Mississippi State, MS
| | - Stephen B. Pruett
- Department of Comparative Biomedical Sciences, College of Veterinary Medicine, Mississippi State University, Mississippi State, MS
| | - Keun Seok Seo
- Department of Comparative Biomedical Sciences, College of Veterinary Medicine, Mississippi State University, Mississippi State, MS
| |
Collapse
|
23
|
Zuo Z, Zhou Z, Chang Y, Liu Y, Shen Y, Li Q, Zhang L. Ribonucleotide reductase M2 (RRM2): Regulation, function and targeting strategy in human cancer. Genes Dis 2024; 11:218-233. [PMID: 37588202 PMCID: PMC10425756 DOI: 10.1016/j.gendis.2022.11.022] [Citation(s) in RCA: 44] [Impact Index Per Article: 44.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 10/26/2022] [Accepted: 11/14/2022] [Indexed: 12/29/2022] Open
Abstract
Ribonucleotide reductase M2 (RRM2) is a small subunit in ribonucleotide reductases, which participate in nucleotide metabolism and catalyze the conversion of nucleotides to deoxynucleotides, maintaining the dNTP pools for DNA biosynthesis, repair, and replication. RRM2 performs a critical role in the malignant biological behaviors of cancers. The structure, regulation, and function of RRM2 and its inhibitors were discussed. RRM2 gene can produce two transcripts encoding the same ORF. RRM2 expression is regulated at multiple levels during the processes from transcription to translation. Moreover, this gene is associated with resistance, regulated cell death, and tumor immunity. In order to develop and design inhibitors of RRM2, appropriate strategies can be adopted based on different mechanisms. Thus, a greater appreciation of the characteristics of RRM2 is a benefit for understanding tumorigenesis, resistance in cancer, and tumor microenvironment. Moreover, RRM2-targeted therapy will be more attention in future therapeutic approaches for enhancement of treatment effects and amelioration of the dismal prognosis.
Collapse
Affiliation(s)
- Zanwen Zuo
- Innovative Drug R&D Center, College of Life Sciences, Huaibei Normal University, Huaibei, Anhui 235000, China
- National “111” Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei Key Laboratory of Industrial Microbiology, Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), and School of Food and Biological Engineering, Hubei University of Technology, Wuhan, Hubei 430068, China
| | - Zerong Zhou
- Innovative Drug R&D Center, College of Life Sciences, Huaibei Normal University, Huaibei, Anhui 235000, China
- National “111” Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei Key Laboratory of Industrial Microbiology, Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), and School of Food and Biological Engineering, Hubei University of Technology, Wuhan, Hubei 430068, China
| | - Yuzhou Chang
- Department of Biomedical Informatics, The Ohio State University, Columbus, OH 43210, USA
| | - Yan Liu
- School of Agriculture and Biology, and Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Yuping Shen
- College of Chemistry and Bioengineering, Hunan University of Science and Engineering, Yongzhou, Hunan 425199, China
| | - Qizhang Li
- Innovative Drug R&D Center, College of Life Sciences, Huaibei Normal University, Huaibei, Anhui 235000, China
- National “111” Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei Key Laboratory of Industrial Microbiology, Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), and School of Food and Biological Engineering, Hubei University of Technology, Wuhan, Hubei 430068, China
| | - Lei Zhang
- Innovative Drug R&D Center, College of Life Sciences, Huaibei Normal University, Huaibei, Anhui 235000, China
- Department of Pharmaceutical Botany, School of Pharmacy, Naval Medical University, Shanghai 200433, China
| |
Collapse
|
24
|
Asghari F, Karimi MH, Pourfathollah AA. mTORC1 inhibition may improve T lymphocytes affected by aging. Immunopharmacol Immunotoxicol 2023; 45:719-729. [PMID: 37581412 DOI: 10.1080/08923973.2023.2232101] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Accepted: 06/23/2023] [Indexed: 08/16/2023]
Abstract
BACKGROUND Due to the increase of the elderly's population and related social and economic problems, it is very important to provide strategies on health. In this regard, induction of T lymphocytes responses, the most important cells of the immune system, may be a good approach. Among different agents considered as antiaging factors, mTORC1 pathway inhibitors are significant. So, the purpose of this study was to evaluate the effect of two mTORC1 inhibitors, Everolimus and Metformin, on age-related features of activated T cells. MATERIALS AND METHODS Optimum doses of drugs was determined with evaluating the effect of treatments on IL-2 gene expression. T cells isolated from old and young mice were treated with drugs and PHA. IL-2 production was evaluated by ELISA. Also, the expression of CD28, PD-1, and KLRG-1, proliferation, and intracellular oxidative stress were assessed by flow cytometry-based assays, phenotyping, CFSE, and DCF-DA assay respectively. RESULTS Both drugs increased IL-2 production in the T cells of old mice. Also, using drugs especially Metformin could improve age-related phenotypical markers and increase the proliferation of T cells of old mice significantly. In addition, Metformin and Everolimus reduced intracellular oxidative stress in aged cells. However, the effect of both drugs on the T cells of young mice wasn't significant or was in opposite to the results of old mice T cells. DISCUSSION In line with studies noting mTOR inhibitors as antiaging drugs, Metformin and Everolimus may improve T cells affected from aging in vitro, and a decrease in intracellular oxidative stress may be one of their mechanism of function.
Collapse
Affiliation(s)
- F Asghari
- Department of Immunology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - M H Karimi
- Larestan University of Medical Sciences, Larestan, Iran
- Transplant Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - A A Pourfathollah
- Department of Immunology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| |
Collapse
|
25
|
Cui X, Li CG, Gao H, Cheng M, Jiang F. Boosting regulatory T cell-dependent immune tolerance by activation of p53. Int Immunopharmacol 2023; 125:111167. [PMID: 37931392 DOI: 10.1016/j.intimp.2023.111167] [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: 08/20/2023] [Revised: 10/13/2023] [Accepted: 10/31/2023] [Indexed: 11/08/2023]
Abstract
Regulatory T cells (Tregs) have critical roles in maintaining immune hemostasis and have important anti-inflammatory functions in diseases. Recently, we identified that CX-5461 (a selective RNA polymerase I inhibitor and p53 activator) acted as a potent immunosuppressive agent, which prevented allogeneic acute rejection in animal models via a molecular mechanism distinct from all those of conventional immunosuppressive drugs. Unexpectedly, we discovered that CX-5461 could promote Treg differentiation. In this review, we have summarized the evidence for a potential role of p53 in mediating Treg differentiation and its possible mechanisms, including regulation of FoxP3 transcription, regulation of the expression of PTEN (phosphatase and tensin homolog), as well as protein-protein interaction with the transcription factor STAT5 (signal transducer and activator of transcription 5). Evidence also suggests that pharmacological p53 activators may potentially be used to boost Treg-mediated immune tolerance. Based on these data, we argue that novel p53 activators such as CX-5461 may represent a distinct class of immunosuppressants that repress conventional T cell-mediated alloimmunity with concomitant boosting of Treg-dependent immune tolerance.
Collapse
Affiliation(s)
- Xiaopei Cui
- Shandong Key Laboratory of Cardiovascular Proteomics and Department of Geriatric Medicine, Qilu Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province, China
| | - Chun-Guang Li
- NICM Health Research Institute, Western Sydney University, Westmead, NSW 2145, Australia
| | - Haiqing Gao
- Shandong Key Laboratory of Cardiovascular Proteomics and Department of Geriatric Medicine, Qilu Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province, China
| | - Mei Cheng
- Shandong Key Laboratory of Cardiovascular Proteomics and Department of Geriatric Medicine, Qilu Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province, China.
| | - Fan Jiang
- Shandong Key Laboratory of Cardiovascular Proteomics and Department of Geriatric Medicine, Qilu Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province, China.
| |
Collapse
|
26
|
Perpiñán E, Sanchez-Fueyo A, Safinia N. Immunoregulation: the interplay between metabolism and redox homeostasis. FRONTIERS IN TRANSPLANTATION 2023; 2:1283275. [PMID: 38993920 PMCID: PMC11235320 DOI: 10.3389/frtra.2023.1283275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Accepted: 11/13/2023] [Indexed: 07/13/2024]
Abstract
Regulatory T cells are fundamental for the induction and maintenance of immune homeostasis, with their dysfunction resulting in uncontrolled immune responses and tissue destruction predisposing to autoimmunity, transplant rejection and several inflammatory and metabolic disorders. Recent discoveries have demonstrated that metabolic processes and mitochondrial function are critical for the appropriate functioning of these cells in health, with their metabolic adaptation, influenced by microenvironmental factors, seen in several pathological processes. Upon activation regulatory T cells rearrange their oxidation-reduction (redox) system, which in turn supports their metabolic reprogramming, adding a layer of complexity to our understanding of cellular metabolism. Here we review the literature surrounding redox homeostasis and metabolism of regulatory T cells to highlight new mechanistic insights of these interlinked pathways in immune regulation.
Collapse
Affiliation(s)
| | | | - N. Safinia
- Department of Inflammation Biology, School of Immunology and Microbial Sciences, Institute of Liver Studies, James Black Centre, King’s College London, London, United Kingdom
| |
Collapse
|
27
|
Olajuyin AM, Olajuyin AK, Zhang X, Hu Q. Immunomodulatory macrophages and Treg in pulmonary hypertension. COMPARATIVE CLINICAL PATHOLOGY 2023; 33:163-173. [DOI: 10.1007/s00580-023-03540-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Accepted: 11/20/2023] [Indexed: 01/03/2025]
|
28
|
Zhang Y, Zhang D, Chen L, Zhou J, Ren B, Chen H. The progress of autoimmune hepatitis research and future challenges. Open Med (Wars) 2023; 18:20230823. [PMID: 38025543 PMCID: PMC10655690 DOI: 10.1515/med-2023-0823] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 09/24/2023] [Accepted: 09/28/2023] [Indexed: 12/01/2023] Open
Abstract
Autoimmune hepatitis (AIH) is a chronic liver inflammatory disease with various immune system manifestations, showing a global trend of increased prevalence. AIH is diagnosed through histological abnormalities, clinical manifestations, and biochemical indicators. The biochemical markers involve interfacial hepatitis, transaminase abnormalities, positive autoantibodies, etc. Although AIH pathogenesis is unclear, gene mutations and immunological factors could be the leading factors. AIH usually presents as a chronic liver disease and sometimes as acute hepatitis, making it challenging to distinguish it from drug-related hepatitis due to similar clinical symptoms. Normalizing transaminases and serum IgG levels is essential in assessing the remission status of AIH treatment. Glucocorticoids and azathioprine are the first-line AIH treatment, with lifelong maintenance therapy in some patients. The quality of life and survival can be improved after appropriate treatment. However, certain limitations jeopardize the quality of treatment, including long treatment cycles, side effects, poor patient compliance, and inability to inhibit liver fibrosis and cirrhosis. Accurate AIH animal models will help us understand the pathophysiology of the disease while providing fresh perspectives for avoiding and treating AIH. This review will help us understand AIH better, from the cellular and molecular causes to the clinical features, and will provide insight into new therapy techniques with fewer side effects.
Collapse
Affiliation(s)
- Yang Zhang
- Graduate Department of Zhejiang Chinese Medicine University, Hangzhou, Zhejiang, China
- Department of Infectious Diseases, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China
| | - Dehe Zhang
- Department of Infectious Diseases, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China
| | - Ling Chen
- Department of Infectious Diseases, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China
| | - Jing Zhou
- Department of Infectious Diseases, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China
| | - Binbin Ren
- Department of Infectious Diseases, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China
| | - Haijun Chen
- Department of Infectious Diseases, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China
| |
Collapse
|
29
|
Luo Z, Zhang Y, Saleh QW, Zhang J, Zhu Z, Tepel M. Metabolic regulation of forkhead box P3 alternative splicing isoforms and their impact on health and disease. Front Immunol 2023; 14:1278560. [PMID: 37868998 PMCID: PMC10588449 DOI: 10.3389/fimmu.2023.1278560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Accepted: 09/25/2023] [Indexed: 10/24/2023] Open
Abstract
Forkhead Box P3 (FOXP3) is crucial for the development and suppressive function of human regulatory T cells (Tregs). There are two predominant FOXP3 splicing isoforms in healthy humans, the full-length isoform and the isoform lacking exon 2, with different functions and regulation mechanisms. FOXP3 splicing isoforms show distinct abilities in the cofactor interaction and the nuclear translocation, resulting in different effects on the differentiation, cytokine secretion, suppressive function, linage stability, and environmental adaptation of Tregs. The balance of FOXP3 splicing isoforms is related to autoimmune diseases, inflammatory diseases, and cancers. In response to environmental challenges, FOXP3 transcription and splicing can be finely regulated by T cell antigen receptor stimulation, glycolysis, fatty acid oxidation, and reactive oxygen species, with various signaling pathways involved. Strategies targeting energy metabolism and FOXP3 splicing isoforms in Tregs may provide potential new approaches for the treatment of autoimmune diseases, inflammatory diseases, and cancers. In this review, we summarize recent discoveries about the FOXP3 splicing isoforms and address the metabolic regulation and specific functions of FOXP3 splicing isoforms in Tregs.
Collapse
Affiliation(s)
- Zhidan Luo
- Department of Geriatrics, Chongqing General Hospital, Chongqing, China
- Cardiovascular and Renal Research, Department of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Yihua Zhang
- Department of Cardiology, Chongqing Fifth People’s Hospital, Chongqing, China
| | - Qais Waleed Saleh
- Cardiovascular and Renal Research, Department of Molecular Medicine, University of Southern Denmark, Odense, Denmark
- Department of Nephrology, Odense University Hospital, Odense, Denmark
| | - Jie Zhang
- Department of Geriatrics, Chongqing General Hospital, Chongqing, China
| | - Zhiming Zhu
- Department of Hypertension and Endocrinology, Daping Hospital, Chongqing, China
| | - Martin Tepel
- Cardiovascular and Renal Research, Department of Molecular Medicine, University of Southern Denmark, Odense, Denmark
- Department of Nephrology, Odense University Hospital, Odense, Denmark
| |
Collapse
|
30
|
Larange A, Takazawa I, Kakugawa K, Thiault N, Ngoi S, Olive ME, Iwaya H, Seguin L, Vicente-Suarez I, Becart S, Verstichel G, Balancio A, Altman A, Chang JT, Taniuchi I, Lillemeier B, Kronenberg M, Myers SA, Cheroutre H. A regulatory circuit controlled by extranuclear and nuclear retinoic acid receptor α determines T cell activation and function. Immunity 2023; 56:2054-2069.e10. [PMID: 37597518 PMCID: PMC10552917 DOI: 10.1016/j.immuni.2023.07.017] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 03/08/2023] [Accepted: 07/25/2023] [Indexed: 08/21/2023]
Abstract
Ligation of retinoic acid receptor alpha (RARα) by RA promotes varied transcriptional programs associated with immune activation and tolerance, but genetic deletion approaches suggest the impact of RARα on TCR signaling. Here, we examined whether RARα would exert roles beyond transcriptional regulation. Specific deletion of the nuclear isoform of RARα revealed an RARα isoform in the cytoplasm of T cells. Extranuclear RARα was rapidly phosphorylated upon TCR stimulation and recruited to the TCR signalosome. RA interfered with extranuclear RARα signaling, causing suboptimal TCR activation while enhancing FOXP3+ regulatory T cell conversion. TCR activation induced the expression of CRABP2, which translocates RA to the nucleus. Deletion of Crabp2 led to increased RA in the cytoplasm and interfered with signalosome-RARα, resulting in impaired anti-pathogen immunity and suppressed autoimmune disease. Our findings underscore the significance of subcellular RA/RARα signaling in T cells and identify extranuclear RARα as a component of the TCR signalosome and a determinant of immune responses.
Collapse
Affiliation(s)
- Alexandre Larange
- Center for Autoimmunity and Inflammation, La Jolla Institute for Immunology, La Jolla, CA 92037, USA; Center for Cancer Immunotherapy, La Jolla Institute for Immunology, La Jolla, CA 92037, USA
| | - Ikuo Takazawa
- Center for Autoimmunity and Inflammation, La Jolla Institute for Immunology, La Jolla, CA 92037, USA; Center for Cancer Immunotherapy, La Jolla Institute for Immunology, La Jolla, CA 92037, USA
| | - Kiyokazu Kakugawa
- Laboratory for Immune Crosstalk, RIKEN Center for Integrative Medical Sciences, 1-7-22 Suehiro, Tsurumi-ku, Yokohama 230-0045, Japan
| | - Nicolas Thiault
- Center for Autoimmunity and Inflammation, La Jolla Institute for Immunology, La Jolla, CA 92037, USA; Center for Cancer Immunotherapy, La Jolla Institute for Immunology, La Jolla, CA 92037, USA
| | - SooMun Ngoi
- School of Biological Sciences, University of California San Diego, La Jolla, CA 92093, USA
| | - Meagan E Olive
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA 02142, USA
| | - Hitoshi Iwaya
- Center for Autoimmunity and Inflammation, La Jolla Institute for Immunology, La Jolla, CA 92037, USA; Center for Cancer Immunotherapy, La Jolla Institute for Immunology, La Jolla, CA 92037, USA
| | - Laetitia Seguin
- Center for Autoimmunity and Inflammation, La Jolla Institute for Immunology, La Jolla, CA 92037, USA; Center for Cancer Immunotherapy, La Jolla Institute for Immunology, La Jolla, CA 92037, USA
| | - Ildefonso Vicente-Suarez
- Center for Autoimmunity and Inflammation, La Jolla Institute for Immunology, La Jolla, CA 92037, USA; Center for Cancer Immunotherapy, La Jolla Institute for Immunology, La Jolla, CA 92037, USA
| | - Stephane Becart
- Center for Autoimmunity and Inflammation, La Jolla Institute for Immunology, La Jolla, CA 92037, USA; Center for Cancer Immunotherapy, La Jolla Institute for Immunology, La Jolla, CA 92037, USA
| | - Greet Verstichel
- Center for Autoimmunity and Inflammation, La Jolla Institute for Immunology, La Jolla, CA 92037, USA; Center for Cancer Immunotherapy, La Jolla Institute for Immunology, La Jolla, CA 92037, USA
| | - Ann Balancio
- Center for Autoimmunity and Inflammation, La Jolla Institute for Immunology, La Jolla, CA 92037, USA; Center for Cancer Immunotherapy, La Jolla Institute for Immunology, La Jolla, CA 92037, USA
| | - Amnon Altman
- Center for Autoimmunity and Inflammation, La Jolla Institute for Immunology, La Jolla, CA 92037, USA; Center for Cancer Immunotherapy, La Jolla Institute for Immunology, La Jolla, CA 92037, USA
| | - John T Chang
- School of Biological Sciences, University of California San Diego, La Jolla, CA 92093, USA
| | - Ichiro Taniuchi
- Laboratory for Transcriptional Regulation, RIKEN Center for Integrative Medical Sciences, 1-7-22 Suehiro, Tsurumi-ku, Yokohama 230-0045, Japan
| | - Bjorn Lillemeier
- Immunobiology and Microbial Pathogenesis Laboratory, IMPL-L, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Mitchell Kronenberg
- Center for Autoimmunity and Inflammation, La Jolla Institute for Immunology, La Jolla, CA 92037, USA; School of Biological Sciences, University of California San Diego, La Jolla, CA 92093, USA; Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA 92037, USA
| | - Samuel A Myers
- Center for Autoimmunity and Inflammation, La Jolla Institute for Immunology, La Jolla, CA 92037, USA; Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA 02142, USA; Laboratory for Immunochemical Circuits, La Jolla Institute for Immunology, La Jolla, CA 92037, USA.
| | - Hilde Cheroutre
- Center for Autoimmunity and Inflammation, La Jolla Institute for Immunology, La Jolla, CA 92037, USA; Center for Cancer Immunotherapy, La Jolla Institute for Immunology, La Jolla, CA 92037, USA; Laboratory for Immune Crosstalk, RIKEN Center for Integrative Medical Sciences, 1-7-22 Suehiro, Tsurumi-ku, Yokohama 230-0045, Japan.
| |
Collapse
|
31
|
Mikami N, Sakaguchi S. Regulatory T cells in autoimmune kidney diseases and transplantation. Nat Rev Nephrol 2023; 19:544-557. [PMID: 37400628 DOI: 10.1038/s41581-023-00733-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/30/2023] [Indexed: 07/05/2023]
Abstract
Regulatory T (Treg) cells that express the transcription factor forkhead box protein P3 (FOXP3) are naturally present in the immune system and have roles in the maintenance of immunological self-tolerance and immune system and tissue homeostasis. Treg cells suppress T cell activation, expansion and effector functions by various mechanisms, particularly by controlling the functions of antigen-presenting cells. They can also contribute to tissue repair by suppressing inflammation and facilitating tissue regeneration, for example, via the production of growth factors and the promotion of stem cell differentiation and proliferation. Monogenic anomalies of Treg cells and genetic variations of Treg cell functional molecules can cause or predispose patients to the development of autoimmune diseases and other inflammatory disorders, including kidney diseases. Treg cells can potentially be utilized or targeted to treat immunological diseases and establish transplantation tolerance, for example, by expanding natural Treg cells in vivo using IL-2 or small molecules or by expanding them in vitro for adoptive Treg cell therapy. Efforts are also being made to convert antigen-specific conventional T cells into Treg cells and to generate chimeric antigen receptor Treg cells from natural Treg cells for adoptive Treg cell therapies with the aim of achieving antigen-specific immune suppression and tolerance in the clinic.
Collapse
Affiliation(s)
- Norihisa Mikami
- Laboratory of Experimental Immunology, Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Shimon Sakaguchi
- Laboratory of Experimental Immunology, Immunology Frontier Research Center, Osaka University, Osaka, Japan.
- Institute for Life and Medical Sciences, Kyoto University, Kyoto, Japan.
| |
Collapse
|
32
|
DeBerge M, Chaudhary R, Schroth S, Thorp EB. Immunometabolism at the Heart of Cardiovascular Disease. JACC Basic Transl Sci 2023; 8:884-904. [PMID: 37547069 PMCID: PMC10401297 DOI: 10.1016/j.jacbts.2022.12.010] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 12/21/2022] [Accepted: 12/27/2022] [Indexed: 08/08/2023]
Abstract
Immune cell function among the myocardium, now more than ever, is appreciated to regulate cardiac function and pathophysiology. This is the case for both innate immunity, which includes neutrophils, monocytes, dendritic cells, and macrophages, as well as adaptive immunity, which includes T cells and B cells. This function is fueled by cell-intrinsic shifts in metabolism, such as glycolysis and oxidative phosphorylation, as well as metabolite availability, which originates from the surrounding extracellular milieu and varies during ischemia and metabolic syndrome. Immune cell crosstalk with cardiac parenchymal cells, such as cardiomyocytes and fibroblasts, is also regulated by complex cellular metabolic circuits. Although our understanding of immunometabolism has advanced rapidly over the past decade, in part through valuable insights made in cultured cells, there remains much to learn about contributions of in vivo immunometabolism and directly within the myocardium. Insight into such fundamental cell and molecular mechanisms holds potential to inform interventions that shift the balance of immunometabolism from maladaptive to cardioprotective and potentially even regenerative. Herein, we review our current working understanding of immunometabolism, specifically in the settings of sterile ischemic cardiac injury or cardiometabolic disease, both of which contribute to the onset of heart failure. We also discuss current gaps in knowledge in this context and therapeutic implications.
Collapse
Affiliation(s)
| | | | - Samantha Schroth
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Edward B. Thorp
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| |
Collapse
|
33
|
Mancuso G, Bechi Genzano C, Fierabracci A, Fousteri G. Type 1 diabetes and inborn errors of immunity: Complete strangers or 2 sides of the same coin? J Allergy Clin Immunol 2023; 151:1429-1447. [PMID: 37097271 DOI: 10.1016/j.jaci.2023.03.026] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 03/31/2023] [Accepted: 03/31/2023] [Indexed: 04/26/2023]
Abstract
Type 1 diabetes (T1D) is a polygenic disease and does not follow a mendelian pattern. Inborn errors of immunity (IEIs), on the other hand, are caused by damaging germline variants, suggesting that T1D and IEIs have nothing in common. Some IEIs, resulting from mutations in genes regulating regulatory T-cell homeostasis, are associated with elevated incidence of T1D. The genetic spectrum of IEIs is gradually being unraveled; consequently, molecular pathways underlying human monogenic autoimmunity are being identified. There is an appreciable overlap between some of these pathways and the genetic variants that determine T1D susceptibility, suggesting that after all, IEI and T1D are 2 sides of the same coin. The study of monogenic IEIs with a variable incidence of T1D has the potential to provide crucial insights into the mechanisms leading to T1D. These insights contribute to the definition of T1D endotypes and explain disease heterogeneity. In this review, we discuss the interconnected pathogenic pathways of autoimmunity, β-cell function, and primary immunodeficiency. We also examine the role of environmental factors in disease penetrance as well as the circumstantial evidence of IEI drugs in preventing and curing T1D in individuals with IEIs, suggesting the repositioning of these drugs also for T1D therapy.
Collapse
Affiliation(s)
- Gaia Mancuso
- Unit of Immunology, Rheumatology, Allergy and Rare Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Camillo Bechi Genzano
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University Irving Medical Center, New York, NY
| | | | - Georgia Fousteri
- Diabetes Research Institute, IRCCS Ospedale San Raffaele, Milan, Italy.
| |
Collapse
|
34
|
Roy S, Batra L. Protein Phosphatase 2A: Role in T Cells and Diseases. J Immunol Res 2023; 2023:4522053. [PMID: 37234102 PMCID: PMC10208765 DOI: 10.1155/2023/4522053] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2023] [Revised: 05/02/2023] [Accepted: 05/04/2023] [Indexed: 05/27/2023] Open
Abstract
Protein phosphatase 2A (PP2A) is a serine-threonine phosphatase that plays an important role in the regulation of cell proliferation and signal transduction. The catalytic activity of PP2A is integral in the maintenance of physiological functions which gets severely impaired in its absence. PP2A plays an essential role in the activation, differentiation, and functions of T cells. PP2A suppresses Th1 cell differentiation while promoting Th2 cell differentiation. PP2A fosters Th17 cell differentiation which contributes to the pathogenesis of systemic lupus erythematosus (SLE) by enhancing the transactivation of the Il17 gene. Genetic deletion of PP2A in Tregs disrupts Foxp3 expression due to hyperactivation of mTORC1 signaling which impairs the development and immunosuppressive functions of Tregs. PP2A is important in the induction of Th9 cells and promotes their antitumor functions. PP2A activation has shown to reduce neuroinflammation in a mouse model of experimental autoimmune encephalomyelitis (EAE) and is now used to treat multiple sclerosis (MS) clinically. In this review, we will discuss the structure and functions of PP2A in T cell differentiation and diseases and therapeutic applications of PP2A-mediated immunotherapy.
Collapse
Affiliation(s)
- Suyasha Roy
- Immuno-Biology Laboratory, Translational Health Science and Technology Institute, Faridabad, India
- Laboratory of Molecular Immunology, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Lalit Batra
- Regional Biocontainment Laboratory, Center for Predictive Medicine, University of Louisville, Louisville, KY, USA
| |
Collapse
|
35
|
Tanaka T, Tawara M, Suzuki H, Kaneko MK, Kato Y. Identification of the Binding Epitope of an Anti-Mouse CCR6 Monoclonal Antibody (C 6Mab-13) Using 1× Alanine Scanning. Antibodies (Basel) 2023; 12:antib12020032. [PMID: 37218898 DOI: 10.3390/antib12020032] [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: 03/16/2023] [Revised: 03/28/2023] [Accepted: 04/17/2023] [Indexed: 05/24/2023] Open
Abstract
CC chemokine receptor 6 (CCR6) is one of the members of the G-protein-coupled receptor (GPCR) family that is upregulated in many immune-related cells, such as B lymphocytes, effector and memory T cells, regulatory T cells, and immature dendritic cells. The coordination between CCR6 and its ligand CC motif chemokine ligand 20 (CCL20) is deeply involved in the pathogenesis of various diseases, such as cancer, psoriasis, and autoimmune diseases. Thus, CCR6 is an attractive target for therapy and is being investigated as a diagnostic marker for various diseases. In a previous study, we developed an anti-mouse CCR6 (mCCR6) monoclonal antibody (mAb), C6Mab-13 (rat IgG1, kappa), that was applicable for flow cytometry by immunizing a rat with the N-terminal peptide of mCCR6. In this study, we investigated the binding epitope of C6Mab-13 using an enzyme-linked immunosorbent assay (ELISA) and the surface plasmon resonance (SPR) method, which were conducted with respect to the synthesized point-mutated-peptides within the 1-20 amino acid region of mCCR6. In the ELISA results, C6Mab-13 lost its ability to react to the alanine-substituted peptide of mCCR6 at Asp11, thereby identifying Asp11 as the epitope of C6Mab-13. In our SPR analysis, the dissociation constants (KD) could not be calculated for the G9A and D11A mutants due to the lack of binding. The SPR analysis demonstrated that the C6Mab-13 epitope comprises Gly9 and Asp11. Taken together, the key binding epitope of C6Mab-13 was determined to be located around Asp11 on mCCR6. Based on the epitope information, C6Mab-13 could be useful for further functional analysis of mCCR6 in future studies.
Collapse
Affiliation(s)
- Tomohiro Tanaka
- Department of Molecular Pharmacology, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Japan
| | - Mayuki Tawara
- Department of Molecular Pharmacology, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Japan
| | - Hiroyuki Suzuki
- Department of Molecular Pharmacology, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Japan
| | - Mika K Kaneko
- Department of Molecular Pharmacology, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Japan
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Japan
| | - Yukinari Kato
- Department of Molecular Pharmacology, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Japan
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Japan
| |
Collapse
|
36
|
Grajchen E, Loix M, Baeten P, Côrte-Real BF, Hamad I, Vanherle S, Haidar M, Dehairs J, Broos JY, Ntambi JM, Zimmermann R, Breinbauer R, Stinissen P, Hellings N, Verberk SGS, Kooij G, Giera M, Swinnen JV, Broux B, Kleinewietfeld M, Hendriks JJA, Bogie JFJ. Fatty acid desaturation by stearoyl-CoA desaturase-1 controls regulatory T cell differentiation and autoimmunity. Cell Mol Immunol 2023; 20:666-679. [PMID: 37041314 DOI: 10.1038/s41423-023-01011-2] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Accepted: 03/23/2023] [Indexed: 04/13/2023] Open
Abstract
The imbalance between pathogenic and protective T cell subsets is a cardinal feature of autoimmune disorders such as multiple sclerosis (MS). Emerging evidence indicates that endogenous and dietary-induced changes in fatty acid metabolism have a major impact on both T cell fate and autoimmunity. To date, however, the molecular mechanisms that underlie the impact of fatty acid metabolism on T cell physiology and autoimmunity remain poorly understood. Here, we report that stearoyl-CoA desaturase-1 (SCD1), an enzyme essential for the desaturation of fatty acids and highly regulated by dietary factors, acts as an endogenous brake on regulatory T-cell (Treg) differentiation and augments autoimmunity in an animal model of MS in a T cell-dependent manner. Guided by RNA sequencing and lipidomics analysis, we found that the absence of Scd1 in T cells promotes the hydrolysis of triglycerides and phosphatidylcholine through adipose triglyceride lipase (ATGL). ATGL-dependent release of docosahexaenoic acid enhanced Treg differentiation by activating the nuclear receptor peroxisome proliferator-activated receptor gamma. Our findings identify fatty acid desaturation by SCD1 as an essential determinant of Treg differentiation and autoimmunity, with potentially broad implications for the development of novel therapeutic strategies and dietary interventions for autoimmune disorders such as MS.
Collapse
Affiliation(s)
- Elien Grajchen
- Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
- University MS Center Hasselt, Pelt, Belgium
| | - Melanie Loix
- Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
- University MS Center Hasselt, Pelt, Belgium
| | - Paulien Baeten
- University MS Center Hasselt, Pelt, Belgium
- Neuro-Immune Connections and Repair Lab, Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
| | - Beatriz F Côrte-Real
- Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
- University MS Center Hasselt, Pelt, Belgium
- VIB Laboratory of Translational Immunomodulation, VIB Center for Inflammation Research, Hasselt University, Diepenbeek, Belgium
| | - Ibrahim Hamad
- Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
- University MS Center Hasselt, Pelt, Belgium
- VIB Laboratory of Translational Immunomodulation, VIB Center for Inflammation Research, Hasselt University, Diepenbeek, Belgium
| | - Sam Vanherle
- Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
- University MS Center Hasselt, Pelt, Belgium
| | - Mansour Haidar
- Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
- University MS Center Hasselt, Pelt, Belgium
| | - Jonas Dehairs
- Department of Oncology, Laboratory of Lipid Metabolism and Cancer, LKI - Leuven Cancer Institute, KU Leuven - University of Leuven, Leuven, Belgium
| | - Jelle Y Broos
- Department of Molecular Cell Biology and Immunology, Amsterdam University Medical Center, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, MS Center Amsterdam, Amsterdam, The Netherlands
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, The Netherlands
| | - James M Ntambi
- Department of Biochemistry, Department of Nutritional Sciences, University of Wisconsin-Madison, Madison, USA
| | - Robert Zimmermann
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
- BioTechMed-Graz, Graz, Austria
| | - Rolf Breinbauer
- BioTechMed-Graz, Graz, Austria
- Institute of Organic Chemistry, Graz University of Technology, Graz, Austria
| | - Piet Stinissen
- Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
- University MS Center Hasselt, Pelt, Belgium
| | - Niels Hellings
- University MS Center Hasselt, Pelt, Belgium
- Neuro-Immune Connections and Repair Lab, Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
| | - Sanne G S Verberk
- Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
- University MS Center Hasselt, Pelt, Belgium
| | - Gijs Kooij
- Department of Molecular Cell Biology and Immunology, Amsterdam University Medical Center, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, MS Center Amsterdam, Amsterdam, The Netherlands
| | - Martin Giera
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, The Netherlands
| | - Johannes V Swinnen
- Department of Oncology, Laboratory of Lipid Metabolism and Cancer, LKI - Leuven Cancer Institute, KU Leuven - University of Leuven, Leuven, Belgium
| | - Bieke Broux
- University MS Center Hasselt, Pelt, Belgium
- Neuro-Immune Connections and Repair Lab, Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
- Cardiovascular Research Institute Maastricht, Department of Internal Medicine, Maastricht University, Maastricht, The Netherlands
| | - Markus Kleinewietfeld
- Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
- University MS Center Hasselt, Pelt, Belgium
- VIB Laboratory of Translational Immunomodulation, VIB Center for Inflammation Research, Hasselt University, Diepenbeek, Belgium
| | - Jerome J A Hendriks
- Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
- University MS Center Hasselt, Pelt, Belgium
| | - Jeroen F J Bogie
- Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium.
- University MS Center Hasselt, Pelt, Belgium.
| |
Collapse
|
37
|
Fu W, Wu G. Targeting mTOR for Anti-Aging and Anti-Cancer Therapy. Molecules 2023; 28:molecules28073157. [PMID: 37049920 PMCID: PMC10095787 DOI: 10.3390/molecules28073157] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 03/24/2023] [Accepted: 03/31/2023] [Indexed: 04/05/2023] Open
Abstract
The balance between anabolism and catabolism is disrupted with aging, with the rate of anabolism being faster than that of catabolism. Therefore, mTOR, whose major function is to enhance anabolism and inhibit catabolism, has become a potential target of inhibition for anti-aging therapy. Interestingly, it was found that the downregulation of the mTOR signaling pathway had a lifespan-extending effect resembling calorie restriction. In addition, the mTOR signaling pathway promotes cell proliferation and has been regarded as a potential anti-cancer target. Rapamycin and rapalogs, such as everolimus, have proven to be effective in preventing certain tumor growth. Here, we reviewed the basic knowledge of mTOR signaling, including both mTORC1 and mTORC2. Then, for anti-aging, we cited a lot of evidence to discuss the role of targeting mTOR and its anti-aging mechanism. For cancer therapy, we also discussed the role of mTOR signaling in different types of cancers, including idiopathic pulmonary fibrosis, tumor immunity, etc. In short, we discussed the research progress and both the advantages and disadvantages of targeting mTOR in anti-aging and anti-cancer therapy. Hopefully, this review may promote more ideas to be generated for developing inhibitors of mTOR signaling to fight cancer and extend lifespan.
Collapse
Affiliation(s)
- Wencheng Fu
- State Key Laboratory of Microbial Metabolism, School of Life Sciences & Biotechnology, the Joint International Research Laboratory of Metabolic & Developmental Sciences MOE, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Geng Wu
- State Key Laboratory of Microbial Metabolism, School of Life Sciences & Biotechnology, the Joint International Research Laboratory of Metabolic & Developmental Sciences MOE, Shanghai Jiao Tong University, Shanghai 200240, China
| |
Collapse
|
38
|
Wang D, Bauersachs J, Berliner D. Immune Checkpoint Inhibitor Associated Myocarditis and Cardiomyopathy: A Translational Review. BIOLOGY 2023; 12:biology12030472. [PMID: 36979163 PMCID: PMC10045178 DOI: 10.3390/biology12030472] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 03/13/2023] [Accepted: 03/16/2023] [Indexed: 03/30/2023]
Abstract
Immune checkpoint inhibitors (ICIs) have revolutionized oncology and transformed the treatment of various malignancies. By unleashing the natural immunological brake of the immune system, ICIs were initially considered an effective, gentle therapy with few side effects. However, accumulated clinical knowledge reveals that ICIs are associated with inflammation and tissue damage in multiple organs, leading to immune-related adverse effects (irAEs). Most irAEs involve the skin and gastrointestinal tract; however, cardiovascular involvement is associated with very high mortality rates, and its underlying pathomechanisms are poorly understood. Ranging from acute myocarditis to chronic cardiomyopathies, ICI-induced cardiotoxicity can present in various forms and entities. Revealing the inciting factors, understanding the pathogenesis, and identifying effective treatment strategies are needed to improve the care of tumor patients and our understanding of the immune and cardiovascular systems.
Collapse
Affiliation(s)
- Dong Wang
- Department of Cardiology and Angiology, Hannover Medical School, 30625 Hannover, Germany
| | - Johann Bauersachs
- Department of Cardiology and Angiology, Hannover Medical School, 30625 Hannover, Germany
| | - Dominik Berliner
- Department of Cardiology and Angiology, Hannover Medical School, 30625 Hannover, Germany
| |
Collapse
|
39
|
Gu Q, Tung KS, Lorenz UM. Treg-specific deletion of the phosphatase SHP-1 impairs control of inflammation in vivo. Front Immunol 2023; 14:1139326. [PMID: 37006301 PMCID: PMC10060847 DOI: 10.3389/fimmu.2023.1139326] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Accepted: 02/28/2023] [Indexed: 03/18/2023] Open
Abstract
Introduction To achieve a healthy and functional immune system, a delicate balance exists between the activation of conventional T cells (Tcon cells) and the suppression by regulatory T cells (Treg). The tyrosine phosphatase SHP-1, a negative regulator of TCR signaling, shapes this 'activation-suppression' balance by modulating Tcon cell resistance to Treg-mediated suppression. Treg cells also express SHP-1, but its role in influencing Treg function is still not fully understood. Methods We generated a Treg-specific SHP-1 deletion model, Foxp3Cre+ Shp-1f/f , to address how SHP-1 affects Treg function and thereby contributes to T cell homeostasis using a combination of ex vivo studies and in vivo models of inflammation and autoimmunity. Results We show that SHP-1 modulates Treg suppressive function at different levels. First, at the intracellular signaling level in Treg cells, SHP-1 attenuates TCR-dependent Akt phosphorylation, with loss of SHP-1 driving Treg cells towards a glycolysis pathway. At the functional level, SHP-1 expression limits the in vivo accumulation of CD44hiCD62Llo T cells within the steady state Tcon populations (both CD8+ as well as CD4+ Tcon). Further, SHP-1-deficient Treg cells are less efficient in suppressing inflammation in vivo; mechanistically, this appears to be due to a failure to survive or a defect in migration of SHP-1-deficient Treg cells to peripheral inflammation sites. Conclusion Our data identify SHP-1 as an important intracellular mediator for fine-tuning the balance between Treg-mediated suppression and Tcon activation/resistance.
Collapse
Affiliation(s)
- QinLei Gu
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA, United States
- Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, VA, United States
| | - Kenneth S. Tung
- Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, VA, United States
- Department of Pathology, University of Virginia, Charlottesville, VA, United States
| | - Ulrike M. Lorenz
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA, United States
- Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, VA, United States
- Department of Pathology and Immunology, Washington University in St. Louis, Saint Louis, MO, United States
| |
Collapse
|
40
|
Meitei HT, Lal G. T cell receptor signaling in the differentiation and plasticity of CD4 + T cells. Cytokine Growth Factor Rev 2023; 69:14-27. [PMID: 36028461 DOI: 10.1016/j.cytogfr.2022.08.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 08/17/2022] [Indexed: 02/07/2023]
Abstract
CD4+ T cells are critical components of the adaptive immune system. The T cell receptor (TCR) and co-receptor signaling cascades shape the phenotype and functions of CD4+ T cells. TCR signaling plays a crucial role in T cell development, antigen recognition, activation, and differentiation upon recognition of foreign- or auto-antigens. In specific autoimmune conditions, altered TCR repertoire is reported and can predispose autoimmunity with organ-specific inflammation and tissue damage. TCR signaling modulates various signaling cascades and regulates epigenetic and transcriptional regulation during homeostasis and disease conditions. Understanding the mechanism by which coreceptors and cytokine signals control the magnitude of TCR signal amplification will aid in developing therapeutic strategies to treat inflammation and autoimmune diseases. This review focuses on the role of the TCR signaling cascade and its components in the activation, differentiation, and plasticity of various CD4+ T cell subsets.
Collapse
Affiliation(s)
| | - Girdhari Lal
- National Centre for Cell Science, SPPU campus, Ganeshkhind, Pune, MH 411007, India.
| |
Collapse
|
41
|
Laukova M, Glatman Zaretsky A. Regulatory T cells as a therapeutic approach for inflammatory bowel disease. Eur J Immunol 2023; 53:e2250007. [PMID: 36562391 PMCID: PMC10107179 DOI: 10.1002/eji.202250007] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 11/20/2022] [Accepted: 12/22/2022] [Indexed: 12/24/2022]
Abstract
Foxp3+ T regulatory (Treg) cells suppress inflammation and are essential for maintaining tissue homeostasis. A growing appreciation of tissue-specific Treg functions has built interest in leveraging the endogenous suppressive mechanisms of these cells into cellular therapeutics in organ-specific diseases. Notably, Treg cells play a critical role in maintaining the intestinal environment. As a barrier site, the gut requires Treg cells to mediate interactions with the microbiota, support barrier integrity, and regulate the immune system. Without fully functional Treg cells, intestinal inflammation and microbial dysbiosis ensue. Thus, there is a particular interest in developing Treg cellular therapies for intestinal inflammatory disease, such as inflammatory bowel disease (IBD). This article reviews some of the critical pathways that are dysregulated in IBD, Treg cell mechanisms of suppression, and the efforts and approaches in the field to develop these cells as a cellular therapy for IBD.
Collapse
|
42
|
Wen J, Zhao W, Shu X. A novel cuproptosis-related LncRNA signature: Prognostic and therapeutic value for low grade glioma. Front Oncol 2023; 12:1087762. [PMID: 36776374 PMCID: PMC9909527 DOI: 10.3389/fonc.2022.1087762] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Accepted: 12/28/2022] [Indexed: 01/27/2023] Open
Abstract
Background As a common primary intracranial tumor, the diagnosis and therapy of low-grade glioma (LGG) remains a pivotal barrier. Cuproptosis, a new way induces cell death, has attracted worldwide attention. However, the relationship between cuproptosis and LGG remains unknown. Our study is all about finding out if there are any genes related to coproptosis that can be used to predict the outcome of LGG. Methods RNA data and clinical information were selected from Cancer Genome Atlas (TCGA) datasets and the Genotype-Tissue Expression (GTEx), 5 lncRNAs (GAS5.AS1, MYLK.AS1, AC142472.1, AC011346.1, AL359643.3) were identified by Cox univariate and multivariate regression, as well as LASSO Cox regression. In the training and test sets, a dual validation of the predictive signature comprised of these 5 lncRNAs was undertaken. The findings demonstrate that the risk model is able to predict the survival regression of LGG patients and has a good performance in either the KM curve approach or the ROC curve. GO, GSEA and KEGG were carried out to explore the possible molecular processes that affecting the prognosis of LGG. The characteristics of immune microenvironment were investigated by using CIBERSORT, ESTIMATE and ssGSEA. Results We identified five lncRNAs related with cuproptosis that were closely associated with the prognosis of LGG and used these five lncRNAs to develop a risk model. Using this risk model, LGG patients were then divided into high-risk and low-risk groups. The two patient groups had significantly distinct survival characteristics. Analyses of Gene Ontology (GO) and the Kyoto Encyclopedia of Genes and Genomes (KEGG) revealed that the differential genes of the two patient groups were primarily concentrated in neural active ligand-receptor interaction and cytokine-cytokine receptor interaction. The ssGSEA score determined the information related to immune infiltration, and the two groups were differentially expressed in immune subpopulations such as T cells and B cells as well. Conclusion Our study discovered 5 cuproptosis-related lncRNAs which contribute to predicting patients' survival of LGG and provide ideas for the exploration of new targets for LGG in the future.
Collapse
Affiliation(s)
- Jun Wen
- Chongqing Cancer Multi-Omics Big Data Application Engineering Research Center, Chongqing University Cancer Hospital, Chongqing, China
| | - Wenting Zhao
- The First Clinical College, Hubei University of Chinese Medicine, Wuhan, China
| | - Xiaolei Shu
- Chongqing Cancer Multi-Omics Big Data Application Engineering Research Center, Chongqing University Cancer Hospital, Chongqing, China
| |
Collapse
|
43
|
Recent insights into the role of Akt in CD4 T-cell activation and differentiation: alternative splicing and beyond. IMMUNOMETABOLISM (COBHAM (SURREY, ENGLAND)) 2023; 5:e00015. [PMID: 36710922 PMCID: PMC9869951 DOI: 10.1097/in9.0000000000000015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Accepted: 11/03/2022] [Indexed: 01/31/2023]
Abstract
The activation and differentiation of CD4+ T cells is a complex process that is controlled by many factors. A critical component of the signaling pathway triggered following T-cell receptor (TCR) engagement is the serine threonine kinase Akt. Akt is involved in the control of many cellular processes including proliferation, metabolism, and differentiation of specific TH-cell subsets. Recent work has shown that, depending on the nature or strength of the TCR activation, Akt may activate different sets of substrates which then lead to differential cellular outcomes. Akt plays an important role in controlling the strength of the TCR signal and several recent studies have identified novel mechanisms including control of the expression of negative regulators of TCR signaling, and the influence on regulatory T cells (Treg) and TH17 differentiation. Many of these functions are mediated via control of the FoxO family of transcription factors, that play an important role in metabolism and Th cell differentiation. A theme that is emerging is that Akt does not function in the same way in all T-cell types. We highlight differences between CD4 and CD8 T cells as well as between Treg, TH17, and TFH cells. While Akt activity has been implicated in the control of alternative splicing in tumor cells, recent studies are emerging that indicate that similar functions may exist in CD4 T cells. In this mini review, we highlight some of the recent advances in these areas of Akt function that demonstrate the varied role that Akt plays in the function of CD4 T cells.
Collapse
|
44
|
Degechisa ST, Dabi YT. Leptin Deficiency May Influence the Divergence of Cell-Mediated Immunity Between Lepromatous and Tuberculoid Leprosy Patients. J Inflamm Res 2022; 15:6719-6728. [PMID: 36536644 PMCID: PMC9758981 DOI: 10.2147/jir.s389845] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Accepted: 11/29/2022] [Indexed: 04/28/2024] Open
Abstract
Leprosy is a disease caused by an intracellular bacillus bacterium called Mycobacterium leprae which lives and multiplies in the hosts' macrophages and Schwann cells. Depending on the degree of the host's cell-mediated immunity (CMI) response to the bacilli, the disease manifests itself in five clinical spectra ranging from polar tuberculoid (TT) to polar lepromatous leprosy (LL). A very high level of T helper 1 (Th1) driven bacilli-specific CMI is seen in the TT form, whereas this response is essentially nonexistent in the LL form. As a result, there is very low or absent bacillary load and localized nodular lesions in TT patients. On the contrary, LL patients presented with high bacillary load and generalized lesions due to low CMI response. The mechanism underlying this divergence of CMI response is not clearly elucidated yet. However, mounting evidence links it to an elevated number of Th1 and Th17 suppressing CD4+ CD25+ FOXP3+ T regulatory cells (Treg cells) which are abundantly found in LL than in TT patients. The predominance of these cells in LL patients is partly attributed to a deficiency of leptin, the cytokine-like peptide hormone, in these patients. Becausea normal level of leptin promotes the proliferation and preferential differentiation of effector T cells (Th1 and Th17) while inhibiting the growth and functional responsiveness of the Treg cells. In contrast, leptin deficiency or neutralization was reported to exert the opposite effect on Treg cells and effector T cells. Other smaller subsets of lymphocytes such as gamma delta (γδ) T cells and B regulatory cells are also modulated by leptin level in the pathogenesis of leprosy. Leptin may therefore regulate the divergence of CMI between TT and LL patients by regulating the homeostasis of effector T cells and Treg cells, and this review will examine the underlying mechanism for this.
Collapse
Affiliation(s)
- Sisay Teka Degechisa
- Medical Biochemistry Department, College of Health Sciences, Addis Ababa University, Addis Abeba, Ethiopia
- Medical Laboratory Science Department, College of Medicine and Health Sciences, Arba Minch University, Arba Minch, Ethiopia
| | - Yosef Tsegaye Dabi
- Medical Biochemistry Department, College of Health Sciences, Addis Ababa University, Addis Abeba, Ethiopia
- Medical Laboratory Science Department, Wollega University, Nekemte, Ethiopia
| |
Collapse
|
45
|
Burkard T, Herrero San Juan M, Dreis C, Kiprina A, Namgaladze D, Siebenbrodt K, Luger S, Foerch C, Pfeilschifter JM, Weigert A, Radeke HH. Differential expression of CD8 defines phenotypically distinct cytotoxic T cells in cancer and multiple sclerosis. Clin Transl Med 2022; 12:e1068. [PMID: 36504430 PMCID: PMC9742381 DOI: 10.1002/ctm2.1068] [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: 07/09/2022] [Revised: 09/13/2022] [Accepted: 09/19/2022] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Cytotoxic T lymphocytes take on a leading role in many immune-related diseases. They function as key effector immune cells fighting cancer cells, but they are also considerably involved in autoimmune diseases. Common to both situations, CD8+ T cells need to adapt their metabolism and effector function to the harsh and nutrient-deprived conditions of the disease-associated microenvironment. METHODS We used an in vitro starvation as well as rapamycin treatment protocol mimicking nutrient deprivation to generate CD8Low versus CD8High T cells and performed FACS-Sorting followed by transcriptomic profiling of the cytotoxic T cell subsets. Prominent markers identified in the CD8Low versus the CD8High T cells were then used to investigate the presence of these cell subsets in immune-related human diseases. Employing cancer tissue microarrays and PhenOptics multispectral imaging as well as flow cytometry, we studied these CD8+ T cell subsets in cancer and relapsing-remitting multiple sclerosis patients. RESULTS Starvation induced a decreased expression of CD8, yielding a CD8Low T cell subpopulation with an altered transcriptomic signature and reduced effector function. CD8Low T cell showed enhanced ST2L and IL6ST (CD130) expression compared to CD8High T cells which expressed elevated KLRD1 (CD94) and granzyme B levels within the tumour microenvironment (TME). Spatial analysis revealed the presence of CD8High T cells in close proximity to tumour cells, while the CD8Low T cells resided at the tumour boundaries. Importantly, the number of tumour-infiltrating CD8Low T lymphocytes correlated with a poor prognosis as well as with enhanced cancer progression in human mammary carcinoma. We also found a reduced frequency of CD8Low T lymphocytes in a cohort of relapse (disease active) multiple sclerosis patients compared to healthy subjects during immune cell starvation in vitro. CONCLUSIONS In summary, our data show that functionally distinct cytotoxic T lymphocytes can be identified based on their expression of CD8. Indicating a more general role in CD8 T cell immunity, these cells may play opposing roles in the TME, and also in the pathophysiology of autoimmune diseases such as multiple sclerosis.
Collapse
Affiliation(s)
- Tobias Burkard
- Pharmazentrum Frankfurt/ZAFESInstitute of Pharmacology and ToxicologyHospital of the Goethe UniversityFrankfurt am MainGermany
| | - Martina Herrero San Juan
- Pharmazentrum Frankfurt/ZAFESInstitute of Pharmacology and ToxicologyHospital of the Goethe UniversityFrankfurt am MainGermany
| | - Caroline Dreis
- Pharmazentrum Frankfurt/ZAFESInstitute of Pharmacology and ToxicologyHospital of the Goethe UniversityFrankfurt am MainGermany
| | - Anastasiia Kiprina
- Faculty of MedicineInstitute of Biochemistry IGoethe‐University Frankfurt/MainFrankfurt am MainGermany
| | - Dmitry Namgaladze
- Faculty of MedicineInstitute of Biochemistry IGoethe‐University Frankfurt/MainFrankfurt am MainGermany
| | - Kai Siebenbrodt
- Department of NeurologyGoethe University Hospital FrankfurtFrankfurt am MainGermany
- Epilepsy Center Frankfurt Rhine‐MainDepartment of NeurologyUniversity Hospital Frankfurt, Frankfurt, Germany
| | - Sebastian Luger
- Department of NeurologyGoethe University Hospital FrankfurtFrankfurt am MainGermany
| | - Christian Foerch
- Department of NeurologyGoethe University Hospital FrankfurtFrankfurt am MainGermany
| | - Josef M. Pfeilschifter
- Pharmazentrum Frankfurt/ZAFESInstitute of Pharmacology and ToxicologyHospital of the Goethe UniversityFrankfurt am MainGermany
| | - Andreas Weigert
- Faculty of MedicineInstitute of Biochemistry IGoethe‐University Frankfurt/MainFrankfurt am MainGermany
- Frankfurt Cancer InstituteGoethe‐University FrankfurtFrankfurtGermany
- Cardio‐Pulmonary Institute (CPI)FrankfurtGermany
| | - Heinfried H. Radeke
- Pharmazentrum Frankfurt/ZAFESInstitute of Pharmacology and ToxicologyHospital of the Goethe UniversityFrankfurt am MainGermany
| |
Collapse
|
46
|
Willman J, Willman M, Reddy R, Fusco A, Sriram S, Mehkri Y, Charles J, Goeckeritz J, Lucke‐Wold B. Gut microbiome and neurosurgery: Implications for treatment. CLINICAL AND TRANSLATIONAL DISCOVERY 2022; 2. [PMID: 36268259 PMCID: PMC9577538 DOI: 10.1002/ctd2.139] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Accepted: 09/19/2022] [Indexed: 11/06/2022]
Abstract
AbstractIntroductionThe aim of this review is to summarize the current understanding of the gut‐brain axis (GBA), its impact on neurosurgery, and its implications for future treatment.BackgroundAn abundance of research has established the existence of a collection of pathways between the gut microbiome and the central nervous system (CNS), commonly known as the GBA. Complicating this relationship, the gut microbiome bacterial diversity appears to change with age, antibiotic exposure and a number of external and internal factors.MethodsIn this paper, we present the current understanding of the key protective and deleterious roles the gut microbiome plays in the pathogenesis of several common neurosurgical concerns.ResultsSpecifically, we examine how spinal cord injury, traumatic brain injury and stroke may cause gut microbial dysbiosis. Furthermore, this link appears to be bidirectional as gut dysbiosis contributes to secondary CNS injury in each of these ailment settings. This toxic cycle may be broken, and the future secondary damage rescued by timely, therapeutic, gut microbiome modification. In addition, a robust gut microbiome appears to improve outcomes in brain tumour treatment. There are several primary routes by which microbiome dysbiosis may be ameliorated, including faecal microbiota transplant, oral probiotics, bacteriophages, genetic modification of gut microbiota and vagus nerve stimulation.ConclusionThe GBA represents an important component of patient care in the field of neurosurgery. Future research may illuminate ideal methods of therapeutic microbiome modulation in distinct pathogenic settings.
Collapse
Affiliation(s)
- Jonathan Willman
- College of Medicine University of Florida Gainesville Florida USA
| | - Matthew Willman
- College of Medicine University of Florida Gainesville Florida USA
| | - Ramya Reddy
- College of Medicine University of Florida Gainesville Florida USA
| | - Anna Fusco
- College of Medicine University of Florida Gainesville Florida USA
| | - Sai Sriram
- College of Medicine University of Florida Gainesville Florida USA
| | - Yusuf Mehkri
- College of Medicine University of Florida Gainesville Florida USA
| | - Jude Charles
- Department of Neurosurgery Jackson Memorial Hospital Miami Florida USA
| | - Joel Goeckeritz
- College of Medicine University of Florida Gainesville Florida USA
| | - Brandon Lucke‐Wold
- Department of Neurosurgery University of Florida Gainesville Florida USA
| |
Collapse
|
47
|
Rush-Kittle J, Gámez-Díaz L, Grimbacher B. Inborn errors of immunity associated with defects of self-tolerance checkpoints: The CD28 family. Pediatr Allergy Immunol 2022; 33:e13886. [PMID: 36564875 DOI: 10.1111/pai.13886] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 10/31/2022] [Accepted: 11/01/2022] [Indexed: 12/11/2022]
Abstract
One of the causes of inborn errors of immunity is immune dysregulation. The inability of the immune system to regulate the extent of its activity has several deleterious effects, including autoimmunity, recurrent infections, and malignancy. In recent years, many proteins in the CD28 family - CD28, ICOS, CTLA-4, PD-1, and BTLA - have come into the focus of several research areas for their consequential role in the upregulation or downregulation of the immune response. In this review, we will discuss the structure and function of these proteins, as well as provide an overview of the clinical picture of patients with genetic defects.
Collapse
Affiliation(s)
- Jorrell Rush-Kittle
- Center for Chronic Immunodeficiency, University Medical Center Freiburg, Freiburg, Germany
| | - Laura Gámez-Díaz
- Center for Chronic Immunodeficiency, University Medical Center Freiburg, Freiburg, Germany
| | - Bodo Grimbacher
- Center for Chronic Immunodeficiency, University Medical Center Freiburg, Freiburg, Germany
| |
Collapse
|
48
|
Yu W, Li C, Zhang D, Li Z, Xia P, Liu X, Cai X, Yang P, Ling J, Zhang J, Zhang M, Yu P. Advances in T Cells Based on Inflammation in Metabolic Diseases. Cells 2022; 11:cells11223554. [PMID: 36428983 PMCID: PMC9688178 DOI: 10.3390/cells11223554] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Revised: 10/26/2022] [Accepted: 11/02/2022] [Indexed: 11/12/2022] Open
Abstract
With the increasing incidence of metabolic diseases year by year and their impact on the incidence of cardiovascular diseases, metabolic diseases have attracted great attention as a major health care problem, but there is still no effective treatment. Oxidative stress and inflammation are the main mechanisms leading to metabolic diseases. T cells are involved in the inflammatory response, which can also regulate the development of metabolic diseases, CD4+ T cells and CD8+ T cells are mainly responsible for the role. Th1 and Th17 differentiated from CD4+ T promote inflammation, while Th2 and Treg inhibit inflammation. CD8+ T cells also contribute to inflammation. The severity and duration of inflammatory reactions can also lead to different degrees of progression of metabolic diseases. Moreover, mTOR, PI3K-Akt, and AMPK signaling pathways play unique roles in the regulation of T cells, which provide a new direction for the treatment of metabolic diseases in the future. In this review, we will elaborate on the role of T cells in regulating inflammation in various metabolic diseases, the signaling pathways that regulate T cells in metabolic diseases, and the latest research progress.
Collapse
Affiliation(s)
- Wenlu Yu
- Department of Metabolism and Endocrinology, The Second Affiliated Hospital of Nanchang University, Nanchang 330000, China
- School of Ophthalmology and Optometry, Nanchang University, Nanchang 330000, China
| | - Chunxiu Li
- Department of Metabolism and Endocrinology, The Second Affiliated Hospital of Nanchang University, Nanchang 330000, China
- School of Ophthalmology and Optometry, Nanchang University, Nanchang 330000, China
| | - Deju Zhang
- Food and Nutritional Sciences, School of Biological Sciences, The University of Hong Kong, Pokfulam Road, Hong Kong 999077, China
| | - Zhangwang Li
- Department of Metabolism and Endocrinology, The Second Affiliated Hospital of Nanchang University, Nanchang 330000, China
- The Second Clinical Medical College, Nanchang University, Nanchang 330000, China
| | - Panpan Xia
- Department of Metabolism and Endocrinology, The Second Affiliated Hospital of Nanchang University, Nanchang 330000, China
| | - Xiao Liu
- Department of Cardiology, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou 510000, China
| | - Xia Cai
- Department of Metabolism and Endocrinology, The Second Affiliated Hospital of Nanchang University, Nanchang 330000, China
| | - Pingping Yang
- Department of Metabolism and Endocrinology, The Second Affiliated Hospital of Nanchang University, Nanchang 330000, China
| | - Jitao Ling
- Department of Metabolism and Endocrinology, The Second Affiliated Hospital of Nanchang University, Nanchang 330000, China
| | - Jing Zhang
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang 330000, China
- Correspondence: (J.Z.); (P.Y.)
| | - Meiying Zhang
- Department of Metabolism and Endocrinology, The Second Affiliated Hospital of Nanchang University, Nanchang 330000, China
| | - Peng Yu
- Department of Metabolism and Endocrinology, The Second Affiliated Hospital of Nanchang University, Nanchang 330000, China
- Correspondence: (J.Z.); (P.Y.)
| |
Collapse
|
49
|
Direct AKT activation in tumor-infiltrating lymphocytes markedly increases interferon-γ (IFN-γ) for the regression of tumors resistant to PD-1 checkpoint blockade. Sci Rep 2022; 12:18509. [PMID: 36323740 PMCID: PMC9630443 DOI: 10.1038/s41598-022-23016-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Accepted: 10/21/2022] [Indexed: 11/17/2022] Open
Abstract
PD-1 immune checkpoint blockade against inhibitory receptors such as receptor programmed cell death-1 (PD-1), has revolutionized cancer treatment. Effective immune reactivity against tumour antigens requires the infiltration and activation of tumour-infiltrating T-cells (TILs). In this context, ligation of the antigen-receptor complex (TCR) in combination with the co-receptor CD28 activates the intracellular mediator AKT (or PKB, protein kinase B) and its downstream targets. PD-1 inhibits the activation of AKT/PKB. Given this, we assessed whether the direct activation of AKT might be effective in activating the immune system to limit the growth of tumors that are resistant to PD-1 checkpoint blockade. We found that the small molecule activator of AKT (SC79) limited growth of a B16 tumor and an EMT-6 syngeneic breast tumor model that are poorly responsive to PD-1 immunotherapy. In the case of B16 tumors, direct AKT activation induced (i) a reduction of suppressor regulatory (Treg) TILs and (ii) an increase in effector CD8+ TILs. SC79 in vivo therapy caused a major increase in the numbers of CD4+ and CD8+ TILs to express interferon-γ (IFN-γ). This effect on IFN-γ expression distinguished responsive from non-responsive anti-tumor responses and could be recapitulated ex vivo with human T-cells. In CD4+FoxP3+Treg TILs, AKT induced IFN-γ expression was accompanied by a loss of suppressor activity, the conversation to CD4+ helper Th1-like TILs and a marked reduction in phospho-SHP2. In CD8+ TILs, we observed an increase in the phospho-activation of PLC-γ. Further, the genetic deletion of the transcription factor T-bet (Tbx21) blocked the increased IFN-γ expression on all subsets while ablating the therapeutic benefits of SC79 on tumor growth. Our study shows that AKT activation therapy acts to induce IFN-γ on CD4 and CD8 TILs that is accompanied by the intra-tumoral conversation of suppressive Tregs into CD4+Th1-like T-cells and augmented CD8 responses.
Collapse
|
50
|
Yenyuwadee S, Aliazis K, Wang Q, Christofides A, Shah R, Patsoukis N, Boussiotis VA. Immune cellular components and signaling pathways in the tumor microenvironment. Semin Cancer Biol 2022; 86:187-201. [PMID: 35985559 PMCID: PMC10735089 DOI: 10.1016/j.semcancer.2022.08.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Accepted: 08/12/2022] [Indexed: 11/24/2022]
Abstract
During the past decade there has been a revolution in cancer therapeutics by the emergence of antibody-based and cell-based immunotherapies that modulate immune responses against tumors. These new therapies have extended and improved the therapeutic efficacy of chemo-radiotherapy and have offered treatment options to patients who are no longer responding to these classic anti-cancer treatments. Unfortunately, tumor eradication and long-lasting responses are observed in a small fraction of patients, whereas the majority of patients respond only transiently. These outcomes indicate that the maximum potential of immunotherapy has not been reached due to incomplete knowledge of the cellular and molecular mechanisms that guide the development of successful anti-tumor immunity and its failure. In this review, we discuss recent discoveries about the immune cellular composition of the tumor microenvironment (TME) and the role of key signaling mechanisms that compromise the function of immune cells leading to cancer immune escape.
Collapse
Affiliation(s)
- Sasitorn Yenyuwadee
- Division of Hematology-Oncology, Beth Israel Deaconess Medical Center; Department of Medicine Beth Israel Deaconess Medical Center, Harvard Medical School; Department of Dermatology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Konstantinos Aliazis
- Division of Hematology-Oncology, Beth Israel Deaconess Medical Center; Department of Medicine Beth Israel Deaconess Medical Center, Harvard Medical School
| | - Qi Wang
- Division of Hematology-Oncology, Beth Israel Deaconess Medical Center; Department of Medicine Beth Israel Deaconess Medical Center, Harvard Medical School
| | - Anthos Christofides
- Division of Hematology-Oncology, Beth Israel Deaconess Medical Center; Department of Medicine Beth Israel Deaconess Medical Center, Harvard Medical School
| | - Rushil Shah
- Division of Hematology-Oncology, Beth Israel Deaconess Medical Center; Department of Medicine Beth Israel Deaconess Medical Center, Harvard Medical School
| | - Nikolaos Patsoukis
- Division of Hematology-Oncology, Beth Israel Deaconess Medical Center; Department of Medicine Beth Israel Deaconess Medical Center, Harvard Medical School; Cancer Center, Beth Israel Deaconess Medical Center, Harvard Medical School Boston, MA 02215, USA.
| | - Vassiliki A Boussiotis
- Division of Hematology-Oncology, Beth Israel Deaconess Medical Center; Department of Medicine Beth Israel Deaconess Medical Center, Harvard Medical School; Cancer Center, Beth Israel Deaconess Medical Center, Harvard Medical School Boston, MA 02215, USA.
| |
Collapse
|