1
|
Lu Y, Ma Q, Yu L, Liu X, Chen P, Liu W. Circulating CD45RA -Foxp3 ++ Treg cells serve as a biomarker for predicting minimal clinical manifestations status of myasthenia gravis. Life Sci 2024; 358:123162. [PMID: 39433086 DOI: 10.1016/j.lfs.2024.123162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 07/28/2024] [Accepted: 10/17/2024] [Indexed: 10/23/2024]
Abstract
AIMS Regulatory T cells (Tregs) are key mediators of the induction of immune tolerance; however, the mechanisms by which they regulate myasthenia gravis (MG) are not fully understood. This study aimed to explore the characteristics of Tregs and their subpopulations in the peripheral blood of patients with minimal clinical manifestations (MM) of MG and identify biomarkers that predict MM-MG for treatment guidance. MATERIALS AND METHODS The clinical data of patients with general MG who visited our hospital were retrospectively analyzed. Age- and sex-matched volunteers were selected as healthy controls (HC). Flow cytometry was used to determine the proportion, function, and subpopulations of total Tregs. A correlation analysis was conducted for subpopulation proportions and MG disease severity. KEY FINDINGS A total of 27 cases of MM-MG, 40 cases of naїve-MG, and 33 cases of HC were included in this study. The number of total Tregs and the suppressive function of total Tregs were elevated in patients with MM-MG compared to those of patients with naїve-MG. Further analysis revealed that the frequency of CD45RA-Foxp3++ Tregs (a-Tregs) negatively correlated with quantitative myasthenia gravis (QMG) scores for patients with naїve-MG. In addition, the number of a-Tregs was significantly greater in patients with MM-MG than in patients with naїve-MG, and CD45RA-Foxp3+ Tregs expressed higher and lower levels of CTLA-4 and CXCR3, respectively. SIGNIFICANCE CD45RA-Foxp3++ Tregs were significantly more abundant and highly expressed surface inhibitory molecules in patients with MM-MG. This profile may serve as a predictive biomarker for MM-MG.
Collapse
Affiliation(s)
- Yaru Lu
- Department of Neurology, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou 450003, Henan, China; Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, Guangzhou 510080, Guangdong, China
| | - Qian Ma
- Department of Neurology, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai 519000, Guangdong, China
| | - Lu Yu
- Department of Neurology, Guizhou Provincial People's Hospital, Guiyang 550002, China
| | - Xiaoxi Liu
- Department of Neurology, Nanfang Hospital of Southern Medical University, Guangzhou 510515, Guangdong, China
| | - Pei Chen
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, Guangzhou 510080, Guangdong, China
| | - Weibin Liu
- Department of Neurology, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou 450003, Henan, China.
| |
Collapse
|
2
|
Uța C, Tîrziu A, Zimbru EL, Zimbru RI, Georgescu M, Haidar L, Panaitescu C. Alloimmune Causes of Recurrent Pregnancy Loss: Cellular Mechanisms and Overview of Therapeutic Approaches. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:1896. [PMID: 39597081 PMCID: PMC11596804 DOI: 10.3390/medicina60111896] [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: 10/30/2024] [Revised: 11/16/2024] [Accepted: 11/18/2024] [Indexed: 11/29/2024]
Abstract
Recurrent pregnancy loss (RPL) is a complex early pregnancy complication affecting 1-2% of couples and is often linked to immune dysfunction. Aberrations in T and B cell subpopulations, as well as natural killer (NK) cell activity, are particularly influential, with studies showing that abnormal NK cell activation and imbalances in T and B cell subtypes contribute to immune-mediated miscarriage risk. Successful pregnancy requires a tightly regulated balance between pro-inflammatory and anti-inflammatory immune responses. In the early stages, inflammation supports processes such as trophoblast invasion and spiral artery remodeling, but this must be tempered to prevent immune rejection of the fetus. In this review, we explore the underlying immune mechanisms of RPL, focusing on how dysregulated T, B, and NK cell function disrupts maternal tolerance. Specifically, we discuss the essential role of uterine NK cells in the early stages of vascular remodeling in the decidua and regulate the depth of invasion by extravillous trophoblasts. Furthermore, we focus on the delicate Treg dynamics that enable the maintenance of optimal immune homeostasis, where the balance, and not only the quantity of Tregs, is crucial for fostering maternal-fetal tolerance. Other T cell subpopulations, such as Th1, Th2, and Th17 cells, also contribute to immune imbalance, with Th1 and Th17 cells promoting inflammation and potentially harming fetal tolerance, while Th2 cells support immune tolerance. Finally, we show how changes in B cell subpopulations and their functions have been associated with adverse pregnancy outcomes. We further discuss current therapeutic strategies aimed at correcting these immune imbalances, including intravenous immunoglobulin (IVIg), glucocorticoids, and TNF-α inhibitors, examining their efficacy, challenges, and potential side effects. By highlighting both the therapeutic benefits and limitations of these interventions, we aim to offer a balanced perspective on clinical applications for women facing immune-related causes of RPL.
Collapse
Affiliation(s)
- Cristina Uța
- Center of Immuno-Physiology and Biotechnologies, Department of Functional Sciences, “Victor Babeș” University of Medicine and Pharmacy, 2 Eftimie Murgu Square, 300041 Timisoara, Romania; (C.U.); (E.-L.Z.); (R.-I.Z.); (M.G.); (C.P.)
- Timis County Emergency Clinical Hospital “Pius Brinzeu”, 156 Liviu Rebreanu Bd., 300723 Timisoara, Romania
| | - Alexandru Tîrziu
- Center of Immuno-Physiology and Biotechnologies, Department of Functional Sciences, “Victor Babeș” University of Medicine and Pharmacy, 2 Eftimie Murgu Square, 300041 Timisoara, Romania; (C.U.); (E.-L.Z.); (R.-I.Z.); (M.G.); (C.P.)
- Department of Functional Sciences, Physiology Discipline, Faculty of Medicine, “Victor Babeș” University of Medicine and Pharmacy Timișoara, 2 Eftimie Murgu Square, 300041 Timişoara, Romania
- Institute of Cardiovascular Diseases Timisoara, 13A Gheorghe Adam Street, 300310 Timisoara, Romania
| | - Elena-Larisa Zimbru
- Center of Immuno-Physiology and Biotechnologies, Department of Functional Sciences, “Victor Babeș” University of Medicine and Pharmacy, 2 Eftimie Murgu Square, 300041 Timisoara, Romania; (C.U.); (E.-L.Z.); (R.-I.Z.); (M.G.); (C.P.)
- Department of Functional Sciences, Physiology Discipline, Faculty of Medicine, “Victor Babeș” University of Medicine and Pharmacy Timișoara, 2 Eftimie Murgu Square, 300041 Timişoara, Romania
- Research Center for Gene and Cellular Therapies in the Treatment of Cancer—OncoGen, Timis County Emergency Clinical Hospital “Pius Brinzeu”, 156 Liviu Rebreanu Bd., 300723 Timisoara, Romania
| | - Răzvan-Ionuț Zimbru
- Center of Immuno-Physiology and Biotechnologies, Department of Functional Sciences, “Victor Babeș” University of Medicine and Pharmacy, 2 Eftimie Murgu Square, 300041 Timisoara, Romania; (C.U.); (E.-L.Z.); (R.-I.Z.); (M.G.); (C.P.)
- Department of Functional Sciences, Physiology Discipline, Faculty of Medicine, “Victor Babeș” University of Medicine and Pharmacy Timișoara, 2 Eftimie Murgu Square, 300041 Timişoara, Romania
- Research Center for Gene and Cellular Therapies in the Treatment of Cancer—OncoGen, Timis County Emergency Clinical Hospital “Pius Brinzeu”, 156 Liviu Rebreanu Bd., 300723 Timisoara, Romania
| | - Marius Georgescu
- Center of Immuno-Physiology and Biotechnologies, Department of Functional Sciences, “Victor Babeș” University of Medicine and Pharmacy, 2 Eftimie Murgu Square, 300041 Timisoara, Romania; (C.U.); (E.-L.Z.); (R.-I.Z.); (M.G.); (C.P.)
- Department of Functional Sciences, Physiology Discipline, Faculty of Medicine, “Victor Babeș” University of Medicine and Pharmacy Timișoara, 2 Eftimie Murgu Square, 300041 Timişoara, Romania
| | - Laura Haidar
- Center of Immuno-Physiology and Biotechnologies, Department of Functional Sciences, “Victor Babeș” University of Medicine and Pharmacy, 2 Eftimie Murgu Square, 300041 Timisoara, Romania; (C.U.); (E.-L.Z.); (R.-I.Z.); (M.G.); (C.P.)
- Department of Functional Sciences, Physiology Discipline, Faculty of Medicine, “Victor Babeș” University of Medicine and Pharmacy Timișoara, 2 Eftimie Murgu Square, 300041 Timişoara, Romania
| | - Carmen Panaitescu
- Center of Immuno-Physiology and Biotechnologies, Department of Functional Sciences, “Victor Babeș” University of Medicine and Pharmacy, 2 Eftimie Murgu Square, 300041 Timisoara, Romania; (C.U.); (E.-L.Z.); (R.-I.Z.); (M.G.); (C.P.)
- Department of Functional Sciences, Physiology Discipline, Faculty of Medicine, “Victor Babeș” University of Medicine and Pharmacy Timișoara, 2 Eftimie Murgu Square, 300041 Timişoara, Romania
| |
Collapse
|
3
|
Mebrek ML, Abaab T, Lemeiter D, Breckler M, Hervé R, Petit M, Clavel G, Sigaux J, Boissier MC, Semerano L, Biton J, Bessis N. Impairment of regulatory T cell stability in axial spondyloarthritis: role of EZH2 and pSTAT5. Front Immunol 2024; 15:1484321. [PMID: 39569199 PMCID: PMC11576896 DOI: 10.3389/fimmu.2024.1484321] [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: 08/21/2024] [Accepted: 10/10/2024] [Indexed: 11/22/2024] Open
Abstract
Background and objectives Axial spondyloarthritis (axSpA) is a chronic inflammatory disease involving the spine, peripheral joints, and entheses. Functional impairment of regulatory T cells (Treg) is linked to inflammatory diseases, but limited data is available regarding Treg involvement in axSpA. Treg stability refers to their ability to maintain their functions and characteristics in pro-inflammatory environments. EZH2 and phosphorylated STAT5 (pSTAT5) play a critical role in maintaining Treg stability. We aimed to characterize Treg stability in patients with axSpA. Methods Peripheral blood mononuclear cells (PBMCs) from axSpA patients, either naïve from targeted therapy or treated by TNF inhibitors (TNFi), and from healthy donors (HD), were freshly isolated. Expression of stability (EZH2, pSTAT5) and suppressive (TNFR2 and CD39) markers by Treg was analyzed by flow cytometry. Results EZH2 expression by Treg was decreased in axSpA patients as compared to HD (p<0.01). Mechanistic study showed that inhibition of EZH2 attenuated Treg differentiation and suppressive phenotype in vitro. EZH2 was predominantly expressed by highly suppressive TNFR2+ and CD39+ Treg. Additionally, axSpA patients also exhibited a reduced frequency of pSTAT5+ Treg compared to HD (p<0.05), and pSTAT5+ Treg frequency increased at 3 months of TNFi treatment compared to baseline (p<0.05). This last result suggested a restoration of Treg stability upon TNFi treatment. Conclusion By highlighting a deficient expression of EZH2 and pSTAT5 by Treg, we revealed an impaired Treg stability in axSpA. Deciphering the pathways influenced by these molecules is necessary to assess the potential therapeutic benefits of restoring Treg stability in axSpA.
Collapse
Affiliation(s)
- Majda Lyna Mebrek
- UMR 1125 INSERM, Bobigny, France
- Sorbonne Paris Nord University, Bobigny, France
| | - Tessnime Abaab
- UMR 1125 INSERM, Bobigny, France
- Sorbonne Paris Nord University, Bobigny, France
| | - Delphine Lemeiter
- UMR 1125 INSERM, Bobigny, France
- Sorbonne Paris Nord University, Bobigny, France
| | - Magali Breckler
- UMR 1125 INSERM, Bobigny, France
- Sorbonne Paris Nord University, Bobigny, France
| | - Roxane Hervé
- UMR 1125 INSERM, Bobigny, France
- Sorbonne Paris Nord University, Bobigny, France
| | - Mylène Petit
- UMR 1125 INSERM, Bobigny, France
- Sorbonne Paris Nord University, Bobigny, France
- Assistance-Publique Hôpitaux de Paris (AP-HP), GHUPSSD, Department of Rheumatology, Bobigny, France
| | - Gaëlle Clavel
- UMR 1125 INSERM, Bobigny, France
- Sorbonne Paris Nord University, Bobigny, France
- Department of Internal Medicine, Fondation Rothschild, Paris, France
| | - Johanna Sigaux
- UMR 1125 INSERM, Bobigny, France
- Sorbonne Paris Nord University, Bobigny, France
- Assistance-Publique Hôpitaux de Paris (AP-HP), GHUPSSD, Department of Rheumatology, Bobigny, France
| | - Marie-Christophe Boissier
- UMR 1125 INSERM, Bobigny, France
- Sorbonne Paris Nord University, Bobigny, France
- Assistance-Publique Hôpitaux de Paris (AP-HP), GHUPSSD, Department of Rheumatology, Bobigny, France
| | - Luca Semerano
- UMR 1125 INSERM, Bobigny, France
- Sorbonne Paris Nord University, Bobigny, France
- Assistance-Publique Hôpitaux de Paris (AP-HP), GHUPSSD, Department of Rheumatology, Bobigny, France
| | - Jérôme Biton
- UMR 1125 INSERM, Bobigny, France
- Sorbonne Paris Nord University, Bobigny, France
| | - Natacha Bessis
- UMR 1125 INSERM, Bobigny, France
- Sorbonne Paris Nord University, Bobigny, France
| |
Collapse
|
4
|
Yu S, Yan J, Fang Y, Ye Y, Bu B. Effect of thymectomy on the frequencies of peripheral regulatory B and T lymphocytes in patients with Myasthenia gravis-a pilot study. Int J Neurosci 2024; 134:1210-1219. [PMID: 37668142 DOI: 10.1080/00207454.2023.2254922] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Revised: 07/19/2023] [Accepted: 08/29/2023] [Indexed: 09/06/2023]
Abstract
AIM We aimed to investigate the relationship between the peripheral lymphocyte subset frequency and thymectomy in patients with myasthenia gravis (MG). MATERIALS AND METHODS The frequencies of regulatory B (Breg) and regulatory T (Treg) cells in peripheral blood samples obtained from 69 patients with MG and 10 healthy controls were analyzed using flow cytometry. Serum acetylcholine receptor antibodies (AchR-Ab) were measured. Patients with MG were subdivided into pre-thymectomy, post-thymectomy, and normal thymus control group. RESULTS The percentage of Breg cells was significantly decreased in both the pre-thymectomy (7.92 ± 1.30%) and post-thymectomy (8.14 ± 1.34%) groups compared to healthy controls (16.02 ± 2.78%) and reduced in the exacerbation and relapse phase compared to the stable maintenance stage. The proportion of cluster of differentiation (CD) 4 + CD25 + T cells and CD4 + CD25 + CD127low/- Treg cells in MG patients were not significantly different than healthy controls. AchR-Ab titers in aggravating or recurrence patients after thymectomy were significantly higher than that of the stable remission patients (11.13 ± 0.70 and 6.03 ± 0.85 nmol/L, respectively; p < 0.001). CONCLUSION The frequency of Breg cells may serve as a potential indicator of MG prognosis, while Treg cell frequency did not demonstrate the same prognostic ability. The concentration of AchR-Ab can be used as a dynamic monitoring index of disease severity in patients with MG.
Collapse
Affiliation(s)
- Shanshan Yu
- Department of Emergency, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jingjun Yan
- Department of Emergency, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yu Fang
- Department of Emergency, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yan Ye
- Department of Emergency, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Bitao Bu
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
5
|
Qiu Y, Xie E, Xu H, Cheng H, Li G. One-carbon metabolism shapes T cell immunity in cancer. Trends Endocrinol Metab 2024; 35:967-980. [PMID: 38925992 DOI: 10.1016/j.tem.2024.05.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 05/30/2024] [Accepted: 05/31/2024] [Indexed: 06/28/2024]
Abstract
One-carbon metabolism (1CM), comprising folate metabolism and methionine metabolism, serves as an important mechanism for cellular energy provision and the production of vital signaling molecules, including single-carbon moieties. Its regulation is instrumental in sustaining the proliferation of cancer cells and facilitating metastasis; in addition, recent research has shed light on its impact on the efficacy of T cell-mediated immunotherapy. In this review, we consolidate current insights into how 1CM affects T cell activation, differentiation, and functionality. Furthermore, we delve into the strategies for modulating 1CM in both T cells and tumor cells to enhance the efficacy of adoptively transferred T cells, overcome metabolic challenges in the tumor microenvironment (TME), and maximize the benefits of T cell-mediated immunotherapy.
Collapse
Affiliation(s)
- Yajing Qiu
- National Key Laboratory of Immunity and Inflammation, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, 215123, Jiangsu, China; Key Laboratory of Synthetic Biology Regulatory Elements, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, 215123, Jiangsu, China
| | - Ermei Xie
- National Key Laboratory of Immunity and Inflammation, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, 215123, Jiangsu, China; Key Laboratory of Synthetic Biology Regulatory Elements, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, 215123, Jiangsu, China
| | - Haipeng Xu
- Fujian Medical University Cancer Hospital, Fujian Cancer Hospital, Fujian, 350011, China
| | - Hongcheng Cheng
- National Key Laboratory of Immunity and Inflammation, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, 215123, Jiangsu, China; Key Laboratory of Synthetic Biology Regulatory Elements, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, 215123, Jiangsu, China.
| | - Guideng Li
- National Key Laboratory of Immunity and Inflammation, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, 215123, Jiangsu, China; Key Laboratory of Synthetic Biology Regulatory Elements, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, 215123, Jiangsu, China.
| |
Collapse
|
6
|
Cui Y, Hackett RG, Ascue J, Muralidaran V, Patil D, Kang J, Kaufman SS, Khan K, Kroemer A. Innate and Adaptive Immune Responses in Intestinal Transplant Rejection: Through the Lens of Inflammatory Bowel and Intestinal Graft-Versus-Host Diseases. Gastroenterol Clin North Am 2024; 53:359-382. [PMID: 39068000 DOI: 10.1016/j.gtc.2024.01.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/30/2024]
Abstract
Intestinal transplantation is a life-saving procedure utilized for patients failing total parenteral nutrition. However, intestinal transplantattion remains plagued with low survival rates and high risk of allograft rejection. The authors explore roles of innate (macrophages, natural killer cells, innate lymphoid cells) and adaptive immune cells (Th1, Th2, Th17, Tregs) in inflammatory responses, particularly inflammatory bowel disease and graft versus host disease, and correlate these findings to intestinal allograft rejection, highlighting which effectors exacerbate or suppress intestinal rejection. Better understanding of this immunology can open further investigation into potential biomolecular targets to develop improved therapeutic treatment options and immunomonitoring techniques to combat allograft rejection and enhance patient lives.
Collapse
Affiliation(s)
- Yuki Cui
- MedStar Georgetown Transplant Institute, MedStar Georgetown University Hospital and the Center for Translational Transplant Medicine, Georgetown University Medical Center, Washington, DC, USA
| | - Ryan G Hackett
- MedStar Georgetown Transplant Institute, MedStar Georgetown University Hospital and the Center for Translational Transplant Medicine, Georgetown University Medical Center, Washington, DC, USA
| | - Jhalen Ascue
- MedStar Georgetown Transplant Institute, MedStar Georgetown University Hospital and the Center for Translational Transplant Medicine, Georgetown University Medical Center, Washington, DC, USA
| | - Vinona Muralidaran
- MedStar Georgetown Transplant Institute, MedStar Georgetown University Hospital and the Center for Translational Transplant Medicine, Georgetown University Medical Center, Washington, DC, USA
| | - Digvijay Patil
- MedStar Georgetown Transplant Institute, MedStar Georgetown University Hospital and the Center for Translational Transplant Medicine, Georgetown University Medical Center, Washington, DC, USA
| | - Jiman Kang
- MedStar Georgetown Transplant Institute, MedStar Georgetown University Hospital and the Center for Translational Transplant Medicine, Georgetown University Medical Center, Washington, DC, USA; Department of Biochemistry and Molecular & Cellular Biology, Georgetown University, Washington, DC, USA
| | - Stuart S Kaufman
- MedStar Georgetown Transplant Institute, MedStar Georgetown University Hospital and the Center for Translational Transplant Medicine, Georgetown University Medical Center, Washington, DC, USA
| | - Khalid Khan
- MedStar Georgetown Transplant Institute, MedStar Georgetown University Hospital and the Center for Translational Transplant Medicine, Georgetown University Medical Center, Washington, DC, USA
| | - Alexander Kroemer
- MedStar Georgetown Transplant Institute, MedStar Georgetown University Hospital and the Center for Translational Transplant Medicine, Georgetown University Medical Center, Washington, DC, USA.
| |
Collapse
|
7
|
Vakrakou AG, Kourepini E, Skordos I, Nieto N, Panoutsakopoulou V, Paschalidis N. Osteopontin Regulates Treg Cell Stability and Function with Implications for Anti-Tumor Immunity and Autoimmunity. Cancers (Basel) 2024; 16:2952. [PMID: 39272810 PMCID: PMC11393878 DOI: 10.3390/cancers16172952] [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: 07/29/2024] [Revised: 08/16/2024] [Accepted: 08/20/2024] [Indexed: 09/15/2024] Open
Abstract
Foxp3-expressing regulatory T (Treg) cells represent the most highly immunosuppressive cell in the tumor microenvironment (TME) that halts effective anti-tumor immunity. Osteopontin (Opn), an extracellular matrix (ECM) glycophosphoprotein, plays key roles in many types of immune-related diseases and is associated with cancer aggressiveness when expressed by tumor cells. However, its role in Foxp3Treg heterogeneity, function, and stability in the TME is poorly defined. We generated mice with a Foxp3-specific deletion of Opn and assessed the ability of Opn-deficient Tregs to suppress inflammation. As these mice aged, they developed a scurfy-like syndrome characterized by aberrant and excessive activation of effector T cells. We evaluated and further confirmed the reduced suppressive capacity of Opn-deficient Tregs in an in vivo suppression assay of colitis. We also found that mice with Opn-deficient Foxp3+ Tregs have enhanced anti-tumor immunity and reduced tumor burden, associated with an unstable Treg phenotype, paralleled by reduced Foxp3 expression in tumor-infiltrating lymphocytes. Finally, we observed reduced Foxp3 and Helios expression in Opn-deficient Tregs compared to wild-type controls after in vitro activation. Our findings indicate that targeting Opn in Tregs reveals vigorous and effective ways of promoting Treg instability and dysfunction in the TME, facilitating anti-tumor immunity.
Collapse
Affiliation(s)
- Aigli G Vakrakou
- Laboratory of Neuroimmunology, First Department of Neurology, Aeginition Hospital, National and Kapodistrian, University of Athens, 21 Papadiamantopoulou, Ilisia, 11528 Athens, Greece
| | - Evangelia Kourepini
- Biomedical Research Foundation, Academy of Athens, 4 Soranou Efessiou Street, 11527 Athens, Greece
| | - Ioannis Skordos
- Biomedical Research Foundation, Academy of Athens, 4 Soranou Efessiou Street, 11527 Athens, Greece
| | - Natalia Nieto
- Department of Pathology, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Vily Panoutsakopoulou
- Biomedical Research Foundation, Academy of Athens, 4 Soranou Efessiou Street, 11527 Athens, Greece
| | - Nikolaos Paschalidis
- Biomedical Research Foundation, Academy of Athens, 4 Soranou Efessiou Street, 11527 Athens, Greece
| |
Collapse
|
8
|
Wang M, Wang S, Yuan G, Gao M, Zhao X, Chu Z, Gao D. Causal role of immune cells in bipolar disorder: a Mendelian randomization study. Front Psychiatry 2024; 15:1411280. [PMID: 39220183 PMCID: PMC11362081 DOI: 10.3389/fpsyt.2024.1411280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Accepted: 07/30/2024] [Indexed: 09/04/2024] Open
Abstract
Background The understanding of the immunological mechanisms underlying bipolar disorder (BD) has enhanced in recent years due to the extensive use of high-density genetic markers for genotyping and advancements in genome-wide association studies (GWAS). However, studies on the relationship between immune cells and the risk of BD remain limited, necessitating further investigation. Methods Bidirectional two-sample Mendelian Randomization (MR) analysis was employed to investigate the causal association between immune cell morphologies and bipolar disorder. Immune cell traits were collected from a research cohort in Sardinia, whereas the GWAS summary statistics for BD were obtained from the Psychiatric Genomics Consortium. Sensitivity analyses were conducted, and the combination of MR-Egger and MR-Presso was used to assess horizontal pleiotropy. Cochran's Q test was employed to evaluate heterogeneity, and the results were adjusted for false discovery rate (FDR). Results The study identified six immune cell phenotypes significantly associated with BD incidence (P< 0.01). These phenotypes include IgD- CD27- %lymphocyte, CD33br HLA DR+ CD14- AC, CD8 on CD28+ CD45RA+ CD8br, CD33br HLA DR+ AC, CD14 on CD14+ CD16+ monocyte, and HVEM on CD45RA- CD4+. After adjusting the FDR to 0.2, two immune cell phenotypes remained statistically significant: IgD-CD27-% lymphocyte (OR=1.099, 95% CI: 1.051-1.149, P = 3.51E-05, FDR=0.026) and CD33br HLA DR+ CD14-AC (OR=0.981, 95% CI: 0.971-0.991, P = 2.17E-04, FDR=0.079). In the reverse MR analysis, BD significantly impacted the phenotypes of four monocytes (P< 0.01), including CD64 on CD14+ CD16+ monocyte, CD64 on monocyte, CX3CR1 on CD14- CD16-, CD64 on CD14+ CD16- monocyte. However, after applying the FDR correction (FDR < 0.2), no statistically significant results were observed. Conclusions This MR investigation reveals associations between immune cell phenotypes, bipolar disorder, and genetics, providing novel perspectives on prospective therapeutic targets for bipolar disorder.
Collapse
Affiliation(s)
- Mengxuan Wang
- Department of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Shuo Wang
- Department of Intelligent and Information Engineering, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Guoshan Yuan
- Department of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Mingzhou Gao
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xiyan Zhao
- Department of Foreign Studies, China University of Petroleum (East China), Qingdao, China
| | - Zhenhan Chu
- Department of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Dongmei Gao
- Department of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
9
|
Zhang C, Liu X, Gu C, Su Y, Lv J, Liu Y, Gao Y, Chen H, Xu N, Xiao J, Xu Z, Su W. Histone deacetylases facilitate Th17-cell differentiation and pathogenicity in autoimmune uveitis via CDK6/ID2 axis. J Adv Res 2024:S2090-1232(24)00313-8. [PMID: 39107200 DOI: 10.1016/j.jare.2024.07.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Revised: 07/25/2024] [Accepted: 07/28/2024] [Indexed: 08/09/2024] Open
Abstract
INTRODUCTION Autoimmune uveitis (AU) is a prevalent ocular autoimmune disease leading to significant visual impairment. However, underlying pathogenesis of AU required to develop more efficient therapy remain unclear. METHODS We isolated peripheral blood mononuclear cells (PBMCs) from AU patients and performed single-cell RNA sequencing (scRNA-seq). Besides, experimental autoimmune uveitis (EAU) model was established and treated with histone deacetylase inhibitor (HDACi) Belinostat or vehicle. We extracted immune cells from Blank, EAU, and HDACi-treated EAU mice and used scRNA-seq, flow cytometry, siRNA, specific inhibitors, and adoptive transfer experiments to explore the role of HDACs and its downstream potential molecular mechanisms in the immune response of EAU and AU. RESULTS We found highly expressed histone deacetylases (HDACs) family in AU patients and identified it as a key factor related to CD4+ effector T cell differentiation in the pathogenesis of AU. Our further studies showed that targeted inhibition of HDACs effectively alleviated EAU, restored its Th17/Treg balance, and reduced inflammatory gene expression, especially in CD4+ T cells. Post-HDACs inhibition, Treg proportions increased with enhanced immunomodulatory effects. Importantly, HDACs exhibited a positive promoting role on Th17 cells. Based on scRNA-seq screening and application of knock-down siRNAs and specific inhibitors in vitro and vivo, we identified CDK6 as a key downstream molecule regulated by HDAC1/3/6 through acetyl-histone H3/p53/p21 axis, which is involved in Th17 pathogenicity and EAU development. Additionally, HDACs-regulated CDK6 formed a positive loop with ID2, inducing PIM1 upregulation, promoting Th17 cell differentiation and pathogenicity, and correlates with AU progression. CONCLUSION Based on the screening of clinical samples and downstream molecular functional validation experiments, we revealed a driving role for HDACs and the HDACs-regulated CDK6/ID2 axis in Th17 cell differentiation and pathogenicity in AU, proposing a promising therapeutic strategy.
Collapse
Affiliation(s)
- Chun Zhang
- Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Xiuxing Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, China.
| | - Chenyang Gu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, China
| | - Yuhan Su
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, China; Department of Clinical Medicine, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou 510060, China
| | - Jianjie Lv
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, China
| | - Yidan Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, China
| | - Yuehan Gao
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, China
| | - Hui Chen
- Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Nanwei Xu
- Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China; Clinical Medicine (Eight-Year Program), West China School of Medicine, Sichuan University, Chengdu 610044, China
| | - Jing Xiao
- Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Zhuping Xu
- Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China.
| | - Wenru Su
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200001, China; State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, China.
| |
Collapse
|
10
|
Yue Y, Ren Y, Lu C, Li P, Zhang G. Epigenetic regulation of human FOXP3+ Tregs: from homeostasis maintenance to pathogen defense. Front Immunol 2024; 15:1444533. [PMID: 39144146 PMCID: PMC11323565 DOI: 10.3389/fimmu.2024.1444533] [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: 06/05/2024] [Accepted: 07/15/2024] [Indexed: 08/16/2024] Open
Abstract
Regulatory T cells (Tregs), characterized by the expression of Forkhead Box P3 (FOXP3), constitute a distinct subset of T cells crucial for immune regulation. Tregs can exert direct and indirect control over immune homeostasis by releasing inhibitory factors or differentiating into Th-like Treg (Th-Treg), thereby actively contributing to the prevention and treatment of autoimmune diseases. The epigenetic regulation of FOXP3, encompassing DNA methylation, histone modifications, and post-translational modifications, governs the development and optimal suppressive function of Tregs. In addition, Tregs can also possess the ability to maintain homeostasis in diverse microenvironments through non-suppressive mechanisms. In this review, we primarily focus on elucidating the epigenetic regulation of Tregs as well as their multifaceted roles within diverse physiological contexts while looking forward to potential strategies involving augmentation or suppression of Tregs activity for disease management, particularly in light of the ongoing global COVID-19 pandemic.
Collapse
Affiliation(s)
| | | | | | | | - Guojun Zhang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
11
|
Blinova VG, Zhdanov DD. Many Faces of Regulatory T Cells: Heterogeneity or Plasticity? Cells 2024; 13:959. [PMID: 38891091 PMCID: PMC11171907 DOI: 10.3390/cells13110959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 05/21/2024] [Accepted: 05/22/2024] [Indexed: 06/21/2024] Open
Abstract
Regulatory T cells (Tregs) are essential for maintaining the immune balance in normal and pathological conditions. In autoimmune diseases and transplantation, they restrain the loss of self-tolerance and promote engraftment, whereas in cancer, an increase in Treg numbers is mostly associated with tumor growth and poor prognosis. Numerous markers and their combinations have been used to identify Treg subsets, demonstrating the phenotypic diversity of Tregs. The complexity of Treg identification can be hampered by the unstable expression of some markers, the decrease in the expression of a specific marker over time or the emergence of a new marker. It remains unclear whether such phenotypic shifts are due to new conditions or whether the observed changes are due to initially different populations. In the first case, cellular plasticity is observed, whereas in the second, cellular heterogeneity is observed. The difference between these terms in relation to Tregs is rather blurred. Considering the promising perspectives of Tregs in regenerative cell-based therapy, the existing confusing data on Treg phenotypes require further investigation and analysis. In our review, we introduce criteria that allow us to distinguish between the heterogeneity and plasticity of Tregs normally and pathologically, taking a closer look at their diversity and drawing the line between two terms.
Collapse
Affiliation(s)
- Varvara G. Blinova
- Laboratory of Medical Biotechnology, Institute of Biomedical Chemistry, Pogodinskaya st. 10/8, 119121 Moscow, Russia;
| | - Dmitry D. Zhdanov
- Laboratory of Medical Biotechnology, Institute of Biomedical Chemistry, Pogodinskaya st. 10/8, 119121 Moscow, Russia;
- Department of Biochemistry, People’s Friendship University of Russia Named after Patrice Lumumba (RUDN University), Miklukho-Maklaya st. 6, 117198 Moscow, Russia
| |
Collapse
|
12
|
Kommoju V, Mariaselvam CM, Bulusu SN, Michael BNR, Kavadichanda C, Thabah MM, Negi VS. Rheumatoid arthritis autologous synovial fluid affects the plasticity and function of peripheral and induced T regulatory cells in vitro. Immunol Lett 2024; 267:106859. [PMID: 38670465 DOI: 10.1016/j.imlet.2024.106859] [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: 08/24/2023] [Revised: 03/01/2024] [Accepted: 04/23/2024] [Indexed: 04/28/2024]
Abstract
The synovial fluid (SF) microenvironment in rheumatoid arthritis (RA) may alter the stability and function of Tregs. In the present study, we assessed cytokine levels and percentage of Tregs, Tregs expressing CXCR3 (Th1-like Treg), CCR6 (Th17-like Treg) in RA peripheral blood (PB) and RA-SF using fluorescence cytometry. Effect of autologous SF on plasticity and function of RA-PB Tregs (pTregs; CD4+CD25hiCD127Lo/-) and induced vimentin-pulsed Tregs (iTregsVIM) was assessed in vitro. Cytokines and percentage of Th1-like and Th17-like Tregs were higher in RA-PB than OA-PB; higher in RA-SF than osteoarthritis (OA)-SF. Compared to OA-SF exposed OA-pTregs, RA-SF exposed RA-pTregs showed higher percentage of Th1-like (11% vs 20%) and Th17-like (16% vs 36%) Tregs; higher T-bet (p = 0.0001), RORγ (p = 0.0001) and lower FOXP3 (p = 0.0001) gene expression; and diminished percentage suppression of autologous T effector cells (36% vs 74%). RA-SF exposed iTregsVIM showed increased percentage of Th1-like and Th17-like Tregs compared to iTregsVIM exposed to AB serum (8% vs 0.1%; 21% vs 0.1%). IL-2, Tocilizumab and 5-azacytidine reduced the conversion of iTregsVIM (8% vs 2.4%; 21% vs 6.9%), when used in combination. To conclude, microenvironment in the RA synovial fluid is possibly responsible for conversion of pTregs into Th-like Tregs and their functional loss. A blockade of cytokine receptors and methyl transferases could inhibit Tregs conversion, providing clinical relevance for future Tregs targeting therapies.
Collapse
Affiliation(s)
- Vallayyachari Kommoju
- Department of Clinical Immunology, Jawaharlal Institute of Postgraduate Medical Education and Research (JIPMER), Pondicherry 605 006, India
| | - Christina Mary Mariaselvam
- Department of Clinical Immunology, Jawaharlal Institute of Postgraduate Medical Education and Research (JIPMER), Pondicherry 605 006, India
| | - Sree Nethra Bulusu
- Department of Clinical Immunology, Jawaharlal Institute of Postgraduate Medical Education and Research (JIPMER), Pondicherry 605 006, India
| | - Benita Nancy Reni Michael
- Department of Clinical Immunology, Jawaharlal Institute of Postgraduate Medical Education and Research (JIPMER), Pondicherry 605 006, India
| | - Chengappa Kavadichanda
- Department of Clinical Immunology, Jawaharlal Institute of Postgraduate Medical Education and Research (JIPMER), Pondicherry 605 006, India
| | - Molly Mary Thabah
- Department of Clinical Immunology, Jawaharlal Institute of Postgraduate Medical Education and Research (JIPMER), Pondicherry 605 006, India
| | - Vir Singh Negi
- Department of Clinical Immunology, Jawaharlal Institute of Postgraduate Medical Education and Research (JIPMER), Pondicherry 605 006, India; All India Institute of Medical Sciences (AIIMS), Bilaspur, Himachal Pradesh 174001, India.
| |
Collapse
|
13
|
Kommoju V, Mariaselvam CM, Bulusu SN, Ganapathy CK, Narasimhan PB, Thabah MM, Negi VS. Conventional Tregs in treatment-naïve rheumatoid arthritis are deficient in suppressive function with an increase in percentage of CXCR3 and CCR6 expressing Tregs. Immunol Res 2024; 72:396-408. [PMID: 38151700 DOI: 10.1007/s12026-023-09444-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Accepted: 11/26/2023] [Indexed: 12/29/2023]
Abstract
In rheumatoid arthritis (RA), immune homeostasis is maintained by T regulatory cells (Tregs) that in an inflammatory milieu can change towards T-helper-like phenotypes (Th-like Tregs). Our aim was to examine the phenotypic and functional characteristics of CD4+CD25+CD127lo/- Tregs, Th-like Tregs and T effector (Teff) cells in the peripheral blood (PB) and synovial fluid (SF) of treatment-naïve early RA, as compared to osteoarthritis (OA) and healthy control (HC) peripheral blood. Frequencies of Tregs, CXCR3, CCR6 expressing Tregs (Th-like Tregs), and Teff cells were analyzed using flow cytometry in RA (n = 80), OA (n = 20), and HC (n = 40). Cytokine concentrations of the respective T cell subsets in plasma and SF were measured using flow cytometric bead array. Tregs sorted from RA and HC PB using magnetic beads were analyzed for functional capacities by CFSE proliferation assay and FOXP3 gene expression using real-time PCR. We observed that the frequencies of Th17 cells in PB and SF were significantly higher in RA when compared to HC, whereas Tregs were lower in PB and high in SF compared to HC and OA respectively. Th1- and Th17-related pro-inflammatory cytokines IL12p70, INF-γ, TNF-α, and IL-6, and IL-17A were significantly higher in the plasma and SF of RA. Tregs expressing CXCR3 (Th1-like Tregs) and CCR6 (Th17-like Treg) were significantly higher in PB and SF of RA compared to controls and was positively associated with seropositivity and disease activity. Treg cells isolated from peripheral blood of RA showed decreased function and reduced FOXP3 gene expression compared to HC. In our study, we have demonstrated higher frequencies of Th1 and Th17 cells and increased circulatory and SF pro-inflammatory cytokines (IL12P70, INF-γ, IL-6, IL-17A, and TNF-α) in RA. This inflammatory milieu might alter total Tregs frequencies and influence conversion of Tregs into Th-like Tregs.
Collapse
Affiliation(s)
- Vallayyachari Kommoju
- Department of Clinical Immunology, Jawaharlal Institute of Postgraduate Medical Education and Research (JIPMER), Pondicherry, 605 006, India
| | - Christina Mary Mariaselvam
- Department of Clinical Immunology, Jawaharlal Institute of Postgraduate Medical Education and Research (JIPMER), Pondicherry, 605 006, India
| | - Sree Nethra Bulusu
- Department of Clinical Immunology, Jawaharlal Institute of Postgraduate Medical Education and Research (JIPMER), Pondicherry, 605 006, India
| | - Chengappa Kavadichanda Ganapathy
- Department of Clinical Immunology, Jawaharlal Institute of Postgraduate Medical Education and Research (JIPMER), Pondicherry, 605 006, India
| | - Prakash Babu Narasimhan
- Department of Clinical Immunology, Jawaharlal Institute of Postgraduate Medical Education and Research (JIPMER), Pondicherry, 605 006, India
| | - Molly Mary Thabah
- Department of Clinical Immunology, Jawaharlal Institute of Postgraduate Medical Education and Research (JIPMER), Pondicherry, 605 006, India
| | - Vir Singh Negi
- Department of Clinical Immunology, Jawaharlal Institute of Postgraduate Medical Education and Research (JIPMER), Pondicherry, 605 006, India.
- All India Institute of Medical Sciences, Bilaspur, Himachal Pradesh, India.
| |
Collapse
|
14
|
Li J, Cheng C, Zhang J. Autoimmune diseases and the risk of bladder cancer: A Mendelian randomization analysis. J Autoimmun 2024; 146:103231. [PMID: 38692170 DOI: 10.1016/j.jaut.2024.103231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 03/25/2024] [Accepted: 04/16/2024] [Indexed: 05/03/2024]
Abstract
OBJECTIVE To investigate the association between autoimmune diseases (AIDs) and bladder cancer (BC) at the genetic level using Mendelian randomization (MR). METHODS Single nucleotide polymorphisms (SNPs) associated with the seven AIDs were extracted from the IEU GWAS database, and the SNPs were quality-controlled using strict screening criteria. The association between AIDs and BC risk was assessed by inverse-variance weighted (IVW), MR-Egger regression and Weighted median method. The heterogeneity of SNPs was evaluated by Cochran Q test. MR-Egger intercept test and MR-PRESSO global test were used to test the horizontal pleiotropy of SNPs. Both sides with potential causal associations were validated using the validation set. RESULTS Our result showed that genetically predicted RA was significantly associated with an increased risk of BC (IVW OR = 1.214, 95 % CI = 1.062-1.388, P = 0.005). MS nominally increased the risk of BC (IVW OR = 1.095, 95 % CI = 1.005-1.193, P = 0.037), consistent with the results of the MR analysis of the BC validation cohort. However SLE, T1D, UC, CD, and MG were not causally associated with BC risk (P > 0.05). The sensitivity analyses showed that there was no heterogeneity or horizontal pleiotropy in our findings. CONCLUSION This study provides evidence of a causal relationship between AIDs and BC risk at the genetic level, confirming a causal relationship between RA and MS in increasing the risk of BC.
Collapse
Affiliation(s)
- Jun Li
- Department of Urology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, China
| | - Cui Cheng
- Department of Urology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, China
| | - Jiajun Zhang
- Department of Urology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, China.
| |
Collapse
|
15
|
Wang Y, Yang H, Hu J, Jiang Y, Ma W, Gao S, Chen D. Preparation and application of fluorescent monoclonal antibodies recognizing goat CD4 +CD25 + regulatory T cells. Appl Microbiol Biotechnol 2024; 108:327. [PMID: 38717623 PMCID: PMC11078799 DOI: 10.1007/s00253-024-13115-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 03/04/2024] [Accepted: 03/19/2024] [Indexed: 05/12/2024]
Abstract
Regulatory T cells (Tregs) are a subset of T cells participating in a variety of diseases including mycoplasmal pneumonia, contagious ecthyma, and so on. The role of Tregs in goat contagious ecthyma is not completely understood due to the lack of species-specific antibodies. Here, we developed a combination of CD4 and CD25 fluorescence monoclonal antibodies (mAb) to recognize goat Tregs and assessed its utility in flow cytometry, immunofluorescence staining. Using immunofluorescence staining, we found that the frequency of Treg cells was positively correlated with the viral load during orf virus infection. These antibodies could serve as important tools to monitor Tregs during orf virus infection in goats. KEY POINTS: • A combination of fluorescent mAbs (C11 and D12) was prepared for the detection of goat Tregs. • C11 and D12 are effective in flow cytometry, immunofluorescence staining, and C11 has excellent species specificity. • The frequency of Treg cells was positively correlated with the viral load during orf virus infection.
Collapse
Affiliation(s)
- Yunpeng Wang
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Haoyue Yang
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Jiajin Hu
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Yuecai Jiang
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Wentao Ma
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China.
| | - Shikong Gao
- Shenmu Animal Husbandry Development Center, Shenmu, 719300, Shaanxi, China.
| | - Dekun Chen
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China.
| |
Collapse
|
16
|
Wood S, Branch J, Vasquez P, DeGuzman MM, Brown A, Sagcal-Gironella AC, Singla S, Ramirez A, Vogel TP. Th17/1 and ex-Th17 cells are detected in patients with polyarticular juvenile arthritis and increase following treatment. Pediatr Rheumatol Online J 2024; 22:32. [PMID: 38431635 PMCID: PMC10908086 DOI: 10.1186/s12969-024-00965-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 02/11/2024] [Indexed: 03/05/2024] Open
Abstract
BACKGROUND A better understanding of the pathogenesis of polyarticular juvenile idiopathic arthritis (polyJIA) is needed to aide in the development of data-driven approaches to guide selection between therapeutic options. One inflammatory pathway of interest is JAK-STAT signaling. STAT3 is a transcription factor critical to the differentiation of inflammatory T helper 17 cells (Th17s). Previous studies have demonstrated increased STAT3 activation in adult patients with rheumatoid arthritis, but less is known about STAT3 activation in polyJIA. We hypothesized that Th17 cells and STAT3 activation would be increased in treatment-naïve polyJIA patients compared to pediatric controls. METHODS Blood from 17 patients with polyJIA was collected at initial diagnosis and again if remission was achieved (post-treatment). Pediatric healthy controls were also collected. Peripheral blood mononuclear cells were isolated and CD4 + T cell subsets and STAT activation (phosphorylation) were evaluated using flow cytometry. Data were analyzed using Mann-Whitney U and Wilcoxon matched-pairs signed rank tests. RESULTS Treatment-naïve polyJIA patients had increased Th17 cells (CD3 + CD4 + interleukin(IL)-17 +) compared to controls (0.15% v 0.44%, p < 0.05), but Tregs (CD3 + CD4 + CD25 + FOXP3 +) from patients did not differ from controls. Changes in STAT3 phosphorylation in CD4 + T cells following ex vivo stimulation were not significantly different in patients compared to controls. We identified dual IL-17 + and interferon (IFN)γ + expressing CD4 + T cells in patients, but not controls. Further, both Th17/1 s (CCR6 + CD161 + IFNγ + IL-17 +) and ex-Th17s (CCR6 + CD161 + IFNγ + IL-17neg) were increased in patients' post-treatment (Th17/1: 0.3% v 0.07%, p < 0.05 and ex-Th17s: 2.3% v 1.4%, p < 0.05). The patients with the highest IL-17 expressing cells post-treatment remained therapy-bound. CONCLUSIONS Patients with polyJIA have increased baseline Th17 cells, potentially reflecting higher tonic STAT3 activation in vivo. These quantifiable immune markers may identify patients that would benefit upfront from pathway-focused biologic therapies. Our data also suggest that inflammatory CD4 + T cell subsets not detected in controls but increased in post-treatment samples should be further evaluated as a tool to stratify patients in remission on medication. Future work will explore these proposed diagnostic and prognostic biomarkers.
Collapse
Affiliation(s)
- Stephanie Wood
- Division of Rheumatology, Department of Pediatrics, Baylor College of Medicine and Texas Children's Hospital, 1102 Bates Street Suite 330, Houston, TX, 77030, USA
- Center for Human Immunobiology, Texas Children's Hospital, 1102 Bates Street Suite 330, Houston, TX, 77030, USA
| | - Justin Branch
- Division of Rheumatology, Department of Pediatrics, Baylor College of Medicine and Texas Children's Hospital, 1102 Bates Street Suite 330, Houston, TX, 77030, USA
- Center for Human Immunobiology, Texas Children's Hospital, 1102 Bates Street Suite 330, Houston, TX, 77030, USA
| | - Priscilla Vasquez
- Division of Rheumatology, Department of Pediatrics, Baylor College of Medicine and Texas Children's Hospital, 1102 Bates Street Suite 330, Houston, TX, 77030, USA
| | - Marietta M DeGuzman
- Division of Rheumatology, Department of Pediatrics, Baylor College of Medicine and Texas Children's Hospital, 1102 Bates Street Suite 330, Houston, TX, 77030, USA
| | - Amanda Brown
- Division of Rheumatology, Department of Pediatrics, Baylor College of Medicine and Texas Children's Hospital, 1102 Bates Street Suite 330, Houston, TX, 77030, USA
| | - Anna Carmela Sagcal-Gironella
- Division of Rheumatology, Department of Pediatrics, Baylor College of Medicine and Texas Children's Hospital, 1102 Bates Street Suite 330, Houston, TX, 77030, USA
| | - Saimun Singla
- Division of Rheumatology, Department of Pediatrics, Baylor College of Medicine and Texas Children's Hospital, 1102 Bates Street Suite 330, Houston, TX, 77030, USA
| | - Andrea Ramirez
- Division of Rheumatology, Department of Pediatrics, Baylor College of Medicine and Texas Children's Hospital, 1102 Bates Street Suite 330, Houston, TX, 77030, USA
| | - Tiphanie P Vogel
- Division of Rheumatology, Department of Pediatrics, Baylor College of Medicine and Texas Children's Hospital, 1102 Bates Street Suite 330, Houston, TX, 77030, USA.
- Center for Human Immunobiology, Texas Children's Hospital, 1102 Bates Street Suite 330, Houston, TX, 77030, USA.
| |
Collapse
|
17
|
Wang Q, Fang X, Sun B, Zhu K, Yao M, Wei S, Zhang A. Rosa roxburghii Tratt juice inhibits NF-κB and increases IL-2 to alleviates the Foxp3-mediated Tregs imbalance in the peripheral blood of arseniasis patients. Food Sci Biotechnol 2024; 33:935-944. [PMID: 38371687 PMCID: PMC10866849 DOI: 10.1007/s10068-023-01384-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 06/19/2023] [Accepted: 07/06/2023] [Indexed: 02/20/2024] Open
Abstract
Arsenic can cause immune inflammation, which is the basis of arsenic-induced damage to multiple organs and systems. Forkhead box P3 (Foxp3)-labelled CD4+CD25+ regulatory T cells (Tregs) play an essential role in maintaining immune homeostasis. Nuclear factor-κb (NF-κB) and Interleukin-2 (IL-2) are critical regulators of Foxp3. Rosa roxburghii Tratt (RRT) is an edible medicinal plant with anti-inflammation effects. In this study, a control group (n = 41) and an arseniasis group (n = 209) were recruited, and screened subjects from the arseniasis patients for RRTJ (n = 46) or placebo (n = 43) to explore the possible mechanism by which RRT alleviates immune inflammation. The results indicated that RRTJ can inhibits NF-κB and increases IL-2, and alleviates the Foxp3-mediated Tregs imbalance in the peripheral blood of arseniasis patients. In summary, these findings suggest a novel intervention or therapeutic target for immune inflammation in arseniasis patients and provide new evidence that RRTJ inhibits immune inflammation. Graphical abstract Supplementary Information The online version contains supplementary material available at 10.1007/s10068-023-01384-0.
Collapse
Affiliation(s)
- Qi Wang
- The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Department of Toxicology, School of Public Health, Guizhou Medical University, Guiyang, 550025 Guizhou People’s Republic of China
| | - Xiaolin Fang
- The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Department of Toxicology, School of Public Health, Guizhou Medical University, Guiyang, 550025 Guizhou People’s Republic of China
| | - Baofei Sun
- The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Department of Toxicology, School of Public Health, Guizhou Medical University, Guiyang, 550025 Guizhou People’s Republic of China
| | - Kai Zhu
- The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Department of Toxicology, School of Public Health, Guizhou Medical University, Guiyang, 550025 Guizhou People’s Republic of China
| | - Maolin Yao
- The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Department of Toxicology, School of Public Health, Guizhou Medical University, Guiyang, 550025 Guizhou People’s Republic of China
| | - Shaofeng Wei
- The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Department of Toxicology, School of Public Health, Guizhou Medical University, Guiyang, 550025 Guizhou People’s Republic of China
| | - Aihua Zhang
- The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Department of Toxicology, School of Public Health, Guizhou Medical University, Guiyang, 550025 Guizhou People’s Republic of China
| |
Collapse
|
18
|
Xuekai L, Yan S, Jian C, Yifei S, Xinyue W, Wenyuan Z, Shuwen H, Xi Y. Advances in reprogramming of energy metabolism in tumor T cells. Front Immunol 2024; 15:1347181. [PMID: 38415258 PMCID: PMC10897011 DOI: 10.3389/fimmu.2024.1347181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 01/29/2024] [Indexed: 02/29/2024] Open
Abstract
Cancer is a leading cause of human death worldwide, and the modulation of the metabolic properties of T cells employed in cancer immunotherapy holds great promise for combating cancer. As a crucial factor, energy metabolism influences the activation, proliferation, and function of T cells, and thus metabolic reprogramming of T cells is a unique research perspective in cancer immunology. Special conditions within the tumor microenvironment and high-energy demands lead to alterations in the energy metabolism of T cells. In-depth research on the reprogramming of energy metabolism in T cells can reveal the mechanisms underlying tumor immune tolerance and provide important clues for the development of new tumor immunotherapy strategies as well. Therefore, the study of T cell energy metabolism has important clinical significance and potential applications. In the study, the current achievements in the reprogramming of T cell energy metabolism were reviewed. Then, the influencing factors associated with T cell energy metabolism were introduced. In addition, T cell energy metabolism in cancer immunotherapy was summarized, which highlighted its potential significance in enhancing T cell function and therapeutic outcomes. In summary, energy exhaustion of T cells leads to functional exhaustion, thus resulting in immune evasion by cancer cells. A better understanding of reprogramming of T cell energy metabolism may enable immunotherapy to combat cancer and holds promise for optimizing and enhancing existing therapeutic approaches.
Collapse
Affiliation(s)
- Liu Xuekai
- Department of Clinical Laboratory, Aerospace Center Hospital, Beijing, China
| | - Song Yan
- Department of Clinical Laboratory, Aerospace Center Hospital, Beijing, China
| | - Chu Jian
- Department of Medical Oncology, Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, Huzhou, China
- Department of Gastroenterology, Fifth School of Clinical Medicine of Zhejiang Chinese Medical University (Huzhou Central Hospital), Huzhou, China
- Department of Key Laboratory of Multiomics Research and Clinical Transformation of Digestive Cancer, Huzhou, China
| | - Song Yifei
- Department of Medical Oncology, Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, Huzhou, China
- Department of Gastroenterology, Fifth School of Clinical Medicine of Zhejiang Chinese Medical University (Huzhou Central Hospital), Huzhou, China
- Department of Key Laboratory of Multiomics Research and Clinical Transformation of Digestive Cancer, Huzhou, China
| | - Wu Xinyue
- Department of Medical Oncology, Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, Huzhou, China
- Department of Gastroenterology, Fifth School of Clinical Medicine of Zhejiang Chinese Medical University (Huzhou Central Hospital), Huzhou, China
- Department of Key Laboratory of Multiomics Research and Clinical Transformation of Digestive Cancer, Huzhou, China
| | - Zhang Wenyuan
- Department of Gynecology, Heyuan Hospital of Traditional Chinese Medicine, Heyuan, China
| | - Han Shuwen
- Department of Medical Oncology, Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, Huzhou, China
- Department of Gastroenterology, Fifth School of Clinical Medicine of Zhejiang Chinese Medical University (Huzhou Central Hospital), Huzhou, China
- Department of Key Laboratory of Multiomics Research and Clinical Transformation of Digestive Cancer, Huzhou, China
| | - Yang Xi
- Department of Medical Oncology, Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, Huzhou, China
- Department of Gastroenterology, Fifth School of Clinical Medicine of Zhejiang Chinese Medical University (Huzhou Central Hospital), Huzhou, China
- Department of Key Laboratory of Multiomics Research and Clinical Transformation of Digestive Cancer, Huzhou, China
| |
Collapse
|
19
|
Ding YJ, Li XN, Xiao Z, Li CY, Jia LH. Low vitamin D during pregnancy is associated with infantile eczema by up-regulation of PI3K/AKT/mTOR signaling pathway and affecting FOXP3 expression: A bidirectional cohort study. J Nutr Biochem 2024; 124:109516. [PMID: 37925089 DOI: 10.1016/j.jnutbio.2023.109516] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 10/26/2023] [Accepted: 10/31/2023] [Indexed: 11/06/2023]
Abstract
Vitamin D has received increasing attention because of its association with atopic disease development. Limited studies that have been done on the impact of maternal vitamin D levels during pregnancy on infantile eczema are still debatable. We wanted to discover the effect of maternal vitamin D on infantile eczema and explore whether regulatory T cells (Treg) play a role in this process. 219 pairs of mothers and children were enrolled. Maternal fasting venous blood was collected in pregnancy's second and third trimesters to determine vitamin D levels. Cord blood and placenta samples were collected during childbirth for detecting levels of genes, proteins and cytokines. Pediatricians followed up the prevalence of eczema in infants within 1 year. The reported rate of vitamin D deficiency and insufficiency was 35.6% and 28.3%. Lower maternal 25(OH)D3 levels were related to a higher risk of infantile eczema. Foxp3 gene expression is lower in cord blood of infants with eczema compared to infants without eczema. There was a positive correlation between maternal 25(OH)D3 levels and the expression of FOXP3 gene in cord blood. Compared to vitamin D sufficiency women, vitamin D deficiency women's placental FOXP3 protein expression was decreased and PI3K/AKT/mTOR protein was up-regulated. Our study demonstrates that low prenatal maternal vitamin D levels increased the risk of infantile eczema aged 0-1 year, which might be related to the downregulating of the FOXP3 gene expression in cord blood and decreased placental FOXP3 protein expression. Low placental FOXP3 protein was related with activating PI3K/AKT/mTOR signaling pathway.
Collapse
Affiliation(s)
- Ya-Jie Ding
- Huizhou Institute for Occupational Health, Huizhou, China; Department of Child and Adolescent Health, School of Public Health, China Medical University, Shenyang, China; Liaoning Key Laboratory of Obesity and Glucose/Lipid Associated Metabolic Diseases, Shenyang, China
| | - Xue-Ning Li
- Department of Pediatrics, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Zhe Xiao
- Department of Child and Adolescent Health, School of Public Health, China Medical University, Shenyang, China; Liaoning Key Laboratory of Obesity and Glucose/Lipid Associated Metabolic Diseases, Shenyang, China
| | - Chen-Yang Li
- Department of Child and Adolescent Health, School of Public Health, China Medical University, Shenyang, China; Liaoning Key Laboratory of Obesity and Glucose/Lipid Associated Metabolic Diseases, Shenyang, China
| | - Li-Hong Jia
- Department of Child and Adolescent Health, School of Public Health, China Medical University, Shenyang, China; Liaoning Key Laboratory of Obesity and Glucose/Lipid Associated Metabolic Diseases, Shenyang, China.
| |
Collapse
|
20
|
Limone F, Couto A, Wang JY, Zhang Y, McCourt B, Huang C, Minkin A, Jani M, McNeer S, Keaney J, Gillet G, Gonzalez RL, Goodman WA, Kadiu I, Eggan K, Burberry A. Myeloid and lymphoid expression of C9orf72 regulates IL-17A signaling in mice. Sci Transl Med 2024; 16:eadg7895. [PMID: 38295187 PMCID: PMC11247723 DOI: 10.1126/scitranslmed.adg7895] [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: 01/26/2023] [Accepted: 01/10/2024] [Indexed: 02/02/2024]
Abstract
A mutation in C9ORF72 is the most common cause of amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). Patients with ALS or FTD often develop autoimmunity and inflammation that precedes or coincides with the onset of neurological symptoms, but the underlying mechanisms are poorly understood. Here, we knocked out murine C9orf72 in seven hematopoietic progenitor compartments by conditional mutagenesis and found that myeloid lineage C9orf72 prevents splenomegaly, loss of tolerance, and premature mortality. Furthermore, we demonstrated that C9orf72 plays a role in lymphoid cells to prevent interleukin-17A (IL-17A) production and neutrophilia. Mass cytometry identified early and sustained elevation of the costimulatory molecule CD80 expressed on C9orf72-deficient mouse macrophages, monocytes, and microglia. Enrichment of CD80 was similarly observed in human spinal cord microglia from patients with C9ORF72-mediated ALS compared with non-ALS controls. Single-cell RNA sequencing of murine spinal cord, brain cortex, and spleen demonstrated coordinated induction of gene modules related to antigen processing and presentation and antiviral immunity in C9orf72-deficient endothelial cells, microglia, and macrophages. Mechanistically, C9ORF72 repressed the trafficking of CD80 to the cell surface in response to Toll-like receptor agonists, interferon-γ, and IL-17A. Deletion of Il17a in C9orf72-deficient mice prevented CD80 enrichment in the spinal cord, reduced neutrophilia, and reduced gut T helper type 17 cells. Last, systemic delivery of an IL-17A neutralizing antibody augmented motor performance and suppressed neuroinflammation in C9orf72-deficient mice. Altogether, we show that C9orf72 orchestrates myeloid costimulatory potency and provide support for IL-17A as a therapeutic target for neuroinflammation associated with ALS or FTD.
Collapse
Affiliation(s)
- Francesco Limone
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, 02138, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA, 02138, USA
- Leiden University Medical Center, LUMC, 2333 ZA Leiden, The Netherlands
| | - Alexander Couto
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, 02138, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA, 02138, USA
| | - Jin-Yuan Wang
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, 02138, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA, 02138, USA
| | - Yingying Zhang
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, 02138, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA, 02138, USA
| | - Blake McCourt
- Department of Pathology, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Cerianne Huang
- Department of Pathology, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Adina Minkin
- Department of Pathology, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Marghi Jani
- Department of Pathology, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Sarah McNeer
- Department of Pathology, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - James Keaney
- Neuroinflammation Focus Area, UCB Biopharma SRL, Braine-l’Alleud, 1420, Belgium
| | - Gaëlle Gillet
- Neuroinflammation Focus Area, UCB Biopharma SRL, Braine-l’Alleud, 1420, Belgium
| | - Rodrigo Lopez Gonzalez
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44196, USA
| | - Wendy A. Goodman
- Department of Pathology, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Irena Kadiu
- Neuroinflammation Focus Area, UCB Biopharma SRL, Braine-l’Alleud, 1420, Belgium
| | - Kevin Eggan
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, 02138, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA, 02138, USA
| | - Aaron Burberry
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, 02138, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA, 02138, USA
- Department of Pathology, Case Western Reserve University, Cleveland, OH, 44106, USA
| |
Collapse
|
21
|
Rodolfi S, Davidson C, Vecellio M. Regulatory T cells in spondyloarthropathies: genetic evidence, functional role, and therapeutic possibilities. Front Immunol 2024; 14:1303640. [PMID: 38288110 PMCID: PMC10822883 DOI: 10.3389/fimmu.2023.1303640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Accepted: 12/28/2023] [Indexed: 01/31/2024] Open
Abstract
Regulatory T cells (Tregs) are a very specialized subset of T lymphocytes: their main function is controlling immune responses during inflammation. T-regs involvement in autoimmune and immune-mediated rheumatic diseases is well-described. Here, we critically review the up-to-date literature findings on the role of Tregs in spondyloarthropathies, particularly in ankylosing spondylitis (AS), a polygenic inflammatory rheumatic disease that preferentially affects the spine and the sacroiliac joints. Genetics discoveries helped in elucidating pathogenic T-regs gene modules and functional involvement. We highlight T-regs tissue specificity as crucial point, as T-regs might have a distinct epigenomic and molecular profiling depending on the different site of tissue inflammation. Furthermore, we speculate about possible therapeutic interventions targeting, or enhancing, Treg cells in spondyloarthropathies.
Collapse
Affiliation(s)
- Stefano Rodolfi
- Department of Rheumatology and Clinical Immunology, IRCCS Humanitas Research Hospital, Milan, Italy
- Department of Biomedical Sciences, Humanitas University, Milan, Italy
| | - Connor Davidson
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Matteo Vecellio
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
- Centro Ricerche Fondazione Italiana Ricerca Sull'Artrite (FIRA), Fondazione Pisana per la Scienza ONLUS, San Giuliano Terme, Italy
| |
Collapse
|
22
|
Abdullah NA, Ali NH, Aljaleel NA. Gene expression profile of immune-check point in response to Trastuzumab therapy in patients with HER-2 positive breast cancer. WIADOMOSCI LEKARSKIE (WARSAW, POLAND : 1960) 2024; 77:1056-1062. [PMID: 39008597 DOI: 10.36740/wlek202405127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/17/2024]
Abstract
OBJECTIVE Aim: To clarify the association between response to Trastuzumab and molecular expression of TIM-3 and FOXP-3 immune checkpoints. PATIENTS AND METHODS Materials and Methods: FOXP-3 and TIM-3 expression in peripheral blood was analyzed using qPCR, and the serum level of Trastuzumab was estimated using an immune sorbent enzyme assay. RESULTS Results: During treatment with Trastuzumab, the FOXP-3 gene expression showed a significant decline throughout one year of treatment, going from 0.85 at cycle 9 to 0.75 at cycle 17. While the TIM-3 gene expression showed a significant up regulation at cycle 9 to 2.8 fold, followed by a reduction in the fold change from 2.8 to 1.7 in the font of reference gene expression. CONCLUSION Conclusions:FOXP-3 and TIM-3 have the potential to be suggestive markers that can anticipate the response to Trastuzumab, but they are not capable of predicting the likelihood of recurrence.
Collapse
|
23
|
Christofi P, Pantazi C, Psatha N, Sakellari I, Yannaki E, Papadopoulou A. Promises and Pitfalls of Next-Generation Treg Adoptive Immunotherapy. Cancers (Basel) 2023; 15:5877. [PMID: 38136421 PMCID: PMC10742252 DOI: 10.3390/cancers15245877] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Revised: 12/13/2023] [Accepted: 12/14/2023] [Indexed: 12/24/2023] Open
Abstract
Regulatory T cells (Tregs) are fundamental to maintaining immune homeostasis by inhibiting immune responses to self-antigens and preventing the excessive activation of the immune system. Their functions extend beyond immune surveillance and subpopulations of tissue-resident Treg cells can also facilitate tissue repair and homeostasis. The unique ability to regulate aberrant immune responses has generated the concept of harnessing Tregs as a new cellular immunotherapy approach for reshaping undesired immune reactions in autoimmune diseases and allo-responses in transplantation to ultimately re-establish tolerance. However, a number of issues limit the broad clinical applicability of Treg adoptive immunotherapy, including the lack of antigen specificity, heterogeneity within the Treg population, poor persistence, functional Treg impairment in disease states, and in vivo plasticity that results in the loss of suppressive function. Although the early-phase clinical trials of Treg cell therapy have shown the feasibility and tolerability of the approach in several conditions, its efficacy has remained questionable. Leveraging the smart tools and platforms that have been successfully developed for primary T cell engineering in cancer, the field has now shifted towards "next-generation" adoptive Treg immunotherapy, where genetically modified Treg products with improved characteristics are being generated, as regards antigen specificity, function, persistence, and immunogenicity. Here, we review the state of the art on Treg adoptive immunotherapy and progress beyond it, while critically evaluating the hurdles and opportunities towards the materialization of Tregs as a living drug therapy for various inflammation states and the broad clinical translation of Treg therapeutics.
Collapse
Affiliation(s)
- Panayiota Christofi
- Gene and Cell Therapy Center, Hematopoietic Cell Transplantation Unit, Hematology Department, George Papanikolaou Hospital, 57010 Thessaloniki, Greece; (P.C.); (C.P.); (I.S.); (E.Y.)
- University General Hospital of Patras, 26504 Rio, Greece
| | - Chrysoula Pantazi
- Gene and Cell Therapy Center, Hematopoietic Cell Transplantation Unit, Hematology Department, George Papanikolaou Hospital, 57010 Thessaloniki, Greece; (P.C.); (C.P.); (I.S.); (E.Y.)
- Department of Genetics, Development and Molecular Biology, School of Biology, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece;
- Institute of Applied Biosciences (INAB), Centre for Research and Technology Hellas (CERTH), 57001 Thessaloniki, Greece
| | - Nikoleta Psatha
- Department of Genetics, Development and Molecular Biology, School of Biology, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece;
| | - Ioanna Sakellari
- Gene and Cell Therapy Center, Hematopoietic Cell Transplantation Unit, Hematology Department, George Papanikolaou Hospital, 57010 Thessaloniki, Greece; (P.C.); (C.P.); (I.S.); (E.Y.)
| | - Evangelia Yannaki
- Gene and Cell Therapy Center, Hematopoietic Cell Transplantation Unit, Hematology Department, George Papanikolaou Hospital, 57010 Thessaloniki, Greece; (P.C.); (C.P.); (I.S.); (E.Y.)
- Department of Medicine, University of Washington, Seattle, WA 98195-7710, USA
| | - Anastasia Papadopoulou
- Gene and Cell Therapy Center, Hematopoietic Cell Transplantation Unit, Hematology Department, George Papanikolaou Hospital, 57010 Thessaloniki, Greece; (P.C.); (C.P.); (I.S.); (E.Y.)
| |
Collapse
|
24
|
Yu S, Gu J, Wang R, Lee S, Shan Y, Wang J, Sun Y, Ma X. TIGIT reverses IFN-α-promoted Th1-like Tregs via in-sequence effects dependent on STAT4. Arthritis Res Ther 2023; 25:221. [PMID: 37978415 PMCID: PMC10655484 DOI: 10.1186/s13075-023-03202-8] [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: 09/25/2023] [Accepted: 10/30/2023] [Indexed: 11/19/2023] Open
Abstract
OBJECTIVES The induction direction of interferon (IFN)-α in T-cell phenotype and function varies depending on the activation state of the cell and the time of stimulation. To assess the effects of elevated IFN-α on regulatory T cells (Tregs) in systemic lupus erythematosus (SLE) patients, we investigated the differentiation of Th1-like Tregs under in-sequence and out-of-sequence conditions and the reversal effect of activating TIGIT on immune suppression. METHODS Phenotypes and activation levels of Tregs from SLE patients and healthy controls were analyzed using flow cytometry. In vitro culture conditions based on the sequence of TCR activation and IFN-α stimulation simulated in-sequence or out-of-sequence effects. CD4+T cells and Tregs were cultured under the above conditions with or without TIGIT agonist. Expression of related characteristic markers and phosphorylation levels of AKT, mTOR, and STATs were detected using flow cytometry and ELISA. RESULTS The frequency of Th1-like Tregs and activation levels of Tregs increased, but TIGIT expression in Tregs decreased in SLE patients. IFN-α promoted the conversation of Tregs to Th1-like Tregs while reducing immunosuppressive function under in-sequence conditions. The STAT4 pathway, but not the STAT1 pathway, was crucial for the IFN-α-mediated in-sequence effects. Reactivation of TIGIT reversed Th1 polarization of Tregs by suppressing AKT/mTOR and STAT4 signaling. CONCLUSIONS Our findings suggest that IFN-α mediated in-sequence effects on Tregs may be responsible for the expansion of Th1-like Tregs in SLE. TIGIT can restore immune suppression damage in Tregs and represents a potential therapeutic target for SLE.
Collapse
Affiliation(s)
- Shihan Yu
- Department of Pediatrics, The First Hospital of China Medical University, No.155, Nanjingbei Street, Heping District, Shenyang, 110001, China
- Shanghai Children's Medical Center Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jia Gu
- Department of Pediatrics, The First Hospital of China Medical University, No.155, Nanjingbei Street, Heping District, Shenyang, 110001, China
- Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Rui Wang
- Department of Pediatrics, The First Hospital of China Medical University, No.155, Nanjingbei Street, Heping District, Shenyang, 110001, China
| | - Seunghyun Lee
- Department of Preventive Medicine, College of Medicine, Chung-Ang University, Seoul, Republic of Korea
| | - Yu Shan
- Department of Pediatrics, Shenyang Women's and Children's Hospital, Shenyang, China
- First Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health, Japan, Kitakyushu, Japan
| | - Jiakai Wang
- Department of Rheumatology and Immunology, The First Hospital of China Medical University, Shenyang, China
| | - Yini Sun
- Department of Critical Care Medicine, The First Hospital of China Medical University, Shenyang, China
| | - Xiaoxue Ma
- Department of Pediatrics, The First Hospital of China Medical University, No.155, Nanjingbei Street, Heping District, Shenyang, 110001, China.
| |
Collapse
|
25
|
Fan HM, Qiao YL, Liu Y, Xu S, Ni HF, Jiao WE, Tao ZZ, Chen SM. Long-term consequences of regulatory T-cell-specific knockout of Notch2 in immune homeostasis. Int Immunopharmacol 2023; 124:111069. [PMID: 37852117 DOI: 10.1016/j.intimp.2023.111069] [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: 08/06/2023] [Revised: 10/01/2023] [Accepted: 10/10/2023] [Indexed: 10/20/2023]
Abstract
AIMS To investigate the long-term alterations in immune function and spontaneous inflammation in mice following specific knockout of Notch2 (Notch2KO) in Treg cells. MAIN METHODS A Treg cell-specific Notch2 knockout mouse model was constructed, and the mice were named Notch2KO mice. The pathological changes and inflammatory cell infiltration in the lungs, skin, and liver of the mice at 2, 6, 9, and 12 months of age were evaluated by HE staining. The expression of Th1/Th2/Th17/Treg transcription factors was detected by Western blotting. The proportion of CD4 + T-cell subsets was determined by flow cytometry. The levels of Th1/Th2/Th17/Treg cytokines were measured by enzyme-linked immunosorbent assays (ELISAs). KEY FINDINGS The expression level of Notch2 in Treg cells from the Notch2KO mice was significantly decreased compared with that in Treg cells from the control mice (P < 0.05). HE staining showed that compared with the control mice, the Notch2KO mice displayed spontaneous inflammation and had a large amount of inflammatory cell infiltration in the lungs and skin (P < 0.05). The number of Treg cells, the expression level of Foxp3, and the level of IL-10 were reduced in the Notch2KO mice compared with the control mice (P < 0.05), and these metrics further decreased with increasing age (P < 0.05). In contrast, the number of Th1/Th2 cells, the expression level of T-bet/GATA3, and the levels of Th1 cytokines (IFN-γ)/Th2 cytokines (IL-4, IL-5, and IL-13) were significantly increased in the Notch2KO mice (P < 0.05), and these metrics further increased with increasing age (P < 0.05). There was no significant change in the number of Th17 cells, the expression of RORγt, or the level of IL-17. Further analysis showed that the balance of Th1/Th2 and Treg/Th17 cells in the Notch2KO mice was shifted, and the ratio showed a downward trend over time (P < 0.05). SIGNIFICANCE The number and function of Treg cells can be severely inhibited by a specific knockout of Notch2 in Treg cells, leading to immune disorders that gradually worsen over time.
Collapse
Affiliation(s)
- Hui-Ming Fan
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, 238 Jie-Fang Road, Wuhan, Hubei 430060, PR China
| | - Yue-Long Qiao
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, 238 Jie-Fang Road, Wuhan, Hubei 430060, PR China
| | - Yan Liu
- Department of Nursing, Renmin Hospital of Wuhan University, 238 Jie-Fang Road, Wuhan, Hubei 430060, PR China
| | - Shan Xu
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, 238 Jie-Fang Road, Wuhan, Hubei 430060, PR China
| | - Hai-Feng Ni
- Department of Otolaryngology Head and Neck Surgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310006, PR China
| | - Wo-Er Jiao
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, 238 Jie-Fang Road, Wuhan, Hubei 430060, PR China
| | - Ze-Zhang Tao
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, 238 Jie-Fang Road, Wuhan, Hubei 430060, PR China; Institute of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, 238 Jie-Fang Road, Wuhan, Hubei 430060, PR China
| | - Shi-Ming Chen
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, 238 Jie-Fang Road, Wuhan, Hubei 430060, PR China; Institute of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, 238 Jie-Fang Road, Wuhan, Hubei 430060, PR China.
| |
Collapse
|
26
|
Freuchet A, Roy P, Armstrong SS, Oliaeimotlagh M, Kumar S, Orecchioni M, Ali AJ, Khan A, Makings J, Lyu Q, Winkels H, Wang E, Durant C, Ghosheh Y, Gulati R, Nettersheim F, Ley K. Identification of human exT reg cells as CD16 +CD56 + cytotoxic CD4 + T cells. Nat Immunol 2023; 24:1748-1761. [PMID: 37563308 PMCID: PMC11022744 DOI: 10.1038/s41590-023-01589-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Accepted: 07/13/2023] [Indexed: 08/12/2023]
Abstract
In atherosclerosis, some regulatory T (Treg) cells become exTreg cells. We crossed inducible Treg and exTreg cell lineage-tracker mice (FoxP3eGFP-Cre-ERT2ROSA26CAG-fl-stop-fl-tdTomato) to atherosclerosis-prone Apoe-/- mice, sorted Treg cells and exTreg cells and determined their transcriptomes by bulk RNA sequencing (RNA-seq). Genes that were differentially expressed between mouse Treg cells and exTreg cells and filtered for their presence in a human single-cell RNA-sequencing (scRNA-seq) panel identified exTreg cell signature genes as CST7, NKG7, GZMA, PRF1, TBX21 and CCL4. Projecting these genes onto the human scRNA-seq with CITE-seq data identified human exTreg cells as CD3+CD4+CD16+CD56+, which was validated by flow cytometry. Bulk RNA-seq of sorted human exTreg cells identified them as inflammatory and cytotoxic CD4+T cells that were significantly distinct from both natural killer and Treg cells. DNA sequencing for T cell receptor-β showed clonal expansion of Treg cell CDR3 sequences in exTreg cells. Cytotoxicity was functionally demonstrated in cell killing and CD107a degranulation assays, which identifies human exTreg cells as cytotoxic CD4+T cells.
Collapse
Affiliation(s)
| | - Payel Roy
- La Jolla Institute for Immunology, La Jolla, CA, USA
| | | | | | - Sunil Kumar
- Immunology Center of Georgia, Augusta University, Augusta, GA, USA
| | - Marco Orecchioni
- La Jolla Institute for Immunology, La Jolla, CA, USA
- Immunology Center of Georgia, Augusta University, Augusta, GA, USA
| | - Amal J Ali
- La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Amir Khan
- Immunology Center of Georgia, Augusta University, Augusta, GA, USA
| | | | - Qingkang Lyu
- Immunology Center of Georgia, Augusta University, Augusta, GA, USA
| | - Holger Winkels
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Clinic III for Internal Medicine, Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Erpei Wang
- La Jolla Institute for Immunology, La Jolla, CA, USA
| | | | - Yanal Ghosheh
- La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Rishab Gulati
- La Jolla Institute for Immunology, La Jolla, CA, USA
| | | | - Klaus Ley
- La Jolla Institute for Immunology, La Jolla, CA, USA.
- Immunology Center of Georgia, Augusta University, Augusta, GA, USA.
| |
Collapse
|
27
|
Zhao M, Chu J, Feng S, Guo C, Xue B, He K, Li L. Immunological mechanisms of inflammatory diseases caused by gut microbiota dysbiosis: A review. Biomed Pharmacother 2023; 164:114985. [PMID: 37311282 DOI: 10.1016/j.biopha.2023.114985] [Citation(s) in RCA: 54] [Impact Index Per Article: 54.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 05/31/2023] [Accepted: 06/01/2023] [Indexed: 06/15/2023] Open
Abstract
The gut microbiota is indispensable for maintaining host health by enhancing the host's digestive capacity, safeguarding the intestinal epithelial barrier, and preventing pathogen invasion. Additionally, the gut microbiota exhibits a bidirectional interaction with the host immune system and promotes the immune system of the host to mature. Dysbiosis of the gut microbiota, primarily caused by factors such as host genetic susceptibility, age, BMI, diet, and drug abuse, is a significant contributor to inflammatory diseases. However, the mechanisms underlying inflammatory diseases resulting from gut microbiota dysbiosis lack systematic categorization. In this study, we summarize the normal physiological functions of symbiotic microbiota in a healthy state and demonstrate that when dysbiosis occurs due to various external factors, the normal physiological functions of the gut microbiota are lost, leading to pathological damage to the intestinal lining, metabolic disorders, and intestinal barrier damage. This, in turn, triggers immune system disorders and eventually causes inflammatory diseases in various systems. These discoveries provide fresh perspectives on how to diagnose and treat inflammatory diseases. However, the unrecognized variables that might affect the link between inflammatory illnesses and gut microbiota, need further studies and extensive basic and clinical research will still be required to investigate this relationship in the future.
Collapse
Affiliation(s)
- Min'an Zhao
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun, Jilin 130021, China; School of Public Health, Jilin University, Changchun, Jilin 130021, China
| | - Jiayi Chu
- School of Public Health, Jilin University, Changchun, Jilin 130021, China
| | - Shiyao Feng
- School of Public Health, Jilin University, Changchun, Jilin 130021, China
| | - Chuanhao Guo
- The Second School of Clinical Medicine of Jilin University, Changchun, Jilin 130041, China
| | - Baigong Xue
- College of Basic Medical Sciences, Jilin University, Changchun, Jilin 130021, China.
| | - Kan He
- Department of Pharmacology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin 130021, China.
| | - Lisha Li
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun, Jilin 130021, China.
| |
Collapse
|
28
|
Sun L, Su Y, Jiao A, Wang X, Zhang B. T cells in health and disease. Signal Transduct Target Ther 2023; 8:235. [PMID: 37332039 PMCID: PMC10277291 DOI: 10.1038/s41392-023-01471-y] [Citation(s) in RCA: 188] [Impact Index Per Article: 188.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 04/21/2023] [Accepted: 04/24/2023] [Indexed: 06/20/2023] Open
Abstract
T cells are crucial for immune functions to maintain health and prevent disease. T cell development occurs in a stepwise process in the thymus and mainly generates CD4+ and CD8+ T cell subsets. Upon antigen stimulation, naïve T cells differentiate into CD4+ helper and CD8+ cytotoxic effector and memory cells, mediating direct killing, diverse immune regulatory function, and long-term protection. In response to acute and chronic infections and tumors, T cells adopt distinct differentiation trajectories and develop into a range of heterogeneous populations with various phenotype, differentiation potential, and functionality under precise and elaborate regulations of transcriptional and epigenetic programs. Abnormal T-cell immunity can initiate and promote the pathogenesis of autoimmune diseases. In this review, we summarize the current understanding of T cell development, CD4+ and CD8+ T cell classification, and differentiation in physiological settings. We further elaborate the heterogeneity, differentiation, functionality, and regulation network of CD4+ and CD8+ T cells in infectious disease, chronic infection and tumor, and autoimmune disease, highlighting the exhausted CD8+ T cell differentiation trajectory, CD4+ T cell helper function, T cell contributions to immunotherapy and autoimmune pathogenesis. We also discuss the development and function of γδ T cells in tissue surveillance, infection, and tumor immunity. Finally, we summarized current T-cell-based immunotherapies in both cancer and autoimmune diseases, with an emphasis on their clinical applications. A better understanding of T cell immunity provides insight into developing novel prophylactic and therapeutic strategies in human diseases.
Collapse
Affiliation(s)
- Lina Sun
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
- Institute of Infection and Immunity, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, China
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi, 710061, China
- Xi'an Key Laboratory of Immune Related Diseases, Xi'an, Shannxi, 710061, China
| | - Yanhong Su
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
- Institute of Infection and Immunity, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, China
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi, 710061, China
- Xi'an Key Laboratory of Immune Related Diseases, Xi'an, Shannxi, 710061, China
| | - Anjun Jiao
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
- Institute of Infection and Immunity, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, China
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi, 710061, China
- Xi'an Key Laboratory of Immune Related Diseases, Xi'an, Shannxi, 710061, China
| | - Xin Wang
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
- Institute of Infection and Immunity, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, China
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi, 710061, China
- Xi'an Key Laboratory of Immune Related Diseases, Xi'an, Shannxi, 710061, China
| | - Baojun Zhang
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China.
- Institute of Infection and Immunity, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, China.
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi, 710061, China.
- Xi'an Key Laboratory of Immune Related Diseases, Xi'an, Shannxi, 710061, China.
| |
Collapse
|
29
|
Chen Z, Huang Y, Wang B, Peng H, Wang X, Wu H, Chen W, Wang M. T cells: an emerging cast of roles in bipolar disorder. Transl Psychiatry 2023; 13:153. [PMID: 37156764 PMCID: PMC10167236 DOI: 10.1038/s41398-023-02445-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 04/19/2023] [Accepted: 04/21/2023] [Indexed: 05/10/2023] Open
Abstract
Bipolar disorder (BD) is a distinctly heterogeneous and multifactorial disorder with a high individual and social burden. Immune pathway dysregulation is an important pathophysiological feature of BD. Recent studies have suggested a potential role for T lymphocytes in the pathogenesis of BD. Therefore, greater insight into T lymphocytes' functioning in patients with BD is essential. In this narrative review, we describe the presence of an imbalance in the ratio and altered function of T lymphocyte subsets in BD patients, mainly in T helper (Th) 1, Th2, Th17 cells and regulatory T cells, and alterations in hormones, intracellular signaling, and microbiomes may be potential causes. Abnormal T cell presence explains the elevated rates of comorbid inflammatory illnesses in the BD population. We also update the findings on T cell-targeting drugs as potentially immunomodulatory therapeutic agents for BD disease in addition to classical mood stabilizers (lithium, valproic acid). In conclusion, an imbalance in T lymphocyte subpopulation ratios and altered function may be involved in the development of BD, and maintaining T cell immune homeostasis may provide an overall therapeutic benefit.
Collapse
Affiliation(s)
- Zhenni Chen
- Department of Laboratory Medicine, the Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
| | - Yiran Huang
- School of Clinical Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, China
| | - Bingqi Wang
- Department of Laboratory Medicine, the Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
| | - Huanqie Peng
- Department of Laboratory Medicine, the Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
| | - Xiaofan Wang
- Department of Laboratory Medicine, the Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
| | - Hongzheng Wu
- Department of Laboratory Medicine, the Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
| | - Wanxin Chen
- Department of Laboratory Medicine, the Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
| | - Min Wang
- Department of Laboratory Medicine, the Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China.
| |
Collapse
|
30
|
Keller LE, Tait Wojno ED, Begum L, Fortier LA. Interleukin-6 neutralization and regulatory T cells are additive in chondroprotection from IL-1β-induced inflammation. J Orthop Res 2023; 41:942-950. [PMID: 36205183 PMCID: PMC10079781 DOI: 10.1002/jor.25453] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 08/25/2022] [Accepted: 10/01/2022] [Indexed: 02/04/2023]
Abstract
Anti-inflammatory Regulatory T cells (Tregs) are enriched in the joints of patients with osteoarthritis (OA) compared to healthy joints. Tregs maintain homeostasis through secretion of anti-inflammatory cytokines and cell-to-cell interactions including immune checkpoint signaling. Interleukin-6 (IL-6) is a pleiotropic cytokine secreted by inflamed synoviocytes and chondrocytes that can inhibit or alter Treg function. This study tested the hypothesis that neutralization of IL-6 would enable Treg anti-inflammatory function to resolve inflammation and catabolism elicited by IL-1β in an equine chondrocyte/synoviocyte/Treg tri-culture OA model. Synoviocyte/chondrocyte co-cultures were stimulated with IL-1β, and treated with αIL-6 neutralizing antibody. Activated Tregs secreting IL-10 were added in direct contact with synoviocytes to create a tri-culture. Neutralization of IL-6 partially restored Treg anti-inflammatory functions and, in combination, reduced IL-1β-stimulated synoviocyte MMP13 expression to control levels and restored Acan expression in chondrocytes. IL-6 neutralization alone decreased Il6 expression in chondrocytes and synoviocytes, mitigating IL-6 positive feedback loop. Although Tregs were the primary producers of anti-inflammatory IL-10 and IL-4, they also produced pro-inflammatory IL-17A, as detected by ELIA, which may have been responsible for incomplete rescue of synoviocyte/chondrocyte homeostasis following IL-1β stimulation. Treg secretion of IL-10, IL-4, and IL-17A was not altered by tri-culture conditions or presence of αIL-6, therefore, it was unlikely that Treg phenotype instability occurred. The significant effect of chondrocyte/synoviocyte donor, but not Treg donor, on gene expression and IL-6 concentration in conditioned media, indicated that personalized therapy considering the patient's OA status might be needed for successful implementation of immunotherapy in the context of OA.
Collapse
Affiliation(s)
- Laura E. Keller
- Cornell University, College of Veterinary Medicine, Department of Clinical Sciences
| | | | - Laila Begum
- Cornell University, College of Veterinary Medicine, Department of Clinical Sciences
| | - Lisa A. Fortier
- Cornell University, College of Veterinary Medicine, Department of Clinical Sciences
| |
Collapse
|
31
|
Jian L, Hu Y, Gao M, Shu L. Promoted Generation of T Helper 1-Like Regulatory T Cells After Transient Middle Cerebral Artery Occlusion in Type-2 Diabetic Mice. Immunol Invest 2023:1-17. [PMID: 37076315 DOI: 10.1080/08820139.2023.2197009] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/21/2023]
Abstract
BACKGROUND Regulatory T cells (Tregs) play a remarkable role in modulating post-ischemic neuroinflammation. However, the characteristics of Tregs in diabetic ischemic stroke remain unknown. METHODS Transient middle cerebral artery occlusion (MCAO) was conducted on leptin receptor-mutated db/db mice and db/+ mice. The number, cytokine production, and signaling features of Tregs in peripheral blood and ipsilateral hemispheres were evaluated by flow cytometry. Treg plasticity was assessed by the adoptive transfer of splenic Tregs into mice. The effect of ipsilateral macrophages/microglia on Treg plasticity was determined by in vitro co-culture analysis. RESULTS db/db mice had more infiltrating Tregs in their ipsilateral hemispheres than db/+ mice. Infiltrating Tregs in db/db mice expressed higher transforming growth factor-β (TGF-β), interleukin-10 (IL-10), forkhead box P3 (Foxp3), interferon-γ (IFN-γ), tumor necrosis factor-α (TNF-α), and T-box expressed in T cells (T-bet) in comparison to infiltrating Tregs in db/+ mice, suggesting promoted generation of T helper 1 (Th1)-like Tregs in the brains of db/db mice after stroke. The post-ischemic brain microenvironment of db/db mice significantly up-regulated IFN-γ, TNF-α, T-bet, IL-10, and TGF-β in infiltrating Tregs. Moreover, ipsilateral macrophages/microglia remarkably enhanced the expression of IFN-γ, TNF-α, and T-bet but not IL-10 and TGF-β in Tregs. db/db macrophages/microglia were more potent in up-regulating IFN-γ, TNF-α, and T-bet than db/+ macrophages/microglia. Interleukin-12 (IL-12) blockage partially abolished the modulatory effect of macrophages/microglia on Tregs. CONCLUSION The generation of Th1-like Tregs was promoted in the brains of type 2 diabetic mice after stroke. Our study reveals significant Treg plasticity in diabetic stroke.Abbreviations: Foxp3: forkhead box P3; IFN-γ: interferon-γ; IL-10: interleukin-10; IL-12: interleukin-12; MCAO: middle cerebral artery occlusion; PBS: phosphate-buffered saline; STAT1: Signal transducer and activator of transcription 1; STAT5: Signal transducer and activator of transcription 1; T-bet: T-box expressed in T cells; TGF-β: transforming growth factor-β; Th1: T helper 1; TNF-α: tumor necrosis factor-α; Tregs: regulatory T cells. Foxp3: forkhead box P3; IFN-γ: interferon-γ; IL-10: interleukin-10; IL-12: interleukin-12; MCAO: middle cerebral artery occlusion; PBS: phosphate-buffered saline; STAT1: Signal transducer and activator of transcription 1; STAT5: Signal transducer and activator of transcription 1; T-bet: T-box expressed in T cells; TGF-β: transforming growth factor-β; Th1: T helper 1; TNF-α: tumor necrosis factor-α; Tregs: regulatory T cells.
Collapse
Affiliation(s)
- Lei Jian
- The department of Endocrinology, Renhe Hospital of China Three Gorges University, Yichang City, Hubei Province, China
| | - Yanqi Hu
- The department of Endocrinology, Renhe Hospital of China Three Gorges University, Yichang City, Hubei Province, China
| | - Mingjie Gao
- The Department of Neurology, Renhe Hospital of China Three Gorges University, Yichang City, Hubei Province, China
| | - Long Shu
- The Department of Neurology, Renhe Hospital of China Three Gorges University, Yichang City, Hubei Province, China
| |
Collapse
|
32
|
Műzes G, Sipos F. Autoimmunity and Carcinogenesis: Their Relationship under the Umbrella of Autophagy. Biomedicines 2023; 11:biomedicines11041130. [PMID: 37189748 DOI: 10.3390/biomedicines11041130] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Revised: 04/04/2023] [Accepted: 04/06/2023] [Indexed: 04/11/2023] Open
Abstract
The immune system and autophagy share a functional relationship. Both innate and adaptive immune responses involve autophagy and, depending on the disease’s origin and pathophysiology, it may have a detrimental or positive role on autoimmune disorders. As a “double-edged sword” in tumors, autophagy can either facilitate or impede tumor growth. The autophagy regulatory network that influences tumor progression and treatment resistance is dependent on cell and tissue types and tumor stages. The connection between autoimmunity and carcinogenesis has not been sufficiently explored in past studies. As a crucial mechanism between the two phenomena, autophagy may play a substantial role, though the specifics remain unclear. Several autophagy modifiers have demonstrated beneficial effects in models of autoimmune disease, emphasizing their therapeutic potential as treatments for autoimmune disorders. The function of autophagy in the tumor microenvironment and immune cells is the subject of intensive study. The objective of this review is to investigate the role of autophagy in the simultaneous genesis of autoimmunity and malignancy, shedding light on both sides of the issue. We believe our work will assist in the organization of current understanding in the field and promote additional research on this urgent and crucial topic.
Collapse
Affiliation(s)
- Györgyi Műzes
- Immunology Division, Department of Internal Medicine and Hematology, Semmelweis University, 1088 Budapest, Hungary
| | - Ferenc Sipos
- Immunology Division, Department of Internal Medicine and Hematology, Semmelweis University, 1088 Budapest, Hungary
| |
Collapse
|
33
|
Shen MY, Jiang BP, Zhang MF, Wang X, Zhu H, Gu ZN, Zhou XP, Lu Y, Zhou LL. MicroRNA-143-3p ameliorates collagen-induced arthritis by polarizing naive CD4 + T cells into Treg cells. J Clin Lab Anal 2023; 37:e24845. [PMID: 36861291 PMCID: PMC10098049 DOI: 10.1002/jcla.24845] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 01/04/2023] [Accepted: 01/26/2023] [Indexed: 03/03/2023] Open
Abstract
BACKGROUND Rheumatoid arthritis (RA) is a persistent and systemic autoimmunity disease. The abnormal differentiation of Treg cells is important in pathogenesis. Despite previous studies showed that microRNAs (miRNAs, miR) are pivotal modulators of Treg cells, the effect of miRNAs on Treg cell differentiation and function is not clear. Our study wants to reveal the relationship of miR-143-3p with the differentiative ability and biofunction of Treg cells during the development of RA. METHODS The Expressing level of miR-143-3p and cell factor generation in peripheral blood (PB) of RA sufferers were identified by ELISA or RT-qPCR. The roles of miR-143-3p in Treg cell differentiation were studied via ShRNA/lentivirus transfection. Male DBA/1 J mice were separated into control, model, control mimics, and miR-143-3p mimics groups to analyze the anti-arthritis efficacy, the differentiative ability of Treg cells, and the expression level of miR-143-3p. RESULTS Our team discovered that the Expressing level of miR-143-3p was related to RA disease activities in a negative manner, and remarkably related to antiinflammation cell factor IL-10. In vitro, the expression of miR-143-3p in the CD4+ T cells upregulated the percentage of CD4+ CD25+ Fxop3+ cells (Tregs) and forkhead box protein 3 (Foxp3) mRNA expression. Evidently, miR-143-3p mimic intervention considerably upregulated the content of Treg cells in vivo, validly avoided CIA progression, and remarkably suppressed the inflammatory events of joints in mice. CONCLUSION Our findings indicated that miR-143-3p could ameliorate CIA through polarizing naive CD4+ T cells into Treg cells, which may be a novel strategy to treat autoimmune diseases such as RA.
Collapse
Affiliation(s)
- Mei-Yu Shen
- School of Pharmacy, Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China
| | - Bao-Ping Jiang
- School of Pharmacy, Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China
| | - Ming-Fei Zhang
- School of Pharmacy, Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China
| | - Xiang Wang
- School of Pharmacy, Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China
| | - Hong Zhu
- Department of Rheumatology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China.,The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China
| | - Zhen-Ning Gu
- Department of Rheumatology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China.,The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China
| | - Xue-Ping Zhou
- The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yan Lu
- Department of Rheumatology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China.,The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China
| | - Ling-Ling Zhou
- School of Pharmacy, Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
34
|
Innate and adaptive immune abnormalities underlying autoimmune diseases: the genetic connections. SCIENCE CHINA. LIFE SCIENCES 2023:10.1007/s11427-021-2187-3. [PMID: 36738430 PMCID: PMC9898710 DOI: 10.1007/s11427-021-2187-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 10/10/2022] [Indexed: 02/05/2023]
Abstract
With the exception of an extremely small number of cases caused by single gene mutations, most autoimmune diseases result from the complex interplay between environmental and genetic factors. In a nutshell, etiology of the common autoimmune disorders is unknown in spite of progress elucidating certain effector cells and molecules responsible for pathologies associated with inflammatory and tissue damage. In recent years, population genetics approaches have greatly enriched our knowledge regarding genetic susceptibility of autoimmunity, providing us with a window of opportunities to comprehensively re-examine autoimmunity-associated genes and possible pathways. In this review, we aim to discuss etiology and pathogenesis of common autoimmune disorders from the perspective of human genetics. An overview of the genetic basis of autoimmunity is followed by 3 chapters detailing susceptibility genes involved in innate immunity, adaptive immunity and inflammatory cell death processes respectively. With such attempts, we hope to expand the scope of thinking and bring attention to lesser appreciated molecules and pathways as important contributors of autoimmunity beyond the 'usual suspects' of a limited subset of validated therapeutic targets.
Collapse
|
35
|
Jacob J, Volpe A, Peng Q, Lechler RI, Smyth LA, Lombardi G, Fruhwirth GO. Radiolabelling of Polyclonally Expanded Human Regulatory T Cells (Treg) with 89Zr-oxine for Medium-Term In Vivo Cell Tracking. Molecules 2023; 28:1482. [PMID: 36771148 PMCID: PMC9920634 DOI: 10.3390/molecules28031482] [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: 12/15/2022] [Revised: 01/25/2023] [Accepted: 01/31/2023] [Indexed: 02/05/2023] Open
Abstract
Regulatory T cells (Tregs) are a promising candidate cell therapy to treat autoimmune diseases and aid the longevity of transplanted solid organs. Despite increasing numbers of clinical trials using human Treg therapy, important questions pertaining to their in vivo fate, distribution, and function remain unanswered. Treg accumulation in relevant tissues was found to be crucial for Treg therapy efficacy, but existing blood-borne biomarkers are unlikely to accurately reflect the tissue state. Non-invasive Treg tracking by whole-body imaging is a promising alternative and can be achieved by direct radiolabelling of Tregs and following the radiolabelled cells with positron emission tomography (PET). Our goal was to evaluate the radiolabelling of polyclonal Tregs with 89Zr to permit their in vivo tracking by PET/CT for longer than one week with current preclinical PET instrumentation. We used [89Zr]Zr(oxinate)4 as the cell-labelling agent and achieved successful radiolabelling efficiency of human Tregs spanning 0.1-11.1 Bq 89Zr/Treg cell, which would be compatible with PET tracking beyond one week. We characterized the 89Zr-Tregs, assessing their phenotypes, and found that they were not tolerating these intracellular 89Zr amounts, as they failed to survive or expand in a 89Zr-dose-dependent manner. Even at 0.1 Bq 89Zr per Treg cell, while 89Zr-Tregs remained functional as determined by a five-day-long effector T cell suppression assay, they failed to expand beyond day 3 in vitro. Moreover, PET imaging revealed signs of 89Zr-Treg death after adoptive transfer in vivo. In summary, 89Zr labelling of Tregs at intracellular radioisotope amounts compatible with cell tracking over several weeks did not achieve the desired outcomes, as 89Zr-Tregs failed to expand and survive. Consequently, we conclude that indirect Treg labelling is likely to be the most effective alternative method to satisfy the requirements of this cell tracking scenario.
Collapse
Affiliation(s)
- Jacinta Jacob
- MRC Centre for Transplantation, Peter Gorer Department of Immunobiology, School of Immunology and Microbial Science, King’s College London, Guy’s Hospital, Tower Wing, 5th Floor, Great Maze Pond, London SE1 9RT, UK
| | - Alessia Volpe
- Imaging Therapies and Cancer Group, Comprehensive Cancer Centre, School of Cancer and Pharmaceutical Sciences, King’s College London, Guy’s Campus, New Hunt’s House, 2nd Floor, Great Maze Pond, London SE1 1UL, UK
| | - Qi Peng
- MRC Centre for Transplantation, Peter Gorer Department of Immunobiology, School of Immunology and Microbial Science, King’s College London, Guy’s Hospital, Tower Wing, 5th Floor, Great Maze Pond, London SE1 9RT, UK
- Imaging Therapies and Cancer Group, Comprehensive Cancer Centre, School of Cancer and Pharmaceutical Sciences, King’s College London, Guy’s Campus, New Hunt’s House, 2nd Floor, Great Maze Pond, London SE1 1UL, UK
| | - Robert I. Lechler
- MRC Centre for Transplantation, Peter Gorer Department of Immunobiology, School of Immunology and Microbial Science, King’s College London, Guy’s Hospital, Tower Wing, 5th Floor, Great Maze Pond, London SE1 9RT, UK
| | - Lesley A. Smyth
- School of Health, Sport and Bioscience, Stratford Campus, University of East London, London E15 4LZ, UK
| | - Giovanna Lombardi
- MRC Centre for Transplantation, Peter Gorer Department of Immunobiology, School of Immunology and Microbial Science, King’s College London, Guy’s Hospital, Tower Wing, 5th Floor, Great Maze Pond, London SE1 9RT, UK
| | - Gilbert O. Fruhwirth
- Imaging Therapies and Cancer Group, Comprehensive Cancer Centre, School of Cancer and Pharmaceutical Sciences, King’s College London, Guy’s Campus, New Hunt’s House, 2nd Floor, Great Maze Pond, London SE1 1UL, UK
| |
Collapse
|
36
|
Qiuping L, Pan P, Zhenzhen L, Zhen Z, Xuezhu Z, Shuting L. Acupuncture regulates the Th17/Treg balance and improves cognitive deficits in a rat model of vascular dementia. Heliyon 2023; 9:e13346. [PMID: 36816326 PMCID: PMC9929319 DOI: 10.1016/j.heliyon.2023.e13346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 12/20/2022] [Accepted: 01/25/2023] [Indexed: 02/02/2023] Open
Abstract
Objective The present study was developed to explore the impact of acupuncture on the Th17/Treg balance in the brain and the periphery and associated changes in cognitive deficits in a rat model of vascular dementia (VD). Methods Male Wistar rats (8 weeks old) were randomly assigned to sham-operated (Gs, n = 10), and operation (n = 30) groups. A VD model was established for all rats in the operation group via the permanent bilateral occlusion of the common carotid artery. Behavioral screening of these rats was conducted via a hidden platform trial at 2 months post-operation. These operation group rats were then further subdivided into impaired (Gi) and acupuncture (Ga) groups (n = 10/group). Acupuncture was performed over a 21-day period for rats in the Ga group. A Morris water maze (MWM) test was used to assess cognitive function for rats in all groups. Flow cytometry and fluorescent staining were used to detect Th17 and Treg cells in samples from these animals based on IL-17/FoxP3 or CD4+FoxP3+/CD4+RORγt+ staining profiles. Results Relative to the Gs group, escape latency values for rats in the Gi group were significantly increased. Following treatment, rats in the Ga group exhibited significant reductions in escape latency values as compared to rats in the Gi group (P < 0.05). The relative Treg proportion in the peripheral blood and spleen additionally trended upwards in these Ga rats as compared to those in the Gi group (P > 0.05), whereas the frequency of Th17 cells in the peripheral blood and spleen of Ga group rats trended downward relative to the Gi group (P > 0.05). Significantly fewer CD4+RORγt+ and RORγt+ cells were detected in the Ga group relative to the Gi group, whereas CD4+FoxP3+ and FoxP3+ cell counts were increased (P < 0.01). Conclusion In summary, VD model rats exhibited dysregulated Th17/Treg homeostasis. Acupuncture treatment was sufficient to reduce the frequency and numbers of Th17 cells in these animals while increasing Treg cell levels, thereby alleviating cognitive deficits with respect to both spatial learning and memory impairment. Consequently, the therapeutic benefits of such acupuncture treatment may be attributable to the regulation of the Th17/Treg balance and associated improvements in cognitive function.
Collapse
Affiliation(s)
- Liu Qiuping
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300380, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300380, China
- Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Pan Pan
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300380, China
- Yunnan University of Traditional Chinese Medicine, Kunming, Yunnan Province, 650500, China
| | - Ling Zhenzhen
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300380, China
- Department of Immune Regulation, Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Zhang Zhen
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300380, China
- Weifang Traditional Chinese Hospital, Shandong Province, 261031, China
| | - Zhang Xuezhu
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300380, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300380, China
| | - Li Shuting
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300380, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300380, China
- Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| |
Collapse
|
37
|
Iraji D, Oftedal BE, Wolff ASB. Th17 Cells: Orchestrators of Mucosal Inflammation and Potential Therapeutic Targets. Crit Rev Immunol 2023; 43:25-52. [PMID: 37831521 DOI: 10.1615/critrevimmunol.2023050360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2023]
Abstract
T helper 17 (Th17) cells represent a specialized subgroup of effector CD4+ T cells known for their role in provoking neutrophil-driven tissue inflammation, particularly within mucosal tissues. Although they are pivotal for defending the host against extracellular bacteria and fungi, they have also been associated with development of various T cell-mediated inflammatory conditions, autoimmune diseases, and even cancer. Notably, Th17 cells exhibit a dual nature, with different Th17 cell subtypes showcasing distinct effector functions and varying capacities to incite autoimmune tissue inflammation. Furthermore, Th17 cells exhibit significant plasticity, which carries important functional implications, both in terms of their expression of cytokines typically associated with other effector T cell subsets and in their interactions with regulatory CD4+ T cells. The intricate balance of Th17 cytokines can also be a double-edged sword in inflammation, autoimmunity, and cancer. Within this article, we delve into the mechanisms that govern the differentiation, function, and adaptability of Th17 cells. We culminate with an exploration of therapeutic potentials in harnessing the power of Th17 cells and their cytokines. Targeted interventions to modulate Th17 responses are emerging as promising strategies for autoimmunity, inflammation, and cancer treatment. By precisely fine-tuning Th17-related pathways, we may unlock new avenues for personalized therapeutic approaches, aiming to restore immune balance, alleviate the challenges of these disorders, and ultimately enhance the quality of life for individuals affected by them.
Collapse
Affiliation(s)
- Dorsa Iraji
- Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Bergithe E Oftedal
- Department of Clinical Science, University of Bergen, Bergen, Norway; Department of Medicine, Haukeland University Hospital, Bergen, Norway
| | - Anette S B Wolff
- Department of Clinical Science, University of Bergen, Bergen, Norway; Department of Medicine, Haukeland University Hospital, Bergen, Norway
| |
Collapse
|
38
|
Cassidy MF, Herbert ZT, Moulton VR. Splicing factor SRSF1 controls autoimmune-related molecular pathways in regulatory T cells distinct from FoxP3. Mol Immunol 2022; 152:140-152. [PMID: 36368121 DOI: 10.1016/j.molimm.2022.10.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 10/26/2022] [Accepted: 10/30/2022] [Indexed: 11/09/2022]
Abstract
Regulatory T cells (Tregs) are vital for maintaining immune self-tolerance, and their impaired function leads to autoimmune disease. Mutations in FoxP3, the master transcriptional regulator of Tregs, leads to immune dysregulation, polyendocrinopathy, enteropathy, X-linked (IPEX) syndrome in humans and the early lethal "scurfy" phenotype with multi-organ autoimmune disease in mice. We recently identified serine/arginine-rich splicing factor 1 (SRSF1) as an indispensable regulator of Treg homeostasis and function. Intriguingly, Treg-conditional SRSF1-deficient mice exhibit early lethal systemic autoimmunity with multi-organ inflammation reminiscent of the scurfy mice. Importantly, SRSF1 is decreased in T cells from patients with the autoimmune disease systemic lupus erythematosus (SLE), and low SRSF1 levels inversely correlate with disease severity. Given that the Treg-specific deficiency of SRSF1 causes similarly profound autoimmune disease outcomes in mice as the deficiency/mutation in FoxP3, we aimed to evaluate the genes and molecular pathways controlled by these two indispensable regulatory proteins. We performed comparative bioinformatic analyses of transcriptomic profiles of Tregs from Srsf1-knockout mice and two Foxp3 mutant mice--the FoxP3-deficient ΔFoxp3 and the Foxp3 M370I mutant mice. We identified 132 differentially expressed genes (DEGs) unique to Srsf1-ko Tregs, 503 DEGs unique to Foxp3 M370I Tregs, and 1367 DEGs unique to ΔFoxp3 Tregs. Gene set enrichment and pathway analysis of DEGs unique to Srsf1-ko Tregs indicate that SRSF1 controls cytokine and immune response pathways. Conversely, FoxP3 controls pathways involved in DNA replication and cell cycle. Besides the distinct gene signatures, we identified only 30 shared genes between all three Treg mutants, mostly contributing to cytokine and immune defense pathways. Prominent genes included the chemokines CXCR6 and CCL1 and the checkpoint inhibitors FASLG and PDCD1. Thus, we demonstrate that SRSF1 and FoxP3 control common and distinct molecular pathways implicated in autoimmunity. Our analyses suggest that SRSF1 controls crucial immune functions in Tregs contributing to immune tolerance, and perturbations in its levels lead to systemic autoimmunity via mechanisms that are largely distinct from FoxP3.
Collapse
Affiliation(s)
- Michael F Cassidy
- Division of Rheumatology and Clinical Immunology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States; Tufts University School of Medicine, Boston, MA, United States.
| | - Zachary T Herbert
- Molecular Biology Core Facilities, Dana Farber Cancer Institute, Harvard Medical School, Boston, MA, United States
| | - Vaishali R Moulton
- Division of Rheumatology and Clinical Immunology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
39
|
Li W, An N, Wang M, Liu X, Mei Z. Interleukin-23 receptor defines T helper 1-like regulatory T cells in oral squamous cell carcinoma. Immun Inflamm Dis 2022; 10:e746. [PMID: 36444617 PMCID: PMC9682469 DOI: 10.1002/iid3.746] [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: 08/22/2022] [Revised: 10/26/2022] [Accepted: 11/08/2022] [Indexed: 11/24/2022] Open
Abstract
BACKGROUND The immune responses play significant roles in the onset, progression, and outcome of oral squamous cell carcinoma (OSCC). CD4+ regulatory T cells (Tregs) significantly impact tumor immunity. However, their role in OSCC development remains elusive. METHODS In a carcinogen-induced mouse OSCC model, interleukin-23 receptor (IL-23R) expression on Tregs and Treg function were determined by flow cytometry. IL-23R overexpression in Tregs was achieved by lentiviral infection, followed by evaluation of the expression of Forkhead box P3 (Foxp3), T-bet, retineic-acid-receptor-related orphan nuclear receptor gamma t, and cytokines by flow cytometry. Adoptive transfer assays were applied to analyze the function of IL-23R- overexpressing Tregs in vivo. The cellular sources of IL-23 were also determined by flow cytometry. RESULTS IL-23R- Tregs and IL-23R+ Tregs were found in the tongues but not spleens of OSCC-bearing mice. IL-23R+ Tregs expressed lower Foxp3 but higher T-bet than IL-23R- Tregs. IL-23R- Tregs produced abundant IL-10 and transforming growth factor (TGF)-β, while IL-23R+ Tregs produced lower IL-10 and TGF-β but remarkably higher interferon (IFN)-γ. Furthermore, IL-23R+ Tregs possessed more phosphorylated signal transducer and activator of transcription (STAT3) and STAT4 than IL-23R- Tregs. IL-23R+ Tregs were less immunosuppressive than IL-23R- Tregs, as evidenced by weaker inhibition of activated conventional T cells. IL-23R overexpression in splenic Tregs remarkably reduced the expression of IL-10 and TGF-β but increased IFN-γ expression when Tregs were adoptively transferred into OSCC-bearing mice. In the OSCC microenvironment, macrophages, dendritic cells, and malignant OSCC cells produced IL-23 which might modulate the function of IL-23R+ Tregs. CONCLUSIONS This study unveils Treg heterogeneity, thus deepening the understanding of Treg biology and tumor immunity in OSCC.
Collapse
Affiliation(s)
- Wei Li
- Department of Head and Neck Surgery, Hubei Cancer Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Ning An
- Department of Head and Neck Surgery, Hubei Cancer Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Mingwei Wang
- Department of Pathology, Hubei Cancer Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Xiguo Liu
- Department of Head and Neck Surgery, Hubei Cancer Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Zhidan Mei
- Department of Head and Neck Surgery, Hubei Cancer Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| |
Collapse
|
40
|
Jiao WE, Xu S, Qiao YL, Kong YG, Sun L, Deng YQ, Yang R, Tao ZZ, Hua QQ, Chen SM. Notch2-dependent GATA3+ Treg cells alleviate allergic rhinitis by suppressing the Th2 cell response. Int Immunopharmacol 2022; 112:109261. [PMID: 36155282 DOI: 10.1016/j.intimp.2022.109261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Revised: 09/10/2022] [Accepted: 09/14/2022] [Indexed: 11/17/2022]
Abstract
The aim of this study was to investigate the role and mechanism of Notch2-dependent GATA3+ Treg cells in allergic rhinitis (AR). Samples were collected from patients in the control and AR groups to detect differences in the numbers of GATA3+ Treg cells and their intracellular Notch2 levels. The effects of Notch2 on GATA3+ Treg cell differentiation and function in vitro were detected. AR mice were subjected to adoptive transfer of GATA3+ Treg cells to detect changes in the allergic inflammatory response and Th2 cells. Mice with Treg cell-specific knockout of Notch2 were constructed, and an AR model was established to detect the changes. The number of GATA3+ Treg cells and intracellular Notch2 expression in peripheral blood of the AR group were decreased compared with the controls (P < 0.05), and the number of GATA3+ Treg cells was significantly negatively correlated with the level of allergen-specific IgE (sIgE; P < 0.01). In vitro experiments showed that Notch2 promoted the differentiation and immunosuppressive function of GATA3+ Treg cells, and Notch2 directly promoted GATA3 transcription in Treg cells (P < 0.05). Animal experiments indicated that adoptive transfer of GATA3+ Treg cells reduced the allergic inflammatory response in AR mice (P < 0.05). The number of GATA3+ Treg cells was decreased in gene knockout mice (P < 0.05), and autoimmune inflammation was observed. After modeling, the allergic inflammatory response was further aggravated (P < 0.05). Overall, our findings indicate that Notch2 alleviates AR by specifically increasing GATA3+ Treg cell differentiation. Notch2 expressed in Treg cells is expected to be a new therapeutic target for AR.
Collapse
Affiliation(s)
- Wo-Er Jiao
- Department of Otolaryngology-Head and Neck Surgery, Central Laboratory, Renmin Hospital of Wuhan University, 238 Jie-Fang Road, Wuhan, Hubei 430060, PR China
| | - Shan Xu
- Department of Otolaryngology-Head and Neck Surgery, Central Laboratory, Renmin Hospital of Wuhan University, 238 Jie-Fang Road, Wuhan, Hubei 430060, PR China
| | - Yue-Long Qiao
- Department of Otolaryngology-Head and Neck Surgery, Central Laboratory, Renmin Hospital of Wuhan University, 238 Jie-Fang Road, Wuhan, Hubei 430060, PR China
| | - Yong-Gang Kong
- Department of Otolaryngology-Head and Neck Surgery, Central Laboratory, Renmin Hospital of Wuhan University, 238 Jie-Fang Road, Wuhan, Hubei 430060, PR China
| | - Liu Sun
- Department of Otolaryngology-Head and Neck Surgery, General Hospital of The Central Theater Command, Wuhan 430070, Hubei, PR China
| | - Yu-Qin Deng
- Department of Otolaryngology-Head and Neck Surgery, Central Laboratory, Renmin Hospital of Wuhan University, 238 Jie-Fang Road, Wuhan, Hubei 430060, PR China
| | - Rui Yang
- Department of Otolaryngology-Head and Neck Surgery, Central Laboratory, Renmin Hospital of Wuhan University, 238 Jie-Fang Road, Wuhan, Hubei 430060, PR China
| | - Ze-Zhang Tao
- Department of Otolaryngology-Head and Neck Surgery, Central Laboratory, Renmin Hospital of Wuhan University, 238 Jie-Fang Road, Wuhan, Hubei 430060, PR China; Institute of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, 238 Jie-Fang Road, Wuhan, Hubei 430060, PR China
| | - Qing-Quan Hua
- Department of Otolaryngology-Head and Neck Surgery, Central Laboratory, Renmin Hospital of Wuhan University, 238 Jie-Fang Road, Wuhan, Hubei 430060, PR China; Institute of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, 238 Jie-Fang Road, Wuhan, Hubei 430060, PR China.
| | - Shi-Ming Chen
- Department of Otolaryngology-Head and Neck Surgery, Central Laboratory, Renmin Hospital of Wuhan University, 238 Jie-Fang Road, Wuhan, Hubei 430060, PR China; Institute of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, 238 Jie-Fang Road, Wuhan, Hubei 430060, PR China.
| |
Collapse
|
41
|
Yang Y, Liu K, Liu M, Zhang H, Guo M. EZH2: Its regulation and roles in immune disturbance of SLE. Front Pharmacol 2022; 13:1002741. [DOI: 10.3389/fphar.2022.1002741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 09/23/2022] [Indexed: 11/13/2022] Open
Abstract
The pathogenesis of systemic lupus erythematosus (SLE) is related to immune homeostasis imbalance. Epigenetic mechanisms have played a significant role in breaking immune tolerance. Enhancer of zeste homolog 2 (EZH2), the specific methylation transferase of lysine at position 27 of histone 3, is currently found to participate in the pathogenesis of SLE through affecting multiple components of the immune system. This review mainly expounds the mechanisms underlying EZH2-mediated disruption of immune homeostasis in SLE patients, hoping to provide new ideas in the pathogenesis of SLE and new targets for future treatment.
Collapse
|
42
|
Lentz LS, Stutz AJ, Meyer N, Schubert K, Karkossa I, von Bergen M, Zenclussen AC, Schumacher A. Human chorionic gonadotropin promotes murine Treg cells and restricts pregnancy-harmful proinflammatory Th17 responses. Front Immunol 2022; 13:989247. [PMID: 36203576 PMCID: PMC9531259 DOI: 10.3389/fimmu.2022.989247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Accepted: 08/25/2022] [Indexed: 11/15/2022] Open
Abstract
An equilibrium between proinflammatory and anti-inflammatory immune responses is essential for maternal tolerance of the fetus throughout gestation. To study the participation of fetal tissue-derived factors in this delicate immune balance, we analyzed the effects of human chorionic gonadotropin (hCG) on murine Treg cells and Th17 cells in vitro, and on pregnancy outcomes, fetal and placental growth, blood flow velocities and remodeling of the uterine vascular bed in vivo. Compared with untreated CD4+CD25+ T cells, hCG increased the frequency of Treg cells upon activation of the LH/CG receptor. hCG, with the involvement of IL-2, also interfered with induced differentiation of CD4+ T cells into proinflammatory Th17 cells. In already differentiated Th17 cells, hCG induced an anti-inflammatory profile. Transfer of proinflammatory Th17 cells into healthy pregnant mice promoted fetal rejection, impaired fetal growth and resulted in insufficient remodeling of uterine spiral arteries, and abnormal flow velocities. Our works show that proinflammatory Th17 cells have a negative influence on pregnancy that can be partly avoided by in vitro re-programming of proinflammatory Th17 cells with hCG.
Collapse
Affiliation(s)
- Lea S. Lentz
- Experimental Obstetrics and Gynecology, Medical Faculty, Health Campus Immunology, Infectilogy and Inflammation (GC-I), Otto-von-Guericke University, Magdeburg, Germany
| | - Annika J. Stutz
- Experimental Obstetrics and Gynecology, Medical Faculty, Health Campus Immunology, Infectilogy and Inflammation (GC-I), Otto-von-Guericke University, Magdeburg, Germany
| | - Nicole Meyer
- Experimental Obstetrics and Gynecology, Medical Faculty, Health Campus Immunology, Infectilogy and Inflammation (GC-I), Otto-von-Guericke University, Magdeburg, Germany
- Department of Environmental Immunology, Helmholtz Centre for Environmental Research, Leipzig, Germany
| | - Kristin Schubert
- Department of Molecular Systems Biology, Helmholtz Centre for Environmental Research, Leipzig, Germany
| | - Isabel Karkossa
- Department of Molecular Systems Biology, Helmholtz Centre for Environmental Research, Leipzig, Germany
| | - Martin von Bergen
- Department of Molecular Systems Biology, Helmholtz Centre for Environmental Research, Leipzig, Germany
- Faculty of Life Sciences, Institute of Biochemistry, University of Leipzig, Leipzig, Germany
- German Centre for Integrative Biodiversity Research (iDiv) Halle-Jena-Leipzig, Leipzig, Germany
| | - Ana C. Zenclussen
- Experimental Obstetrics and Gynecology, Medical Faculty, Health Campus Immunology, Infectilogy and Inflammation (GC-I), Otto-von-Guericke University, Magdeburg, Germany
- Department of Environmental Immunology, Helmholtz Centre for Environmental Research, Leipzig, Germany
| | - Anne Schumacher
- Experimental Obstetrics and Gynecology, Medical Faculty, Health Campus Immunology, Infectilogy and Inflammation (GC-I), Otto-von-Guericke University, Magdeburg, Germany
- Department of Environmental Immunology, Helmholtz Centre for Environmental Research, Leipzig, Germany
- *Correspondence: Anne Schumacher,
| |
Collapse
|
43
|
Qiao YL, Jiao WE, Xu S, Kong YG, Deng YQ, Yang R, Hua QQ, Chen SM. Allergen immunotherapy enhances the immunosuppressive effects of Treg cells to alleviate allergic rhinitis by decreasing PU-1+ Treg cell numbers. Int Immunopharmacol 2022; 112:109187. [PMID: 36037652 DOI: 10.1016/j.intimp.2022.109187] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 08/18/2022] [Accepted: 08/19/2022] [Indexed: 11/18/2022]
Abstract
OBJECTIVE To investigate the role of Tregs and their subtypes in the treatment of allergic rhinitis with allergen immunotherapy (AIT) as well as the underlying mechanism. METHODS 1. Thirty-one healthy controls, 29 Allergic rhinitis (AR) patients and 16 AR patients treated with AIT were recruited. The total nasal symptom scores (TNSSs) were calculated. The serum levels of IgE, IL-2, TNF-α, IFN-γ, IL-4, IL-5, IL-6, IL-10 and IL-17 were measured. 2. Changes in the proportions of CD4+ T cells, Treg cells, Treg subtypes and Th1/Th2/Th9/Th17/Tfh cells in the peripheral blood of the subjects in the three groups were measured. 3. The correlations of Treg cells, Treg subtypes and TNSS with the levels of various cytokines in the AR group and AIT group were analysed. RESULTS 1. Compared with the control group, the TNSS and IgE, IL-5 and IL-6 levels in the AR group were significantly increased, while the IL-2, IFN-γ and IL-10 levels were significantly decreased (P < 0.05). Compared with the AR group, the TNSS and IgE, IL-5 and IL-6 levels in the AIT group were significantly decreased, while the IL-2, IFN-γ and IL-10 levels were significantly increased (P < 0.05). 2. Compared with the control group, the proportions of Tregs, GATA3+ Tregs and Th1 cells in the AR group were significantly reduced, while the proportions of PU-1+ Tregs, T-bet+ Tregs and Th2 cells were significantly increased (P < 0.05). Compared with the AR group, the proportions of Tregs and Th1 cells in the AIT group were significantly increased, while the proportions of PU-1+ Tregs and Th2 cells were decreased (P < 0.05). 3. Correlation analysis showed that Treg cell proportions were negatively correlated with the TNSS, sIgE levels, IL-5 levels and IL-6 levels but positively correlated with the IL-2 and IL-10 levels (P < 0.05). PU-1+ Treg cell proportions were positively correlated with the TNSS, sIgE levels, IL-5 levels and IL-6 levels but negatively correlated with the Treg cell proportions, IL-2 levels and IL-10 levels (P < 0.05). CONCLUSIONS AIT can reduce the proportions of PU-1+ Treg subtypes in AR patients. PU-1+ Treg cell numbers can potentially be used as an indicator to monitor the therapeutic effect of AIT on AR.
Collapse
Affiliation(s)
- Yue-Long Qiao
- Department of Otolaryngology-Head and Neck Surgery, Central Laboratory, Renmin Hospital of Wuhan University, 238 Jie-Fang Road, Wuhan, Hubei 430060, PR China
| | - Wo-Er Jiao
- Department of Otolaryngology-Head and Neck Surgery, Central Laboratory, Renmin Hospital of Wuhan University, 238 Jie-Fang Road, Wuhan, Hubei 430060, PR China
| | - Shan Xu
- Department of Otolaryngology-Head and Neck Surgery, Central Laboratory, Renmin Hospital of Wuhan University, 238 Jie-Fang Road, Wuhan, Hubei 430060, PR China
| | - Yong-Gang Kong
- Department of Otolaryngology-Head and Neck Surgery, Central Laboratory, Renmin Hospital of Wuhan University, 238 Jie-Fang Road, Wuhan, Hubei 430060, PR China
| | - Yu-Qin Deng
- Department of Otolaryngology-Head and Neck Surgery, Central Laboratory, Renmin Hospital of Wuhan University, 238 Jie-Fang Road, Wuhan, Hubei 430060, PR China
| | - Rui Yang
- Department of Otolaryngology-Head and Neck Surgery, Central Laboratory, Renmin Hospital of Wuhan University, 238 Jie-Fang Road, Wuhan, Hubei 430060, PR China
| | - Qing-Quan Hua
- Department of Otolaryngology-Head and Neck Surgery, Central Laboratory, Renmin Hospital of Wuhan University, 238 Jie-Fang Road, Wuhan, Hubei 430060, PR China.
| | - Shi-Ming Chen
- Department of Otolaryngology-Head and Neck Surgery, Central Laboratory, Renmin Hospital of Wuhan University, 238 Jie-Fang Road, Wuhan, Hubei 430060, PR China.
| |
Collapse
|
44
|
Sakowska J, Arcimowicz Ł, Jankowiak M, Papak I, Markiewicz A, Dziubek K, Kurkowiak M, Kote S, Kaźmierczak-Siedlecka K, Połom K, Marek-Trzonkowska N, Trzonkowski P. Autoimmunity and Cancer-Two Sides of the Same Coin. Front Immunol 2022; 13:793234. [PMID: 35634292 PMCID: PMC9140757 DOI: 10.3389/fimmu.2022.793234] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Accepted: 04/12/2022] [Indexed: 02/06/2023] Open
Abstract
Autoimmune disease results from the immune response against self-antigens, while cancer develops when the immune system does not respond to malignant cells. Thus, for years, autoimmunity and cancer have been considered as two separate fields of research that do not have a lot in common. However, the discovery of immune checkpoints and the development of anti-cancer drugs targeting PD-1 (programmed cell death receptor 1) and CTLA-4 (cytotoxic T lymphocyte antigen 4) pathways proved that studying autoimmune diseases can be extremely helpful in the development of novel anti-cancer drugs. Therefore, autoimmunity and cancer seem to be just two sides of the same coin. In the current review, we broadly discuss how various regulatory cell populations, effector molecules, genetic predisposition, and environmental factors contribute to the loss of self-tolerance in autoimmunity or tolerance induction to cancer. With the current paper, we also aim to convince the readers that the pathways involved in cancer and autoimmune disease development consist of similar molecular players working in opposite directions. Therefore, a deep understanding of the two sides of immune tolerance is crucial for the proper designing of novel and selective immunotherapies.
Collapse
Affiliation(s)
- Justyna Sakowska
- Department of Medical Immunology, Medical University of Gdańsk, Gdańsk, Poland
| | - Łukasz Arcimowicz
- International Centre for Cancer Vaccine Science, University of Gdańsk, Gdańsk, Poland
| | - Martyna Jankowiak
- Department of Medical Immunology, Medical University of Gdańsk, Gdańsk, Poland
| | - Ines Papak
- International Centre for Cancer Vaccine Science, University of Gdańsk, Gdańsk, Poland
| | - Aleksandra Markiewicz
- Laboratory of Translational Oncology, Intercollegiate Faculty of Biotechnology, University of Gdańsk and Medical University of Gdańsk, Gdańsk, Poland
| | - Katarzyna Dziubek
- International Centre for Cancer Vaccine Science, University of Gdańsk, Gdańsk, Poland
| | - Małgorzata Kurkowiak
- International Centre for Cancer Vaccine Science, University of Gdańsk, Gdańsk, Poland
| | - Sachin Kote
- International Centre for Cancer Vaccine Science, University of Gdańsk, Gdańsk, Poland
| | | | - Karol Połom
- Department of Surgical Oncology, Medical University of Gdańsk, Gdańsk, Poland
| | - Natalia Marek-Trzonkowska
- International Centre for Cancer Vaccine Science, University of Gdańsk, Gdańsk, Poland
- Laboratory of Immunoregulation and Cellular Therapies, Department of Family Medicine, Medical University of Gdańsk, Gdańsk, Poland
| | - Piotr Trzonkowski
- Department of Medical Immunology, Medical University of Gdańsk, Gdańsk, Poland
| |
Collapse
|
45
|
Frequency of CD4+ regulatory T cells and modulation of CD4+T lymphocyte activation in pleural tuberculoma. Tuberculosis (Edinb) 2022; 134:102210. [DOI: 10.1016/j.tube.2022.102210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 04/17/2022] [Accepted: 04/24/2022] [Indexed: 11/21/2022]
|
46
|
Tagkareli S, Salagianni M, Galani I, Manioudaki M, Pavlos E, Thanopoulou K, Andreakos E. CD103 integrin identifies a high IL-10-producing FoxP3 + regulatory T-cell population suppressing allergic airway inflammation. Allergy 2022; 77:1150-1164. [PMID: 34658046 DOI: 10.1111/all.15144] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 09/10/2021] [Accepted: 10/03/2021] [Indexed: 12/14/2022]
Abstract
BACKGROUND Although FoxP3+ regulatory T (Treg) cells constitute a highly heterogeneous population, with different regulatory potential depending on the disease context, distinct subsets or phenotypes remain poorly defined. This hampers the development of immunotherapy for allergic and autoimmune disorders. The present study aimed at characterizing distinct FoxP3+ Treg subpopulations involved in the suppression of Th2-mediated allergic inflammation in the lung. METHODS We used an established mouse model of allergic airway disease based on ovalbumin sensitization and challenge to analyze FoxP3+ Tregs during the induction and resolution of inflammation, and identify markers that distinguish their most suppressive phenotypes. We also developed a new knock-in mouse model (Foxp3cre Cd103dtr ) enabling the specific ablation of CD103+ FoxP3+ Tregs for functional studies. RESULTS We found that during resolution of allergic airway inflammation in mice >50% of FoxP3+ Treg cells expressed the integrin CD103 which marks FoxP3+ Treg cells of high IL-10 production, increased expression of immunoregulatory molecules such as KLRG1, ICOS and CD127, and enhanced suppressive capacity for Th2-mediated inflammatory responses. CD103+ FoxP3+ Tregs were essential for keeping allergic inflammation under control as their specific depletion in Foxp3cre Cd103dtr mice lead to severe alveocapillary damage, eosinophilic pneumonia, and markedly reduced lifespan of the animals. Conversely, adoptive transfer of CD103+ FoxP3+ Tregs effectively treated disease, attenuating Th2 responses and allergic inflammation in an IL-10-dependent manner. CONCLUSIONS Our study identifies a novel regulatory T-cell population, defined by CD103 expression, programmed to prevent exuberant type 2 inflammation and keep homeostasis in the respiratory tract under control. This has important therapeutic implications.
Collapse
Affiliation(s)
- Sofia Tagkareli
- Laboratory of Immunobiology Center for Clinical, Experimental Surgery and Translational ResearchBiomedical Research Foundation of the Academy of Athens Athens Greece
| | - Maria Salagianni
- Laboratory of Immunobiology Center for Clinical, Experimental Surgery and Translational ResearchBiomedical Research Foundation of the Academy of Athens Athens Greece
| | - Ioanna‐Evdokia Galani
- Laboratory of Immunobiology Center for Clinical, Experimental Surgery and Translational ResearchBiomedical Research Foundation of the Academy of Athens Athens Greece
| | - Maria Manioudaki
- Laboratory of Immunobiology Center for Clinical, Experimental Surgery and Translational ResearchBiomedical Research Foundation of the Academy of Athens Athens Greece
| | - Eleftherios Pavlos
- Laboratory of Immunobiology Center for Clinical, Experimental Surgery and Translational ResearchBiomedical Research Foundation of the Academy of Athens Athens Greece
| | - Kalliopi Thanopoulou
- Laboratory of Immunobiology Center for Clinical, Experimental Surgery and Translational ResearchBiomedical Research Foundation of the Academy of Athens Athens Greece
| | - Evangelos Andreakos
- Laboratory of Immunobiology Center for Clinical, Experimental Surgery and Translational ResearchBiomedical Research Foundation of the Academy of Athens Athens Greece
- Airway Disease Infection Section National Heart and Lung InstituteImperial College London London UK
| |
Collapse
|
47
|
Zhang R, Miao J, Zhang K, Zhang B, Luo X, Sun H, Zheng Z, Zhu P. Th1-Like Treg Cells Are Increased But Deficient in Function in Rheumatoid Arthritis. Front Immunol 2022; 13:863753. [PMID: 35603149 PMCID: PMC9114761 DOI: 10.3389/fimmu.2022.863753] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Accepted: 04/07/2022] [Indexed: 12/22/2022] Open
Abstract
OBJECTIVES This study aimed to investigate the changes in quantity and function of T helper (Th)-like T regulatory (Treg) cell subsets in peripheral blood (PB) and synovial fluid (SF) of rheumatoid arthritis (RA) patients and to understand their relationship with disease activity. METHODS A total of 86 RA patients and 76 gender and age-matched healthy controls (HC) were enrolled in this study. Th-like Treg frequency and function were determined using flow cytometry. The inhibitory function of Th-like Treg cells was detected using an in vitro co-culture suppression assay. RESULTS The proportion and absolute number of Th1-like Treg cells from RA PB and RA SF were significantly higher than those of HC PB. In RA SF, the proportions of Treg cells and Th1-like Treg cells were significantly lower in the elevated erythrocyte sedimentation rate or the C-Reactive Protein group, and in the positive groups of anti-CCP antibody and anti-MCV antibody. Additionally, the proportions of Treg cells and Th1-like Treg cells from RA SF were negatively correlated with disease activity. However, the expression levels of CD73 and TGF-β1 in Th1-like Treg cells were decreased, and these Treg cells could not effectively inhibit the proliferation of effector T (Teff) cells. CONCLUSION Our data indicate that Th1-like Treg cells are the predominant Treg cell subset in RA SF, but their suppressive function is defective. Improving the function of Th1-like Treg cells may control inflammation in joints and provide new strategies for Treg-targeted therapies in RA.
Collapse
Affiliation(s)
- Rui Zhang
- Department of Clinical Immunology, Chinese People's Liberation Army. (PLA) Specialized Research Institute of Rheumatoid & Immunology, Xijing Hospital, The Fourth Military Medical University, Xi’an, China
- Department of Special Service Convalescence, Air Force Healthcare Center for Special Services, Hangzhou, China
| | - Jinlin Miao
- Department of Clinical Immunology, Chinese People's Liberation Army. (PLA) Specialized Research Institute of Rheumatoid & Immunology, Xijing Hospital, The Fourth Military Medical University, Xi’an, China
- National Translational Science Center for Molecular Medicine & Department of Cell Biology, The Fourth Military Medical University, Xi’an, China
| | - Kui Zhang
- Department of Clinical Immunology, Chinese People's Liberation Army. (PLA) Specialized Research Institute of Rheumatoid & Immunology, Xijing Hospital, The Fourth Military Medical University, Xi’an, China
| | - Bei Zhang
- Department of Clinical Immunology, Chinese People's Liberation Army. (PLA) Specialized Research Institute of Rheumatoid & Immunology, Xijing Hospital, The Fourth Military Medical University, Xi’an, China
| | - Xing Luo
- Department of Clinical Immunology, Chinese People's Liberation Army. (PLA) Specialized Research Institute of Rheumatoid & Immunology, Xijing Hospital, The Fourth Military Medical University, Xi’an, China
| | - Haoyang Sun
- Department of Clinical Immunology, Chinese People's Liberation Army. (PLA) Specialized Research Institute of Rheumatoid & Immunology, Xijing Hospital, The Fourth Military Medical University, Xi’an, China
| | - Zhaohui Zheng
- Department of Clinical Immunology, Chinese People's Liberation Army. (PLA) Specialized Research Institute of Rheumatoid & Immunology, Xijing Hospital, The Fourth Military Medical University, Xi’an, China
- *Correspondence: Zhaohui Zheng, ; Ping Zhu,
| | - Ping Zhu
- Department of Clinical Immunology, Chinese People's Liberation Army. (PLA) Specialized Research Institute of Rheumatoid & Immunology, Xijing Hospital, The Fourth Military Medical University, Xi’an, China
- *Correspondence: Zhaohui Zheng, ; Ping Zhu,
| |
Collapse
|
48
|
Cassidy MF, Herbert ZT, Moulton VR. Splicing factor SRSF1 controls distinct molecular programs in regulatory and effector T cells implicated in systemic autoimmune disease. Mol Immunol 2022; 141:94-103. [PMID: 34839165 PMCID: PMC10797198 DOI: 10.1016/j.molimm.2021.11.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 09/15/2021] [Accepted: 11/07/2021] [Indexed: 12/30/2022]
Abstract
Systemic autoimmune diseases are characterized by hyperactive effector T cells (Teffs), aberrant cytokines and chemokines, and dysfunctional regulatory T cells (Tregs). We previously uncovered new roles for serine/arginine-rich splicing factor 1 (SRSF1) in the control of genes involved in T cell signaling and cytokine production in human T cells. SRSF1 levels are decreased in T cells from patients with systemic lupus erythematosus (SLE), and low levels correlate with severe disease. Moreover, T cell-conditional Srsf1-deficient mice recapitulate the autoimmune phenotype, exhibiting CD4 T cell hyperactivity, dysfunctional Tregs, systemic autoimmunity, and tissue inflammation. However, the role of SRSF1 in controlling molecular programs in Teffs and Tregs and how these pathways are implicated in autoimmunity is not known. Here, by comparative bioinformatics analysis, we demonstrate that SRSF1 controls largely distinct gene programs in Tregs and Teffs in vivo. SRSF1 regulates 189 differentially expressed genes (DEGs) unique to Tregs, 582 DEGs unique to Teffs, and 29 DEGs shared between both. Shared genes included IL-17A, IL-17F, CSF1, CXCL10, and CXCR4, and were highly enriched for inflammatory response and cytokine-cytokine receptor interaction pathways. SRSF1 controls distinct pathways in Tregs, which include chemokine signaling and immune cell differentiation, compared with pathways in Teffs, which include cytokine production, T cell homeostasis, and activation. We identified putative mRNA binding targets of SRSF1 which include CSF1, CXCL10, and IL-17F. Finally, comparisons with transcriptomics profiles from lupus-prone MRL/lpr mice reveal that SRSF1 controls genes and pathways implicated in autoimmune disease. The target genes of SRSF1 and putative binding targets we discovered, have known roles in systemic autoimmunity. Our findings suggest that SRSF1 controls distinct molecular pathways in Tregs and Teffs and aberrant SRSF1 levels may contribute to their dysfunction and immunopathogenesis of systemic autoimmune disease.
Collapse
Affiliation(s)
- Michael F Cassidy
- Division of Rheumatology and Clinical Immunology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States; Tufts University School of Medicine, Boston, MA, United States
| | - Zachary T Herbert
- Molecular Biology Core Facilities at Dana-Farber Cancer Institute, Boston, MA, United States
| | - Vaishali R Moulton
- Division of Rheumatology and Clinical Immunology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States.
| |
Collapse
|
49
|
Shimojima Y, Kishida D, Ichikawa T, Takamatsu R, Nomura S, Sekijima Y. Oxidative Stress Promotes Instability of Regulatory T Cells in Antineutrophil Cytoplasmic Antibody-Associated Vasculitis. Front Immunol 2021; 12:789740. [PMID: 34950150 PMCID: PMC8691772 DOI: 10.3389/fimmu.2021.789740] [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: 10/05/2021] [Accepted: 11/22/2021] [Indexed: 02/02/2023] Open
Abstract
We investigated the characteristics of regulatory T cells (Tregs), focusing on the relationship between their stability and reactive oxygen species (ROS), in antineutrophil cytoplasmic antibody-associated vasculitis (AAV). Intracellular expressions of effector cytokines, forkhead box protein 3 (FoxP3), ROS, phosphorylated mammalian target of rapamycin (mTOR), and sirtuin 1 (SIRT1) in Tregs from peripheral blood mononuclear cells (PBMCs) of patients with AAV and healthy controls (HC) were analyzed. The alterations in and functional ability of Tregs were compared before and after resveratrol (RVL) treatment of PBMCs in patients with AAV. Significantly higher expressions of interferon (IFN)-γ, interleukin (IL)-17, IL-4, ROS, and phosphorylated mTOR (pho-mTOR) and lower expression of SIRT1 in CD4+CD25+FoxP3+ cells were found in patients with AAV than in the HC. FoxP3 expression in CD4+CD25+ cells and suppressive function of Tregs were significantly lower in patients with AAV than in the HC. Tregs after RVL treatment demonstrated significant decreases in IFN-γ, ROS, and pho-mTOR levels and increases in FoxP3, SIRT1 levels, and functional activity. Conversely, the direct activation of SIRT1 by SRT1720 resulted in decreased FoxP3 expression, with no reduction in ROS levels. The pho-mTOR levels were significantly higher in Tregs after activation by SRT1720 than in those after RVL treatment. This study suggested that imbalanced changes in Tregs could be attributed to mTOR activation, in which ROS overproduction was predominantly implicated. Therefore, ROS is a key mediator for promoting Tregs instability in AAV.
Collapse
Affiliation(s)
- Yasuhiro Shimojima
- Department of Medicine (Neurology and Rheumatology), Shinshu University School of Medicine, Matsumoto, Japan
| | - Dai Kishida
- Department of Medicine (Neurology and Rheumatology), Shinshu University School of Medicine, Matsumoto, Japan
| | - Takanori Ichikawa
- Department of Medicine (Neurology and Rheumatology), Shinshu University School of Medicine, Matsumoto, Japan
| | - Ryota Takamatsu
- Department of Medicine (Neurology and Rheumatology), Shinshu University School of Medicine, Matsumoto, Japan
| | - Shun Nomura
- Department of Medicine (Neurology and Rheumatology), Shinshu University School of Medicine, Matsumoto, Japan
| | - Yoshiki Sekijima
- Department of Medicine (Neurology and Rheumatology), Shinshu University School of Medicine, Matsumoto, Japan
| |
Collapse
|
50
|
Chen Z, Wang M, Yang S, Shi J, Ji T, Ding W, Jiang L, Fan Z, Chen J, Lu Y. Butyric Acid Protects Against Renal Ischemia-Reperfusion Injury by Adjusting the Treg/Th17 Balance via HO-1/p-STAT3 Signaling. Front Cell Dev Biol 2021; 9:733308. [PMID: 34796171 PMCID: PMC8593469 DOI: 10.3389/fcell.2021.733308] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 08/30/2021] [Indexed: 12/12/2022] Open
Abstract
Immune regulation plays a vital role in ischemia-reperfusion injury (IRI). Butyric acid (BA) has immunomodulatory effects in many diseases, but its immunomodulatory effects during renal IRI are still unclear. Our research shows that BA protected against IRI and significantly improved renal IRI in vivo. In vitro studies showed that BA inhibits Th17 cell differentiation and induces Treg cell differentiation. Mechanism studies have shown that heme oxygenase 1 (HO-1)/STAT3 signaling pathway was involved in the inhibitory effect of BA on Th17 cell differentiation. HO-1 inhibitors can significantly rescue the BA-mediated inhibition of Th17 cell differentiation. We confirmed that BA promotes the differentiation of Th17 cells into Treg cells by regulating the pathway and reduces renal IRI.
Collapse
Affiliation(s)
- Zhen Chen
- The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Miaomiao Wang
- The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Shikun Yang
- Key Laboratory of Liver Transplantation, Hepatobiliary/Liver Transplantation Center, The First Affiliated Hospital of Nanjing Medical University, Chinese Academy of Medical Sciences, Nanjing, China
| | - Jian Shi
- The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Tianhao Ji
- Key Laboratory of Liver Transplantation, Hepatobiliary/Liver Transplantation Center, The First Affiliated Hospital of Nanjing Medical University, Chinese Academy of Medical Sciences, Nanjing, China
| | - Wei Ding
- The Third Affiliated Hospital of Soochow University, Changzhou, China.,Wujin Hospital Affiliated With Jiangsu University, Changzhou, China
| | | | - Zhiwen Fan
- Department of Pathology, Affiliated Nanjing Drum Tower Hospital of Nanjing University School of Medicine, Nanjing, China
| | - Jing Chen
- The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Yunjie Lu
- The Third Affiliated Hospital of Soochow University, Changzhou, China
| |
Collapse
|