1
|
Wiersinga WM, Eckstein AK, Žarković M. Thyroid eye disease (Graves' orbitopathy): clinical presentation, epidemiology, pathogenesis, and management. Lancet Diabetes Endocrinol 2025:S2213-8587(25)00066-X. [PMID: 40324443 DOI: 10.1016/s2213-8587(25)00066-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Revised: 03/03/2025] [Accepted: 03/05/2025] [Indexed: 05/07/2025]
Abstract
Thyroid eye disease (TED; also known as Graves' orbitopathy), causes swollen extraocular muscles and orbital fat. Mechanistically, TED involves lid retraction, oedema and redness of the eyelids and conjunctiva, proptosis, diplopia, and optic neuropathy. Investigation of TED involves assessment of disease activity (inflammation) and disease severity. TED is predominantly mild in 77% of cases, moderate-to-severe in 22%, and rarely sight-threatening in 1% of patients. While most patients with TED have Graves' hyperthyroidism, up to 5% are euthyroid or even hypothyroid. Risk factors include male sex, older age, smoking, diabetes, hypercholesterolaemia, duration of hyperthyroidism, radioactive iodine therapy, and the presence of thyrotropin receptor (TSHR) antibodies (detectable in more than 95% of patients and directly related to TED activity and severity). Genetic immunisation of mice with TSHR, but not with insulin-like growth factor-1 receptor (IGF-1R), provides a reliable animal model of TED, demonstrating that TSHR is the primary autoantigen in the disease. Crosstalk between TSHR and IGF-1R occurs via a β-arrestin scaffold. Teprotumumab, a human monoclonal antibody that blocks IGF-1R without binding to TSHR, has been shown to significantly improve outcomes in moderate-to-severe TED, including greater proptosis reduction compared with intravenous methylprednisolone. However, its disadvantages include side effects (eg, hearing loss in 30% of patients), a high cost, and a high relapse rate. Therefore, intravenous steroids remain the treatment of choice in many parts of the world. Tocilizumab, which blocks the interleukin-6 receptor, is an effective treatment option for patients with TED who are steroid-resistant. This Review further discusses alternative medications, surgical treatments, local measures, and the importance of quality-of-life assessments and multidisciplinary care.
Collapse
Affiliation(s)
- Wilmar M Wiersinga
- Department of Endocrinology and Metabolism, Academic Medical Centre, University of Amsterdam, Amsterdam, Netherlands
| | - Anja K Eckstein
- Department of Ophthalmology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Miloš Žarković
- School of Medicine, University of Belgrade, Belgrade 11000, Serbia; Clinic for Endocrinology, Diabetes and Metabolic Disorders, Clinical Centre of Serbia, Belgrade, Serbia.
| |
Collapse
|
2
|
Wang L, Sun Y, Zhang M, He H, Wang J, Xu H, Hao Y, Zhang W, Wang Y, Chong KKL, Guo H, Shi B, Wang Y. Negative association between serum calcium and glucocorticoid-induced hypertension in thyroid-associated ophthalmopathy patients treated with methylprednisolone. Front Endocrinol (Lausanne) 2025; 16:1548953. [PMID: 40290310 PMCID: PMC12021620 DOI: 10.3389/fendo.2025.1548953] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Accepted: 03/24/2025] [Indexed: 04/30/2025] Open
Abstract
Background Hypertension is a common adverse event after systemic glucocorticoid therapy. Previous studies have suggested that blood pressure (BP) regulation is related to serum calcium. However, whether serum calcium affects the risk of glucocorticoid-induced hypertension remains understudied. Methods We used data from thyroid-associated ophthalmopathy (TAO) patients who completed a course of intravenous methylprednisolone (IVMP). Patients with high BP at baseline, a history of hypertension, and missing data were excluded. Glucocorticoid-induced hypertension was defined as systolic BP (SBP) ≥ 140 mmHg or diastolic BP (DBP) ≥ 90 mmHg during follow-up. Multivariate logistic regression and generalized additive models were used to investigate the associations between serum calcium and glucocorticoid-induced hypertension. Bar charts were used to compare the SBP and DBP fluctuations between patients with and without hypocalcemia. After accounting for missing data, all analyses were repeated in the imputed cohort. Results Serum calcium was negatively correlated with glucocorticoid-induced hypertension after adjusting for covariates with p-value < 0.1 (including age, body mass index, SBP, and DBP). For each 0.1 mmol/L increase in serum calcium, the OR (95% CI) was 0.61 (0.39, 0.95). Furthermore, a nonlinear relationship was observed, with an inflection point at 2.10 mmol/L. After the serum calcium level was converted into a categorical variable, hypocalcemia was positively associated with glucocorticoid-induced hypertension (OR = 3.26, 95% CI = 1.11-9.53). Patients with hypocalcemia exhibited significantly greater SBP fluctuations than patients without hypocalcemia (p < 0.05). These results were stable when adjusting for confounders and in the analyses of the imputed cohort. Conclusions Hypocalcemia was associated with glucocorticoid-induced hypertension in TAO patients. Further research is needed to confirm these findings in larger populations and to investigate whether calcium supplementation before glucocorticoid therapy may reduce such risk.
Collapse
Affiliation(s)
- Ling Wang
- Department of Endocrinology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Yushi Sun
- Department of Endocrinology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Meng Zhang
- Department of Endocrinology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Hairong He
- Center of Investigator Initiated Trials, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Jingya Wang
- Department of Gastroenterology, Xi’an Children’s Hospital, Shaanxi Research Institute for Pediatric Diseases, The Affiliated Children’s Hospital of Xi’an Jiaotong University and National Regional Medical Center for Children (Northwest), Xi’an, China
| | - Huayang Xu
- Department of Endocrinology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Yang Hao
- Department of Ophthalmology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Wenqiang Zhang
- Department of Endocrinology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Yawen Wang
- Department of Laboratory Medicine, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
- Biobank, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
- Shaanxi Engineering Research Center for Biobank and Advanced Medical Research, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Kelvin K. L. Chong
- Department of Ophthalmology and Visual Sciences, The Prince of Wales
Hospital, Hong Kong, Hong Kong SAR, China
- Department of Ophthalmology and Visual Sciences, Faculty of Medicine, The Chinese
University of Hong Kong, Hong Kong, Hong Kong SAR, China
- Hong Kong Eye Hospital, Hong Kong, Hong Kong SAR, China
| | - Hui Guo
- Department of Endocrinology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Bingyin Shi
- Department of Endocrinology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Yue Wang
- Department of Endocrinology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| |
Collapse
|
3
|
Lee ACH, Kahaly GJ. Targeted immunotherapies for Graves' thyroidal & orbital diseases. Front Immunol 2025; 16:1571427. [PMID: 40145088 PMCID: PMC11936961 DOI: 10.3389/fimmu.2025.1571427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Accepted: 02/21/2025] [Indexed: 03/28/2025] Open
Abstract
Background Graves' hyperthyroidism and its associated Graves' orbitopathy are common autoimmune disorders associated with significant adverse health impact. Current standard treatments have limitations regarding efficacy and safety, and most do not specifically target the pathogenic mechanisms. We aim to review the latest development of targeted immunotherapies in these two closely related disorders. Summary Targeted immunotherapies of Graves' hyperthyroidism have recently demonstrated clinical efficacy in early phase clinical studies. They include rituximab, an anti-CD20 monoclonal antibody which causes rapid B cell depletion; ATX-GD-59, an antigen specific immunotherapy which restores immune tolerance to thyrotropin receptor; iscalimab, an anti-CD40 monoclonal antibody which blocks the CD40-CD154 co-stimulatory pathway in B-T cell interaction; and K1-70, a thyrotropin receptor blocking monoclonal antibody. Furthermore, there have been major therapeutic advances in the management of Graves' orbitopathy. Mycophenolate has a dual mechanism of action both inhibiting the proliferation of activated B & T cells as well as the mammalian target of rapamycin growth intracellular pathway. Rituximab appears to be effective in active disease of recent onset without impending dysthyroid optic neuropathy. Both tocilizumab (anti-interleukin 6 receptor monoclonal antibody) and sirolimus (mammalian target of rapamycin inhibitor) showed promise in glucocorticoid resistant active disease. Teprotumumab, an anti-insulin-like growth factor-1 receptor monoclonal antibody, demonstrated remarkable all-round efficacy across a wide disease spectrum. Linsitinib, a dual small molecule inhibitor of insulin-like growth factor-1 receptor and insulin receptor, displayed significant proptosis reduction in its phase 2b/3 study. Finally, Batoclimab, an anti-neonatal fragment crystallizable receptor monoclonal antibody, which blocks recycling of pathogenic thyrotropin receptor antibody, showed promising signals for significant proptosis reduction, disease inactivation, overall response, and improvement of quality of life. Conclusion Therapeutic advances will continue to optimize our management of Graves' hyperthyroidism and its associated orbitopathy in an effective and safe manner.
Collapse
Affiliation(s)
- Alan Chun Hong Lee
- Division of Endocrinology and Metabolism, Department of Medicine, Queen Mary Hospital, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - George J. Kahaly
- Department of Medicine I, Johannes Gutenberg University (JGU) Medical Centre, Mainz, Germany
| |
Collapse
|
4
|
Huang J, Hu Y, Wang S, Liu Y, Sun X, Wang X, Yu H. Single-cell RNA sequencing in autoimmune diseases: New insights and challenges. Pharmacol Ther 2025; 267:108807. [PMID: 39894174 DOI: 10.1016/j.pharmthera.2025.108807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Revised: 01/02/2025] [Accepted: 01/29/2025] [Indexed: 02/04/2025]
Abstract
Autoimmune diseases involve a variety of cell types, yet the intricacies of their individual roles within molecular mechanisms and therapeutic strategies remain poorly understood. Single-cell RNA sequencing (scRNA-seq) offers detailed insights into transcriptional diversity at the single-cell level, significantly advancing research in autoimmune diseases. This article explores how scRNA-seq enhances the understanding of cellular heterogeneity and its potential applications in the etiology, diagnosis, treatment, and prognosis of autoimmune diseases. By revealing a comprehensive cellular landscape, scRNA-seq illuminates the functional regulation of different cell subtypes during disease progression. It aids in identifying diagnostic and prognostic markers, and analyzing cell communication networks to uncover potential therapeutic targets. Despite its valuable contributions, addressing the limitations of scRNA-seq is essential for making further advancements.
Collapse
Affiliation(s)
- Jialing Huang
- Department of Immunology, Special Key Laboratory of Ocular Diseases of Guizhou Province, Zunyi Medical University, Guizhou, China
| | - Yuelin Hu
- Department of Immunology, Special Key Laboratory of Ocular Diseases of Guizhou Province, Zunyi Medical University, Guizhou, China
| | - Shuqing Wang
- Department of Immunology, Special Key Laboratory of Ocular Diseases of Guizhou Province, Zunyi Medical University, Guizhou, China
| | - Yuefang Liu
- School of Basic Medical Sciences, Special Key Laboratory of Gene Detection and Therapy of Guizhou Province, Zunyi Medical University, Guizhou, China
| | - Xin Sun
- School of Basic Medical Sciences, Special Key Laboratory of Gene Detection and Therapy of Guizhou Province, Zunyi Medical University, Guizhou, China
| | - Xin Wang
- School of Basic Medical Sciences, Special Key Laboratory of Gene Detection and Therapy of Guizhou Province, Zunyi Medical University, Guizhou, China
| | - Hongsong Yu
- Department of Immunology, Special Key Laboratory of Ocular Diseases of Guizhou Province, Zunyi Medical University, Guizhou, China; Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Guizhou, China.
| |
Collapse
|
5
|
Al-kuraishy HM, Sulaiman GM, Mohammed HA, Abu-Alghayth MH, Albukhaty S, Jabir MS, Albuhadily AK, Al-Gareeb AI, Klionsky DJ, Abomughaid MM. The role of autophagy in Graves disease: knowns and unknowns. Front Cell Dev Biol 2025; 12:1480950. [PMID: 39834383 PMCID: PMC11743935 DOI: 10.3389/fcell.2024.1480950] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Accepted: 12/12/2024] [Indexed: 01/22/2025] Open
Abstract
Graves disease (GD), an autoimmune disease affects the thyroid gland, results in hyperthyroidisms and goiter. The main cause of GD is not clearly defined; however, stimulating autoantibodies for thyroid stimulating hormone receptor (TSHR) known as thyroid-stimulating immunoglobulins (TSIs) are the primary proposed mechanism. The TSI activation of TSHRs of thyroid gland results in excessive release of thyroid hormones with the subsequent development of hyperthyroidism and goiter. The cellular process of macroautophagy/autophagy is implicated in the pathogenesis of GD and other thyroid diseases. Autophagy plays a critical role in many thyroid diseases and in different stages of the same disease through modulation of immunity and the inflammatory response. In addition, autophagy is also implicated in the pathogenesis of thyroid-associated ophthalmopathy (TAO). However, the exact role of autophagy in GD is not well explained. Therefore, this review discusses how autophagy is intricately involved in the pathogenesis of GD regarding its protective and harmful effects.
Collapse
Affiliation(s)
- Hayder M. Al-kuraishy
- Department of Clinical Pharmacology and Medicine, College of Medicine, Mustansiriyah University, Baghdad, Iraq
| | - Ghassan M. Sulaiman
- Division of Biotechnology, Department of Applied Sciences, University of Technology, Baghdad, Iraq
| | - Hamdoon A. Mohammed
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, Qassim University, Qassim, Saudi Arabia
| | - Mohammed H. Abu-Alghayth
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, University of Bisha, Bisha, Saudi Arabia
| | - Salim Albukhaty
- Department of Laboratory Techniques, Al-Manara College for Medical Sciences, Maysan, Iraq
| | - Majid S. Jabir
- Division of Biotechnology, Department of Applied Sciences, University of Technology, Baghdad, Iraq
| | - Ali K. Albuhadily
- Department of Clinical Pharmacology and Medicine, College of Medicine, Mustansiriyah University, Baghdad, Iraq
| | - Ali I. Al-Gareeb
- Department of Clinical Pharmacology and Medicine, Jabir Ibn Hayyan Medical University, Najaf, Iraq
| | - Daniel J. Klionsky
- Life Sciences Institute, University of Michigan, Ann Arbor, MI, United States
| | - Mosleh M. Abomughaid
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, University of Bisha, Bisha, Saudi Arabia
| |
Collapse
|
6
|
Wang Z, Chen G, Li H, Liu J, Yang Y, Zhao C, Li Y, Shi J, Chen H, Chen G. Zotarolimus alleviates post-trabeculectomy fibrosis via dual functions of anti-inflammation and regulating AMPK/mTOR axis. Int Immunopharmacol 2024; 142:113176. [PMID: 39303539 DOI: 10.1016/j.intimp.2024.113176] [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: 07/24/2024] [Revised: 09/10/2024] [Accepted: 09/11/2024] [Indexed: 09/22/2024]
Abstract
OBJECTIVE Postoperative scar formation is the primary cause of uncontrolled intraocular pressure following trabeculectomy failure. This study aimed to evaluate the efficacy of zotarolimus as an adjuvant anti-scarring agent in the experimental trabeculectomy. METHODS We performed differential gene and Gene Ontology enrichment analysis on rabbit follicular transcriptome sequencing data (GSE156781). New Zealand white Rabbits were randomly assigned into three groups: Surgery only, Surgery with mitomycin-C treatment, Surgery with zotarolimus treatment. Rabbits were euthanized 3 days or 28 days post-trabeculectomy. Pathological sections were analyzed using immunohistochemistry, immunofluorescence, and Masson staining. In vitro, primary human tenon's capsule fibroblasts (HTFs) were stimulated by transforming growth factor-β1 (TGF-β1) and treated with either mitomycin-C or zotarolimus. Cell proliferation and migration were evaluated using cell counting kit-8, cell cycle, and scratch assays. Mitochondrial membrane potential was detected with the JC-1 probe, and reactive oxygen species were detected using the DCFH-DA probe. RNA and protein expressions were quantified using RT-qPCR and immunofluorescence. RESULTS Transcriptome sequencing analysis revealed the involvement of complex immune factors and metabolic disorders in trabeculectomy outcomes. Zotarolimus effectively inhibited fibrosis, reduced proinflammatory factor release and immune cell infiltration, and improved the surgical outcomes of trabeculectomy. In TGF-β1-induced HTFs, zotarolimus reduced fibrosis, proliferation, and migration without cytotoxicity via the dual regulation of the TGF-β1/Smad2/3 and AMPK/AKT/mTOR pathways. CONCLUSION Our study demonstrates that zotarolimus mitigates fibrosis by reducing immune infiltration and correcting metabolic imbalances, offering a potential treatment for improving trabeculectomy surgical outcomes.
Collapse
Affiliation(s)
- Zhiruo Wang
- Department of Ophthalmology, the Second Xiangya Hospital of Central South University, Changsha, China; Hunan Clinical Research Center of Ophthalmic Disease, Changsha, China
| | - Gong Chen
- Department of Ophthalmology, the Second Xiangya Hospital of Central South University, Changsha, China; Hunan Clinical Research Center of Ophthalmic Disease, Changsha, China
| | - Haoyu Li
- Department of Ophthalmology, the Second Xiangya Hospital of Central South University, Changsha, China; Hunan Clinical Research Center of Ophthalmic Disease, Changsha, China
| | - Jingyuan Liu
- Department of Ophthalmology, the Second Xiangya Hospital of Central South University, Changsha, China; Hunan Clinical Research Center of Ophthalmic Disease, Changsha, China
| | - Yuanyuan Yang
- Department of Ophthalmology, the Second Xiangya Hospital of Central South University, Changsha, China; Hunan Clinical Research Center of Ophthalmic Disease, Changsha, China
| | - Cong Zhao
- Department of Ophthalmology, the Second Xiangya Hospital of Central South University, Changsha, China; Hunan Clinical Research Center of Ophthalmic Disease, Changsha, China
| | - Yunping Li
- Department of Ophthalmology, the Second Xiangya Hospital of Central South University, Changsha, China; Hunan Clinical Research Center of Ophthalmic Disease, Changsha, China
| | - Jingming Shi
- Department of Ophthalmology, the Second Xiangya Hospital of Central South University, Changsha, China; Hunan Clinical Research Center of Ophthalmic Disease, Changsha, China
| | - Huihui Chen
- Department of Ophthalmology, the Second Xiangya Hospital of Central South University, Changsha, China; Hunan Clinical Research Center of Ophthalmic Disease, Changsha, China; Clinical Immunology Research Center of Central South University, Changsha, China.
| | - Guochun Chen
- Clinical Immunology Research Center of Central South University, Changsha, China; Department of Nephrology, the Second Xiangya Hospital of Central South University, Changsha, China
| |
Collapse
|
7
|
Liu Z, Ke SR, Shi ZX, Zhou M, Sun L, Sun QH, Xiao B, Wang DL, Huang YJ, Lin JS, Wang HS, Zhang QK, Pan CN, Liang XW, Chen RX, Mao Z, Lin XC. Dynamic transition of Tregs to cytotoxic phenotype amid systemic inflammation in Graves' ophthalmopathy. JCI Insight 2024; 9:e181488. [PMID: 39365735 PMCID: PMC11601897 DOI: 10.1172/jci.insight.181488] [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: 04/12/2024] [Accepted: 09/26/2024] [Indexed: 10/06/2024] Open
Abstract
Graves' disease (GD) is an autoimmune condition that can progress to Graves' ophthalmopathy (GO), leading to irreversible damage to orbital tissues and potential blindness. The pathogenic mechanism is not fully understood. In this study, we conducted single-cell multi-omics analyses on healthy individuals, patients with GD without GO, newly diagnosed patients with GO, and treated patients with GO. Our findings revealed gradual systemic inflammation during GO progression, marked by overactivation of cytotoxic effector T cell subsets, and expansion of specific T cell receptor clones. Importantly, we observed a decline in the immunosuppressive function of activated Treg (aTreg) accompanied by a cytotoxic phenotypic transition. In vitro experiments revealed that dysfunction and transition of GO-autoreactive Treg were regulated by the yin yang 1 (YY1) upon secondary stimulation of thyroid stimulating hormone receptor (TSHR) under inflammatory conditions. Furthermore, adoptive transfer experiments of the GO mouse model confirmed infiltration of these cytotoxic Treg into the orbital lesion tissues. Notably, these cells were found to upregulate inflammation and promote pathogenic fibrosis of orbital fibroblasts (OFs). Our results reveal the dynamic changes in immune landscape during GO progression and provide direct insights into the instability and phenotypic transition of Treg, offering potential targets for therapeutic intervention and prevention of autoimmune diseases.
Collapse
|
8
|
Liu J, Zhao Y, Zhao H. Chimeric antigen receptor T-cell therapy in autoimmune diseases. Front Immunol 2024; 15:1492552. [PMID: 39628482 PMCID: PMC11611814 DOI: 10.3389/fimmu.2024.1492552] [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: 09/07/2024] [Accepted: 10/28/2024] [Indexed: 12/06/2024] Open
Abstract
The administration of T cells that have been modified to carry chimeric antigen receptors (CARs) aimed at B cells has been an effective strategy in treating B cell malignancies. This breakthrough has spurred the creation of CAR T cells intended to specifically reduce or alter the faulty immune responses associated with autoimmune disorders. Early positive outcomes from clinical trials involving CAR T cells that target the B cell protein CD19 in patients suffering from autoimmune diseases driven by B cells have been reported. Additional strategies are being developed to broaden the use of CAR T cell therapy and enhance its safety in autoimmune conditions. These include employing chimeric autoantireceptors (CAAR) to specifically eliminate B cells that are reactive to autoantigens, and using regulatory T cells (Tregs) engineered to carry antigen-specific CARs for precise immune modulation. This discussion emphasizes key factors such as choosing the right target cell groups, designing CAR constructs, defining tolerable side effects, and achieving a lasting immune modification, all of which are critical for safely integrating CAR T cell therapy in treating autoimmune diseases.
Collapse
MESH Headings
- Humans
- Autoimmune Diseases/therapy
- Autoimmune Diseases/immunology
- Receptors, Chimeric Antigen/immunology
- Receptors, Chimeric Antigen/genetics
- Immunotherapy, Adoptive/methods
- Immunotherapy, Adoptive/adverse effects
- Animals
- T-Lymphocytes/immunology
- T-Lymphocytes/transplantation
- T-Lymphocytes, Regulatory/immunology
- B-Lymphocytes/immunology
- Receptors, Antigen, T-Cell/immunology
- Receptors, Antigen, T-Cell/genetics
- Autoantigens/immunology
- Antigens, CD19/immunology
Collapse
Affiliation(s)
- Jie Liu
- Department of Neurosurgery, the Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Yan Zhao
- Department of Respiratory, Lanzhou University Second Hospital, Lanzhou, Gansu, China
| | - Hai Zhao
- Department of Neurosurgery, the Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| |
Collapse
|
9
|
Su L, Mi P, Niu W, Zhou T, Yang W, Chen C, Huang C. Evaluation of 99Tcm-DTPA orbit SPECT/CT combined with thyroid function test in the treatment of radioactive iodine I-131 in patients with thyroid-associated ophthalmopathy-hyperthyroidism. J Med Biochem 2024; 43:897-907. [PMID: 39876911 PMCID: PMC11771966 DOI: 10.5937/jomb0-48734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 05/17/2024] [Indexed: 01/31/2025] Open
Abstract
Background Thyroid-associated ophthalmopathy (TAO) is an autoimmune response to inflammation of the thyroid and orbital tissue. This research evaluated the efficacy of 99Tcm-DTPA orbital SPECT/CT combined with thyroid function test in radioactive iodine I-131 (RAI) treatment of TAO-hyperthyroidism. Methods We retrospectively studied clinical activity score (CAS), blood thyrotropine (TSH), free triiodothyronine (FT3), free thyroxine (FT4), thickness of extra-ocular muscle (EOM), and uptake rate (UR) of 99Tcm-DTPA orbital SPECT/CT of 43 patients after 6 months of treatment with 20 mCi RAI. Parameters were compared before and after RAI in patients assessed as effectively treated (normal thyroid function or hypothyroidism), and correlations between blood FSH, FT3, FT4, thickness of EOM, and UR were analyzed after treatment. Results After RAI, 35 cases (70 eyes, 81.4%) had normal or hypothyroidism, and 8 cases (16 eyes, 18.6%) had hyperthyroidism. Compared with the patients who failed treatment, effectively treated patients had lower CAS, FT3, FT4, and UR and higher blood TSH. In patients with effective treatment, UR of the inferior rectus muscle was positively correlated with FT3 and FT4. Adverse RAI outcomes were associated with smoking and higher iodine-thyroid iodine uptake before treatment. Conclusions Combined with TSH, FT3, and FT4 levels, the reduction of 99Tcm-DTPA orbital SPECT/CT UR also indicates an improvement in the disease course of patients. The UR of the inferior rectus muscle can be an objective index to evaluate the curative effect of TAO patients.
Collapse
Affiliation(s)
- Li Su
- The Central Hospital of Xiaogan, Department of Nuclear Medicine, Xiaogan City, Hubei Province, China
| | - Ping Mi
- The Central Hospital of Xiaogan, Department of Nuclear Medicine, Xiaogan City, Hubei Province, China
| | - Wenqiang Niu
- The Central Hospital of Xiaogan, Department of Nuclear Medicine, Xiaogan City, Hubei Province, China
| | - Ting Zhou
- The Central Hospital of Xiaogan, Department of Nuclear Medicine, Xiaogan City, Hubei Province, China
| | - Wang Yang
- The Central Hospital of Xiaogan, Department of Nuclear Medicine, Xiaogan City, Hubei Province, China
| | - Cheng Chen
- The Central Hospital of Xiaogan, Department of Nuclear Medicine, Xiaogan City, Hubei Province, China
| | - Chenggang Huang
- The Central Hospital of Xiaogan, Department of Nuclear Medicine, Xiaogan City, Hubei Province, China
| |
Collapse
|
10
|
Ahsanuddin S, Wu AY. Single-cell transcriptomics in thyroid eye disease. Taiwan J Ophthalmol 2024; 14:554-564. [PMID: 39803402 PMCID: PMC11717346 DOI: 10.4103/tjo.tjo-d-23-00096] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 07/18/2023] [Indexed: 01/16/2025] Open
Abstract
Thyroid eye disease (TED) is a poorly understood autoimmune condition affecting the retroorbital tissue. Tissue inflammation, expansion, and fibrosis can potentially lead to debilitating sequelae such as vision loss, painful eye movement, proptosis, and eyelid retraction. Current treatment modalities for TED include systemic glucocorticoids, thioamides, methimazole, teprotumumab, beta-blockers, and radioactive iodine; however, it has been reported that up to 10%-20% of TED patients relapse after treatment withdrawal and 20%-30% are unresponsive to mainstay therapy for reasons that have yet to be more clearly elucidated. In the past 4 years, vision researchers have harnessed high-throughput single-cell RNA sequencing to elucidate the diversity of cell types and molecular mechanisms driving the pathogenesis of TED at single-cell resolution. Such studies have provided unprecedented insight regarding novel biomarkers and therapeutic targets in TED. This timely review summarizes recent breakthroughs and emerging opportunities for using single-cell and single-nuclei transcriptomic data to characterize this highly complex disease state. We also provide an overview of current challenges and future applications of this technology to potentially improve patient quality of life and facilitate reversal of disease endpoints.
Collapse
Affiliation(s)
- Sofia Ahsanuddin
- Department of Ophthalmology, Byers Eye Institute, Stanford University School of Medicine, Stanford, CA, USA
- Department of Ophthalmology, Icahn School of Medicine at Mount Sinai, New York City, NY, USA
| | - Albert Y. Wu
- Department of Ophthalmology, Byers Eye Institute, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
11
|
Lin L, Ren R, Xiong Q, Zheng C, Yang B, Wang H. Remodeling of T-cell mitochondrial metabolism to treat autoimmune diseases. Autoimmun Rev 2024; 23:103583. [PMID: 39084278 DOI: 10.1016/j.autrev.2024.103583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 07/23/2024] [Accepted: 07/24/2024] [Indexed: 08/02/2024]
Abstract
T cells are key drivers of the pathogenesis of autoimmune diseases by producing cytokines, stimulating the generation of autoantibodies, and mediating tissue and cell damage. Distinct mitochondrial metabolic pathways govern the direction of T-cell differentiation and function and rely on specific nutrients and metabolic enzymes. Metabolic substrate uptake and mitochondrial metabolism form the foundational elements for T-cell activation, proliferation, differentiation, and effector function, contributing to the dynamic interplay between immunological signals and mitochondrial metabolism in coordinating adaptive immunity. Perturbations in substrate availability and enzyme activity may impair T-cell immunosuppressive function, fostering autoreactive responses and disrupting immune homeostasis, ultimately contributing to autoimmune disease pathogenesis. A growing body of studies has explored how metabolic processes regulate the function of diverse T-cell subsets in autoimmune diseases such as systemic lupus erythematosus (SLE), multiple sclerosis (MS), autoimmune hepatitis (AIH), inflammatory bowel disease (IBD), and psoriasis. This review describes the coordination of T-cell biology by mitochondrial metabolism, including the electron transport chain (ETC), oxidative phosphorylation, amino acid metabolism, fatty acid metabolism, and one‑carbon metabolism. This study elucidated the intricate crosstalk between mitochondrial metabolic programs, signal transduction pathways, and transcription factors. This review summarizes potential therapeutic targets for T-cell mitochondrial metabolism and signaling in autoimmune diseases, providing insights for future studies.
Collapse
Affiliation(s)
- Liyan Lin
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu 610041, China; Sichuan Clinical Research Center for Laboratory Medicine, Chengdu 610041, China; Laboratory Medicine Research Center of West China Hospital, Sichuan University, Chengdu 610041, China
| | - Ruyu Ren
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu 610041, China; Sichuan Clinical Research Center for Laboratory Medicine, Chengdu 610041, China; Laboratory Medicine Research Center of West China Hospital, Sichuan University, Chengdu 610041, China
| | - Qiao Xiong
- Department of Infectious Disease, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Chunfu Zheng
- Department of Microbiology, Immunology & Infection Diseases, University of Calgary, Calgary, Alberta, Canada.
| | - Bin Yang
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu 610041, China; Sichuan Clinical Research Center for Laboratory Medicine, Chengdu 610041, China; Laboratory Medicine Research Center of West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Huiqing Wang
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
12
|
Lai KKH, Liao X, Aljufairi FMAA, Sebastian JU, Ma A, Man Wong Y, Lam Lee C, Chen W, Hu Z, Cheng GPM, Tham CC, Pang CP, Chong KKL. Ocular surface and meibomian gland evaluation in euthyroid Graves' ophthalmopathy. Int Ophthalmol 2024; 44:124. [PMID: 38430354 PMCID: PMC10908617 DOI: 10.1007/s10792-024-02919-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Accepted: 12/24/2023] [Indexed: 03/03/2024]
Abstract
PURPOSE Euthyroid Graves' ophthalmology (EGO) refers to the subgroup of thyroid eye disease patients with distinct clinical presentations. This study evaluated the ocular surface and meibomian gland changes in EGO patients. METHODS A cross-sectional study was conducted at The Chinese University of Hong Kong including 34 EGO patients and 34 age-and sex- matched healthy controls. Outcome measures include anterior segment examination, keratographic and meibographic imaging. RESULTS Between 34 EGO patients and 34 age and sex-matched healthy controls, EGO was associated with a higher ocular surface disease index (P < 0.01), higher severity of meibomian gland dropout (upper: P < 0.001, lower: P < 0.00001) and higher percentage of partial blinking (P = 0.0036). The worse affected eyes of the EGO patients were associated with corneal staining (P = 0.0019), eyelid telangiectasia (P = 0.0009), eyelid thickening (P = 0.0013), eyelid irregularity (P = 0.0054), meibomian gland plugging (P < 0.00001), expressibility (P < 0.00001), and meibum quality (P < 0.00001). When the two eyes of the same EGO patient were compared, the degree of meibomian gland dropout was higher among the worse affected eyes (upper: P < 0.00001, and lower: P < 0.00001). Tear meniscus height, lipid layer thickness, and noninvasive break-up time were comparable between the two eyes of EGO patients and also between EGO patients and healthy controls. TMH was positively correlated with the degree of exophthalmos (r = 0.383, P < 0.05). CONCLUSION EGO patients have more ocular surface complications and meibomian gland dropouts than healthy controls. Almost 60% of them had dry eye symptoms, but aqueous deficiency was not apparent. Further studies are warranted to clarify the mechanism of dry eye in EGO. (249 words).
Collapse
Affiliation(s)
- Kenneth Ka Hei Lai
- Department of Ophthalmology and Visual Sciences, Hong Kong Eye Hospital, The Chinese University of Hong Kong, 147K Argyle Street, Kowloon, Hong Kong, Hong Kong SAR
- Department of Ophthalmology and Visual Sciences, Prince of Wales Hospital, Hong Kong, Hong Kong SAR
| | - Xulin Liao
- Department of Ophthalmology and Visual Sciences, Hong Kong Eye Hospital, The Chinese University of Hong Kong, 147K Argyle Street, Kowloon, Hong Kong, Hong Kong SAR
| | - Fatema Mohamed Ali Abdulla Aljufairi
- Department of Ophthalmology and Visual Sciences, Hong Kong Eye Hospital, The Chinese University of Hong Kong, 147K Argyle Street, Kowloon, Hong Kong, Hong Kong SAR
- Department of Ophthalmology, Salmaniya Medical Complex, Government Hospitals, Manama, Bahrain
| | - Jake Uy Sebastian
- Department of Ophthalmology and Visual Sciences, Hong Kong Eye Hospital, The Chinese University of Hong Kong, 147K Argyle Street, Kowloon, Hong Kong, Hong Kong SAR
- Department of Ophthalmology, Hong Kong Sanatorium and Hospital, Hong Kong, Hong Kong SAR
| | - Andre Ma
- Department of Ophthalmology and Visual Sciences, Prince of Wales Hospital, Hong Kong, Hong Kong SAR
- Hong Kong Eye Hospital, Hong Kong, Hong Kong SAR
| | - Yiu Man Wong
- Department of Ophthalmology and Visual Sciences, Hong Kong Eye Hospital, The Chinese University of Hong Kong, 147K Argyle Street, Kowloon, Hong Kong, Hong Kong SAR
| | - Cheuk Lam Lee
- Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR
- Department of Medicine, The University of Hong Kong, Hong Kong, China
| | - Wanxue Chen
- Department of Ophthalmology and Visual Sciences, Hong Kong Eye Hospital, The Chinese University of Hong Kong, 147K Argyle Street, Kowloon, Hong Kong, Hong Kong SAR
| | - Zhichao Hu
- Department of Ophthalmology and Visual Sciences, Hong Kong Eye Hospital, The Chinese University of Hong Kong, 147K Argyle Street, Kowloon, Hong Kong, Hong Kong SAR
| | - George P M Cheng
- Department of Ophthalmology and Visual Sciences, Hong Kong Eye Hospital, The Chinese University of Hong Kong, 147K Argyle Street, Kowloon, Hong Kong, Hong Kong SAR
- Hong Kong Laser Eye Center, Hong Kong, Hong Kong SAR
| | - Clement C Tham
- Department of Ophthalmology and Visual Sciences, Hong Kong Eye Hospital, The Chinese University of Hong Kong, 147K Argyle Street, Kowloon, Hong Kong, Hong Kong SAR
- Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR
- Hong Kong Eye Hospital, Hong Kong, Hong Kong SAR
| | - Chi Pui Pang
- Department of Ophthalmology and Visual Sciences, Hong Kong Eye Hospital, The Chinese University of Hong Kong, 147K Argyle Street, Kowloon, Hong Kong, Hong Kong SAR
| | - Kelvin K L Chong
- Department of Ophthalmology and Visual Sciences, Hong Kong Eye Hospital, The Chinese University of Hong Kong, 147K Argyle Street, Kowloon, Hong Kong, Hong Kong SAR.
- Hong Kong Eye Hospital, Hong Kong, Hong Kong SAR.
| |
Collapse
|
13
|
Eckstein A, Stöhr M, Görtz GE, Gulbins A, Möller L, Fuehrer-Sakel D, Oeverhaus M. Current Therapeutic Approaches for Graves' Orbitopathy - are Targeted Therapies the Future? Klin Monbl Augenheilkd 2024; 241:48-68. [PMID: 37799096 DOI: 10.1055/a-2186-5548] [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: 10/07/2023]
Abstract
Graves' orbitopathy is an autoimmune disease of the orbit that most frequently occurs with Graves' hyperthyroidism. The occurrence of autoantibodies directed against the TSH receptor (TRAb) is of central importance for the diagnosis and pathogenesis. These autoantibodies are mostly stimulating, and induce uncontrolled hyperthyroidism and tissue remodelling in the orbit and more or less pronounced inflammation. Consequently, patients suffer to a variable extent from periocular swelling, exophthalmos, and fibrosis of the eye muscles and thus restrictive motility impairment with double vision. In recent decades, therapeutic approaches have mainly comprised immunosuppressive treatments and antithyroid drug therapy for hyperthyroidism to inhibit thyroid hormone production. With the recognition that TRAb also activates an important growth factor receptor, IGF1R (insulin-like growth factor 1 receptor), biological agents have been developed. Teprotumumab (an inhibitory IGF1R antibody) has already been approved in the USA and the therapeutic effects are enormous, especially with regard to the reduction of exophthalmos. Side effects are to be considered, especially hyperglycaemia and hearing loss. It is not yet clear whether the autoimmune reaction (development of the TRAb/attraction of immunocompetent cells) is also influenced by anti-IGF1R inhibiting agents. Recurrences after therapy show that the inhibition of antibody development must be included in the therapeutic concept, especially in severe cases.
Collapse
Affiliation(s)
- Anja Eckstein
- Klinik für Augenheilkunde, Universitätsklinikum Essen, Deutschland
| | - Mareile Stöhr
- Klinik für Augenheilkunde, Universitätsklinikum Essen, Deutschland
| | - Gina-Eva Görtz
- Labor für Molekulare Augenheilkunde, Universität Duisburg-Essen, Duisburg, Deutschland
| | - Anne Gulbins
- Labor für Molekulare Augenheilkunde, Universität Duisburg-Essen, Duisburg, Deutschland
| | - Lars Möller
- Klinik für Endokrinologie, Diabetologie und Stoffwechsel, Universitätsklinikum Essen, Deutschland
| | - Dagmar Fuehrer-Sakel
- Klinik für Endokrinologie, Diabetologie und Stoffwechsel, Universitätsklinikum Essen, Deutschland
| | - Michael Oeverhaus
- Klinik für Augenheilkunde, Universitätsklinikum Essen, Deutschland
- Gemeinschaftspraxis Dres. Oeverhaus & Weiß, Rietberg, Deutschland
| |
Collapse
|
14
|
Zhang X, Zhao Q, Li B. Current and promising therapies based on the pathogenesis of Graves' ophthalmopathy. Front Pharmacol 2023; 14:1217253. [PMID: 38035032 PMCID: PMC10687425 DOI: 10.3389/fphar.2023.1217253] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Accepted: 11/01/2023] [Indexed: 12/02/2023] Open
Abstract
Graves' ophthalmopathy (GO) is a hyperthyroidism-related and immune-mediated disease that poses a significant threat to human health. The pathogenesis of GO primarily involves T cells, B cells, and fibroblasts, suggesting a pivotal role for the thyrotropin-antibody-immunocyte-fibroblast axis. Traditional treatment approaches for Graves' disease (GD) or GO encompass antithyroid drugs (ATDs), radioactive iodine, and beta-blockers. However, despite decades of treatment, there has been limited improvement in the global incidence of GO. In recent years, promising therapies, including immunotherapy, have emerged as leading contenders, demonstrating substantial benefits in clinical trials by inhibiting the activation of immune cells like Th1 and B cells. Furthermore, the impact of diet, gut microbiota, and metabolites on GO regulation has been recognized, suggesting the potential of non-pharmaceutical interventions. Moreover, as traditional Chinese medicine (TCM) components have been extensively explored and have shown effective results in treating autoimmune diseases, remarkable progress has been achieved in managing GO with TCM. In this review, we elucidate the pathogenesis of GO, summarize current and prospective therapies for GO, and delve into the mechanisms and prospects of TCM in its treatment.
Collapse
Affiliation(s)
- Xin Zhang
- Eye School of Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Department of Ophthalmology, Chengdu Integrated TCM and Western Medicine Hospital/Chengdu First People’s Hospital, Chengdu, China
- Key Laboratory of Standardization of Chinese Medicine, Ministry of Education, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Qixiang Zhao
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Bei Li
- Department of Ophthalmology, Chengdu Integrated TCM and Western Medicine Hospital/Chengdu First People’s Hospital, Chengdu, China
| |
Collapse
|
15
|
Joachim A, Aussel R, Gélard L, Zhang F, Mori D, Grégoire C, Villazala Merino S, Gaya M, Liang Y, Malissen M, Malissen B. Defective LAT signalosome pathology in mice mimics human IgG4-related disease at single-cell level. J Exp Med 2023; 220:e20231028. [PMID: 37624388 PMCID: PMC10457416 DOI: 10.1084/jem.20231028] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 07/25/2023] [Accepted: 08/08/2023] [Indexed: 08/26/2023] Open
Abstract
Mice with a loss-of-function mutation in the LAT adaptor (LatY136F) develop an autoimmune and type 2 inflammatory disorder called defective LAT signalosome pathology (DLSP). We analyzed via single-cell omics the trajectory leading to LatY136F DLSP and the underlying CD4+ T cell diversification. T follicular helper cells, CD4+ cytotoxic T cells, activated B cells, and plasma cells were found in LatY136F spleen and lung. Such cell constellation entailed all the cell types causative of human IgG4-related disease (IgG4-RD), an autoimmune and inflammatory condition with LatY136F DLSP-like histopathological manifestations. Most previously described T cell-mediated autoimmune manifestations require persistent TCR input. In contrast, following their first engagement by self-antigens, the autoreactive TCR expressed by LatY136F CD4+ T cells hand over their central role in T cell activation to CD28 costimulatory molecules. As a result, all subsequent LatY136F DLSP manifestations, including the production of autoantibodies, solely rely on CD28 engagement. Our findings elucidate the etiology of the LatY136F DLSP and qualify it as a model of IgG4-RD.
Collapse
Affiliation(s)
- Anais Joachim
- Aix Marseille Université, INSERM, CNRS, Centre d’Immunologie de Marseille-Luminy, Marseille, France
| | - Rudy Aussel
- Aix Marseille Université, INSERM, CNRS, Centre d’Immunologie de Marseille-Luminy, Marseille, France
| | - Léna Gélard
- Aix Marseille Université, INSERM, CNRS, Centre d’Immunologie de Marseille-Luminy, Marseille, France
- Centre d’Immunophénomique, INSERM, CNRS, Aix Marseille Université, Marseille, France
| | - Fanghui Zhang
- Aix Marseille Université, INSERM, CNRS, Centre d’Immunologie de Marseille-Luminy, Marseille, France
- School of Laboratory Medicine, Henan Key Laboratory for Immunology and Targeted Therapy, Xinxiang Medical University, Xinxiang, China
| | - Daiki Mori
- Aix Marseille Université, INSERM, CNRS, Centre d’Immunologie de Marseille-Luminy, Marseille, France
- Centre d’Immunophénomique, INSERM, CNRS, Aix Marseille Université, Marseille, France
| | - Claude Grégoire
- Aix Marseille Université, INSERM, CNRS, Centre d’Immunologie de Marseille-Luminy, Marseille, France
| | - Sergio Villazala Merino
- Aix Marseille Université, INSERM, CNRS, Centre d’Immunologie de Marseille-Luminy, Marseille, France
| | - Mauro Gaya
- Aix Marseille Université, INSERM, CNRS, Centre d’Immunologie de Marseille-Luminy, Marseille, France
| | - Yinming Liang
- School of Laboratory Medicine, Henan Key Laboratory for Immunology and Targeted Therapy, Xinxiang Medical University, Xinxiang, China
| | - Marie Malissen
- Aix Marseille Université, INSERM, CNRS, Centre d’Immunologie de Marseille-Luminy, Marseille, France
- Centre d’Immunophénomique, INSERM, CNRS, Aix Marseille Université, Marseille, France
- Laboratory of Immunophenomics, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, China
| | - Bernard Malissen
- Aix Marseille Université, INSERM, CNRS, Centre d’Immunologie de Marseille-Luminy, Marseille, France
- Centre d’Immunophénomique, INSERM, CNRS, Aix Marseille Université, Marseille, France
- Laboratory of Immunophenomics, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, China
| |
Collapse
|
16
|
Wang L, Zhang M, Wang Y, Shi B. Graves' Orbitopathy Models: Valuable Tools for Exploring Pathogenesis and Treatment. Horm Metab Res 2023; 55:745-751. [PMID: 37903495 DOI: 10.1055/a-2161-5417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/01/2023]
Abstract
Graves' orbitopathy (GO) is the most common extrathyroidal complication of Graves' disease (GD) and severely affects quality of life. However, its pathogenesis is still poorly understood, and therapeutic options are limited. Animal models are important tools for preclinical research. The animals in some previous models only exhibited symptoms of hyperthyroidism without ocular lesions. With the improvements achieved in modeling methods, some progressive animal models have been established. Immunization of mice with A subunit of the human thyroid stimulating hormone receptor (TSHR) by either adenovirus or plasmid (with electroporation) is widely used and convincing. These models are successful to identify that the gut microbiota influences the occurrence and severity of GD and GO, and sex-related risk factors may be key contributors to the female bias in the occurrence of GO rather than sex itself. Some data provide insight that macrophages and CD8+ T cells may play an important pathogenic role in the early stage of GO. Our team also replicated the time window from GD onset to GO onset and identified a group of CD4+ cytotoxic T cells. In therapeutic exploration, TSHR derived peptides, fingolimod, and rapamycin offer new potential options. Further clinical trials are needed to investigate these drugs. With the increasing use of these animal models and more in-depth studies of the new findings, scientists will gain a clearer understanding of the pathogenesis of GO and identify more treatments for patients.
Collapse
Affiliation(s)
- Ling Wang
- Department of Endocrinology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Meng Zhang
- Department of Endocrinology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yue Wang
- Department of Endocrinology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Bingyin Shi
- Department of Endocrinology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
17
|
Mo K, Chu Y, Liu Y, Zheng G, Song K, Song Q, Zheng H, Tang Y, Tian X, Yao W, Fang H, Wang K, Jiang Y, Yang D, Chen Y, Huang C, Li T, Qu H, Song X, Zhou J. Targeting hnRNPC suppresses thyroid follicular epithelial cell apoptosis and necroptosis through m 6A-modified ATF4 in autoimmune thyroid disease. Pharmacol Res 2023; 196:106933. [PMID: 37729957 DOI: 10.1016/j.phrs.2023.106933] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 09/16/2023] [Accepted: 09/17/2023] [Indexed: 09/22/2023]
Abstract
Both environmental and genetic factors contribute to the etiology of autoimmune thyroid disease (AITD) including Graves' disease (GD) and Hashimoto's thyroiditis (HT). However, the exact pathogenesis and interactions that occur between environmental factors and genes remain unclear, and therapeutic targets require further investigation due to limited therapeutic options. To solve such problems, this study utilized single-cell transcriptome, whole transcriptome, full-length transcriptome (Oxford nanopore technology), and metabolome sequencing to examine thyroid lesion tissues from 2 HT patients and 2 GD patients as well as healthy thyroid tissue from 1 control subject. HT patients had increased ATF4-positive thyroid follicular epithelial (ThyFoEp) cells, which significantly increased endoplasmic reticulum stress. The enhanced sustained stress resulted in cell death mainly including apoptosis and necroptosis. The ATF4-based global gene regulatory network and experimental validation revealed that N6-methyladenosine (m6A) reader hnRNPC promoted the transcriptional activity, synthesis, and translation of ATF4 through mediating m6A modification of ATF4. Increased ATF4 expression initiated endoplasmic reticulum stress signaling, which when sustained, caused apoptosis and necroptosis in ThyFoEp cells, and mediated HT development. Targeting hnRNPC and ATF4 notably decreased ThyFoEp cell death, thus ameliorating disease progression. Collectively, this study reveals the mechanisms by which microenvironmental cells in HT and GD patients trigger and amplify the thyroid autoimmune cascade response. Furthermore, we identify new therapeutic targets for the treatment of autoimmune thyroid disease, hoping to provide a potential way for targeted therapy.
Collapse
Affiliation(s)
- Ke Mo
- Key Laboratory of Spatiotemporal Single-Cell Technologies and Translational Medicine, Yantai Yuhuangding Hospital of Qingdao University, Yantai 264000, Shandong, China; Experimental Center of BIOQGene, YuanDong International Academy of Life Sciences, 999077, Hong Kong, China
| | - Yongli Chu
- Department of Scientific Research, Yantai Yuhuangding Hospital of Qingdao University, Yantai 264000, Shandong, China
| | - Yang Liu
- Department of Thyroid Surgery, Yantai Yuhuangding Hospital of Qingdao University, Yantai 264000, Shandong, China
| | - Guibin Zheng
- Department of Thyroid Surgery, Yantai Yuhuangding Hospital of Qingdao University, Yantai 264000, Shandong, China
| | - Kaiyu Song
- Department of Endocrinology, Yantai Yuhuangding Hospital of Qingdao University, Yantai 264000, Shandong, China
| | - Qiong Song
- Key Laboratory of Spatiotemporal Single-Cell Technologies and Translational Medicine, Yantai Yuhuangding Hospital of Qingdao University, Yantai 264000, Shandong, China; Experimental Center of BIOQGene, YuanDong International Academy of Life Sciences, 999077, Hong Kong, China
| | - Haitao Zheng
- Department of Thyroid Surgery, Yantai Yuhuangding Hospital of Qingdao University, Yantai 264000, Shandong, China
| | - Yuxiao Tang
- Department of Endocrinology, Yantai Yuhuangding Hospital of Qingdao University, Yantai 264000, Shandong, China
| | - Xinghan Tian
- Department of Critical Care Medicine, Yantai Yuhuangding Hospital of Qingdao University, Yantai 264000, Shandong, China
| | - Wenjie Yao
- Department of Endocrinology, Binzhou Medical University, Yantai 264003, Shandong, China
| | - Han Fang
- Department of Otorhinolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital of Qingdao University, Yantai 264000, Shandong, China
| | - Kejian Wang
- Experimental Center of BIOQGene, YuanDong International Academy of Life Sciences, 999077, Hong Kong, China; Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250300, Shandong, China
| | - Yongqiang Jiang
- Biology Institute, Guangxi Academy of Sciences, Nanning 530007, Guangxi, China
| | - Dengfeng Yang
- Biology Institute, Guangxi Academy of Sciences, Nanning 530007, Guangxi, China
| | - Yixuan Chen
- Experimental Center of BIOQGene, YuanDong International Academy of Life Sciences, 999077, Hong Kong, China; Biology Institute, Guangxi Academy of Sciences, Nanning 530007, Guangxi, China
| | - Chengyu Huang
- Experimental Center of BIOQGene, YuanDong International Academy of Life Sciences, 999077, Hong Kong, China; Biology Institute, Guangxi Academy of Sciences, Nanning 530007, Guangxi, China
| | - Ting Li
- Experimental Center of BIOQGene, YuanDong International Academy of Life Sciences, 999077, Hong Kong, China; Biology Institute, Guangxi Academy of Sciences, Nanning 530007, Guangxi, China
| | - Hongmei Qu
- Departments of Obstetrics and Gynecology, Yantai Yuhuangding Hospital of Qingdao University, Yantai 264000, Shandong, China.
| | - Xicheng Song
- Key Laboratory of Spatiotemporal Single-Cell Technologies and Translational Medicine, Yantai Yuhuangding Hospital of Qingdao University, Yantai 264000, Shandong, China; Department of Otorhinolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital of Qingdao University, Yantai 264000, Shandong, China.
| | - Jin Zhou
- Key Laboratory of Spatiotemporal Single-Cell Technologies and Translational Medicine, Yantai Yuhuangding Hospital of Qingdao University, Yantai 264000, Shandong, China; Department of Endocrinology, Yantai Yuhuangding Hospital of Qingdao University, Yantai 264000, Shandong, China.
| |
Collapse
|
18
|
Tu W, Cao YW, Sun M, Liu Q, Zhao HG. mTOR signaling in hair follicle and hair diseases: recent progress. Front Med (Lausanne) 2023; 10:1209439. [PMID: 37727765 PMCID: PMC10506410 DOI: 10.3389/fmed.2023.1209439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Accepted: 08/23/2023] [Indexed: 09/21/2023] Open
Abstract
Mammalian target of rapamycin (mTOR) signaling pathway is a major regulator of cell proliferation and metabolism, playing significant roles in proliferation, apoptosis, inflammation, and illness. More and more evidences showed that the mTOR signaling pathway affects hair follicle circulation and maintains the stability of hair follicle stem cells. mTOR signaling may be a critical cog in Vitamin D receptor (VDR) deficiency-mediated hair follicle damage and degeneration and related alopecia disorders. This review examines the function of mTOR signaling in hair follicles and hair diseases, and talks about the underlying molecular mechanisms that mTOR signaling regulates.
Collapse
Affiliation(s)
| | | | | | | | - Heng-Guang Zhao
- Department of Dermatology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
19
|
Cao T, Xie F, Shi Y, Xu J, Liu Y, Cui D, Zhang F, Lin L, Li W, Gao Y, Ruan Y, Wang X, Zhu Y, Han B, Xia S, Guo W, Li B, Jing Y. Rapamycin and Low-dose IL-2 Mediate an Immunosuppressive Microenvironment to Inhibit Benign Prostatic Hyperplasia. Int J Biol Sci 2023; 19:3441-3455. [PMID: 37497009 PMCID: PMC10367549 DOI: 10.7150/ijbs.85089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2023] [Accepted: 06/20/2023] [Indexed: 07/28/2023] Open
Abstract
Benign prostatic hyperplasia (BPH) is a condition that becomes more common with age and manifests itself primarily as the expansion of the prostate and surrounding tissues. However, to date, the etiology of BPH remains unclear. In this respect, we performed single-cell RNA sequencing of prostate transition zone tissues from elderly individuals with different prostate volumes to reveal their distinct tissue microenvironment. Ultimately, we demonstrated that a reduced Treg/CD4+ T-cell ratio in the large-volume prostate and a relatively activated immune microenvironment were present, characterized partially by increased expression levels of granzymes, which may promote vascular growth and profibrotic processes and further exacerbate BPH progression. Consistently, we observed that the prostate gland of patients taking immunosuppressive drugs usually remained at a smaller volume. Furthermore, in mouse models, we confirmed that both suppression of the immune system with rapamycin and induction of Treg proliferation with low doses of IL-2 therapy indeed prevented the progression of BPH. Taken together, our findings suggest that an activated immune microenvironment is necessary for prostate volume growth and that Tregs can reverse this immune activation state, thereby inhibiting the progression of BPH.
Collapse
Affiliation(s)
- Tianyu Cao
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Feng Xie
- Department of Immunology and Microbiology, Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Youwei Shi
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Junhao Xu
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yi Liu
- Department of Plastic Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Di Cui
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Fang Zhang
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lihui Lin
- Department of Laboratory Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Weize Li
- Department of Laboratory Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yanting Gao
- Department of Laboratory Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuan Ruan
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaohai Wang
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yiping Zhu
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Bangmin Han
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shujie Xia
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wenhuan Guo
- Department of Pathology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Bin Li
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Department of Respiratory and Critical Care Medicine of Ruijin Hospital, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Thoracic Surgery, Clinical Translational Research Center, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
- Institute of Arthritis Research, Guanghua Integrative Medicine Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Department of Integrated TCM & Western Medicine, Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
- Henan Key Laboratory for Digestive Organ Transplantation, Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Shenzhen Key Laboratory of Immunity and Inflammatory Diseases, Shenzhen, China
| | - Yifeng Jing
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
20
|
Chen Z, Zhang M, Liu Y, Chen Z, Wang L, Wang W, Wang J, He M, Shi B, Wang Y. VEGF-A enhances the cytotoxic function of CD4 + cytotoxic T cells via the VEGF-receptor 1/VEGF-receptor 2/AKT/mTOR pathway. J Transl Med 2023; 21:74. [PMID: 36737819 PMCID: PMC9896805 DOI: 10.1186/s12967-023-03926-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 01/25/2023] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND CD4+ cytotoxic T cells (CD4 CTLs) are CD4+ T cells with major histocompatibility complex-II-restricted cytotoxic function. Under pathologic conditions, CD4 CTLs hasten the development of autoimmune disease or viral infection by enhancing cytotoxicity. However, the regulators of the cytotoxicity of CD4 CTLs are not fully understood. METHODS To explore the potential regulators of the cytotoxicity of CD4 CTLs, bulk RNA and single-cell RNA sequencing (scRNA-seq), enzyme-linked immunosorbent assay, flow cytometry, quantitative PCR, and in-vitro stimulation and inhibition assays were performed. RESULTS In this study, we found that VEGF-A promoted the cytotoxicity of CD4 CTLs through scRNA-seq and flow cytometry. Regarding the specific VEGF receptor (R) involved, VEGF-R1/R2 signaling was activated in CD4 CTLs with increased cytotoxicity, and the VEGF-A effects were inhibited when anti-VEGF-R1/R2 neutralizing antibodies were applied. Mechanistically, VEGF-A treatment activated the AKT/mTOR pathway in CD4 CTLs, and the increases of cytotoxic molecules induced by VEGF-A were significantly reduced when the AKT/mTOR pathway was inhibited. CONCLUSION In conclusion, VEGF-A enhances the cytotoxicity of CD4 CTLs through the VEGF-R1/VEGF-R2/AKT/mTOR pathway, providing insights for the development of novel treatments for disorders associated with CD4 CTLs.
Collapse
Affiliation(s)
- Ziyi Chen
- grid.452438.c0000 0004 1760 8119Department of Endocrinology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Meng Zhang
- grid.452438.c0000 0004 1760 8119Department of Endocrinology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Yufeng Liu
- grid.43169.390000 0001 0599 1243MOE Key Lab for Intelligent Networks & Networks Security, School of Electronic and Information Engineering, Xi’an Jiaotong University, Xi’an, China ,grid.452438.c0000 0004 1760 8119Genome Institute, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China ,grid.452438.c0000 0004 1760 8119BioBank, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Zhe Chen
- grid.452452.00000 0004 1757 9282Department of Spine Surgery, Hong Hui Hospital, Xi’an Jiaotong University, Xi’an, China
| | - Ling Wang
- grid.452438.c0000 0004 1760 8119Department of Endocrinology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Wenjuan Wang
- grid.452438.c0000 0004 1760 8119Department of Hematology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Jincheng Wang
- grid.452438.c0000 0004 1760 8119Department of Hematology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Mingqian He
- grid.452438.c0000 0004 1760 8119Department of Endocrinology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Bingyin Shi
- Department of Endocrinology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.
| | - Yue Wang
- Department of Endocrinology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China. .,MOE Key Lab for Intelligent Networks & Networks Security, School of Electronic and Information Engineering, Xi'an Jiaotong University, Xi'an, China. .,Genome Institute, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.
| |
Collapse
|