1
|
Roztocil E, Husain F, Patrick CC, Feldon SE, Woeller CF. Targeting the Aryl Hydrocarbon Receptor to Attenuate IGF1R Signaling in Thyroid Eye Disease. Thyroid 2025. [PMID: 40257057 DOI: 10.1089/thy.2024.0529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/22/2025]
Abstract
Background: Thyroid eye disease (TED) is an autoimmune disorder characterized by proptosis, inflammation, and fibrosis. Elevated insulin-like growth factor 1 receptor (IGF1R) signaling in TED orbital fibroblasts (OFs) drives the proliferation and biosynthesis of hyaluronan, which causes enlargement of orbital tissue volume. The aryl hydrocarbon receptor (AHR) is a ligand-activated transcription factor that regulates cellular stress responses, metabolism, and inflammation. Given its important role in regulating cellular responses, we hypothesized that activation of the AHR could limit excessive IGF1R signaling in TED OFs, offering therapeutic potential. Methods: We measured IGF1R and AHR expression levels in TED, non-TED, and non-OF controls. OF activation was analyzed using proliferation, hyaluronan accumulation, and migration assays. RNA sequencing was used to detect transcriptome-wide changes in IGF1-treated TED OFs. After gene set enrichment analysis, select gene expression changes were validated by quantitative polymerase chain reaction. OFs were treated with the AHR ligands 6-formylindolo[3,2-b]carbazole (FICZ) or tapinarof with or without IGF1. Western blotting evaluated signaling pathways impacted by AHR and IGF1R signaling. Results: TED OFs showed elevated IGF1R and AHR expression levels compared to controls. IGF1 significantly increased hyaluronan accumulation, proliferation, and migration in TED OFs compared to non-TED OFs. IGF1R signaling altered the expression of hundreds of genes controlling cell migration, proliferation, and metabolism in TED OFs. These genes included TUBA1B, TUBA1C, CRABP2 (upregulated), and IRS2 and SOD3 (downregulated). AHR activation blocked proliferation, migration, hyaluronan production, and gene expression mediated through IGF1R signaling. The AHR inhibited these pathways by reducing phosphorylation of GSK3β, an important mediator of IGF1R/β-catenin mediated signaling. Conclusions: AHR activation represents a promising therapeutic strategy for mitigating TED progression by inhibiting IGF1R signaling. Through modulation of GSK3β-mediated pathways, AHR activation may target additional pathologically relevant pathways beyond those affected by direct IGF1R inhibitors. This research provides novel insights into TED pathophysiology and offers a potential avenue for developing therapies to improve patient outcomes.
Collapse
Affiliation(s)
- Elisa Roztocil
- Flaum Eye Institute, University of Rochester, Rochester, New York, USA
| | - Farha Husain
- Flaum Eye Institute, University of Rochester, Rochester, New York, USA
| | | | - Steven E Feldon
- Flaum Eye Institute, University of Rochester, Rochester, New York, USA
| | - Collynn F Woeller
- Flaum Eye Institute, University of Rochester, Rochester, New York, USA
| |
Collapse
|
2
|
Cieplińska K, Niedziela E, Rdzanek AK, Słuszniak A, Chrapek M, Pałyga I, Kowalska A. Association between clinical activity score and serum sPD-1 and sPD-L1 levels during systemic glucocorticoid treatment for active moderate-to-severe thyroid eye disease. Cytokine 2025; 187:156862. [PMID: 39842384 DOI: 10.1016/j.cyto.2025.156862] [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: 11/11/2024] [Revised: 01/04/2025] [Accepted: 01/16/2025] [Indexed: 01/24/2025]
Abstract
BACKGROUND CD4+ T lymphocytes are key immune cells involved in orbital inflammation in thyroid eye disease (TED). Inhibition of their activity is important in treatment of TED, but effective drugs targeting these cells are lacking. The programmed cell death-1/programmed cell death ligand-1 pathway has been implicated in several T-cell-mediated diseases. Manipulation of this pathway with antagonists or agonists is an attractive therapeutic option. The role of soluble programmed cell death-1 (sPD-1) and soluble programmed cell death ligand-1 (sPD-L1) in regulation of this pathway is debated. This study aimed to investigate the involvement of sPD-1 and sPD-L1 in the pathogenesis of TED, focusing on their utility as novel biomarkers to evaluate disease severity and treatment response. METHODS Thirty patients diagnosed with moderate-to-severe TED associated with Graves' disease were included. Blood samples were collected from patients before and 12 weeks after initiation of intravenous glucocorticosteroid (IVGC) treatment. Disease severity was assessed using the Clinical Activity Score (CAS) before and after IVGC treatment. Thyroid-stimulating hormone, free thyroxine, free triiodothyronine, thyroid-stimulating immunoglobulin, interleukin-6, sPD-1, and sPD-L1 levels were measured. Correlations between sPD-1, sPD-L1, and CAS before and after IVGC treatment were investigated. Serum concentrations of sPD-1 and sPD-L1 before and after IVGC treatment in patients with TED were compared with those in healthy controls (HCs). The changes in the tested protein concentrations upon IVGC treatment and their associations with clinical characteristics were investigated. Enzyme-linked immunosorbent assays were used to measure sPD-1 and sPD-L1 concentrations in peripheral blood serum. RESULTS There was a positive correlation of moderate Spearman's rank strength between sPD-L1 and CAS before and after treatment, and a positive correlation between sPD-1 and sPD-L1. However, no correlation was observed between sPD-1 and CAS. Baseline serum levels of sPD-1 and sPD-L1 did not significantly differ between patients with TED and HCs. There were no correlations between changes in the levels of the tested molecules upon IVGC treatment and the analyzed clinical features. The decreases of sPD-1 and sPD-L1 levels after 12 weeks of IVGC treatment were not significant. CONCLUSION The positive correlation of moderate Spearman's rank strength between sPD-L1 and CAS before and after 12 weeks of treatment indicates that sPD-L1 is involved in the pathogenesis of TED. sPD-L1 may become an additional immunological biomarker to assess the disease activity and monitor the respond to treatment. Although sPD-1 is reported in the literature to have an activating effect on lymphocytes, our study shows that sPD-1 may not play a significant role in the pathogenesis of TED, as its level does not differ significantly between the TED and HC groups and does not correlate with disease activity. Understanding the clinical value of sPD-1 and sPD-L1 is of great practical importance.
Collapse
Affiliation(s)
| | - Emilia Niedziela
- Collegium Medicum, Jan Kochanowski University in Kielce, 25-317 Kielce, Poland; Department of Endocrinology, Holy Cross Cancer Center, 25-734 Kielce, Poland
| | | | - Anna Słuszniak
- Department of Tumor Markers, Holy Cross Cancer Center, 25-734 Kielce, Poland
| | - Magdalena Chrapek
- Department of Mathematics, Faculty of Natural Sciences, Jan Kochanowski University, Kielce 25-406, Poland
| | - Iwona Pałyga
- Collegium Medicum, Jan Kochanowski University in Kielce, 25-317 Kielce, Poland; Department of Endocrinology, Holy Cross Cancer Center, 25-734 Kielce, Poland
| | - Aldona Kowalska
- Collegium Medicum, Jan Kochanowski University in Kielce, 25-317 Kielce, Poland; Department of Endocrinology, Holy Cross Cancer Center, 25-734 Kielce, Poland
| |
Collapse
|
3
|
Wei L, Huang Q, Tu Y, Song S, Zhang X, Yu B, Liu Y, Li Z, Huang Q, Chen L, Liu B, Xu S, Li T, Liu X, Hu X, Liu W, Chi ZL, Wu W. Plasma exosomes from patients with active thyroid-associated orbitopathy induce inflammation and fibrosis in orbital fibroblasts. J Transl Med 2024; 22:546. [PMID: 38849907 PMCID: PMC11157872 DOI: 10.1186/s12967-024-05263-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Accepted: 04/29/2024] [Indexed: 06/09/2024] Open
Abstract
BACKGROUND The pathogenesis of thyroid-associated orbitopathy (TAO) remains incompletely understand. The interaction between immunocytes and orbital fibroblasts (OFs) play a critical role in orbital inflammatory and fibrosis. Accumulating reports indicate that a significant portion of plasma exosomes (Pla-Exos) are derived from immune cells; however, their impact upon OFs function is unclear. METHODS OFs were primary cultured from inactive TAO patients. Exosomes isolated from plasma samples of patients with active TAO and healthy controls (HCs) were utilized for functional and RNA cargo analysis. Functional analysis in thymocyte differentiation antigen-1+ (Thy-1+) OFs measured expression of inflammatory and fibrotic markers (mRNAs and proteins) and cell activity in response to Pla-Exos. RNA cargo analysis was performed by RNA sequencing and RT-qPCR. Thy-1+ OFs were transfected with miR-144-3p mimics/inhibitors to evaluate its regulation of inflammation, fibrosis, and proliferation. RESULTS Pla-Exos derived from active TAO patients (Pla-ExosTAO-A) induced stronger production of inflammatory cytokines and hyaluronic acid (HA) in Thy-1+ OFs while inhibiting their proliferation. Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis and single sample gene set enrichment analysis (ssGSEA) suggested that the difference in mRNA expression levels between Pla-ExosTAO-A and Pla-ExosHC was closely related to immune cells. Differential expression analysis revealed that 62 upregulated and 45 downregulated miRNAs in Pla-ExosTAO-A, with the elevation of miR-144-3p in both Pla-Exos and PBMCs in active TAO group. KEGG analysis revealed that the target genes of differentially expressed miRNA and miR-144-3p enriched in immune-related signaling pathways. Overexpression of the miR-144-3p mimic significantly upregulated the secretion of inflammatory cytokines and HA in Thy-1+ OFs while inhibiting their proliferation. CONCLUSION Pla-Exos derived from patients with active TAO were immune-active, which may be a long-term stimulus casual for inflammatory and fibrotic progression of TAO. Our finding suggests that Pla-Exos could be used as biomarkers or treatment targets in TAO patients.
Collapse
Affiliation(s)
- Li Wei
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
- State Key Laboratory of Ophthalmology, Optometry and Vison Science, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Qinying Huang
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
- State Key Laboratory of Ophthalmology, Optometry and Vison Science, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Yunhai Tu
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
- State Key Laboratory of Ophthalmology, Optometry and Vison Science, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Shihan Song
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
- State Key Laboratory of Ophthalmology, Optometry and Vison Science, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Xiaobo Zhang
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
- State Key Laboratory of Ophthalmology, Optometry and Vison Science, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Bo Yu
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
- State Key Laboratory of Ophthalmology, Optometry and Vison Science, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Yufen Liu
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
- State Key Laboratory of Ophthalmology, Optometry and Vison Science, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Ziwei Li
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
- State Key Laboratory of Ophthalmology, Optometry and Vison Science, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Qing Huang
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
- State Key Laboratory of Ophthalmology, Optometry and Vison Science, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Lili Chen
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
- State Key Laboratory of Ophthalmology, Optometry and Vison Science, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Bo Liu
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
- State Key Laboratory of Ophthalmology, Optometry and Vison Science, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Shenglan Xu
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
- State Key Laboratory of Ophthalmology, Optometry and Vison Science, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Tong Li
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
- State Key Laboratory of Ophthalmology, Optometry and Vison Science, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Xiyuan Liu
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
- State Key Laboratory of Ophthalmology, Optometry and Vison Science, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Xiaozhou Hu
- State Key Laboratory of Ophthalmology, Optometry and Vison Science, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Weijie Liu
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
- State Key Laboratory of Ophthalmology, Optometry and Vison Science, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Zai-Long Chi
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China.
- State Key Laboratory of Ophthalmology, Optometry and Vison Science, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China.
| | - Wencan Wu
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China.
- State Key Laboratory of Ophthalmology, Optometry and Vison Science, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China.
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain health), Wenzhou, 32500, China.
| |
Collapse
|
4
|
Alluli A, Fonseca G, Matthews J, Eidelman DH, Baglole CJ. Regulation of long non-coding RNA expression by aryl hydrocarbon receptor activation. Toxicol Lett 2024; 391:13-25. [PMID: 38036013 DOI: 10.1016/j.toxlet.2023.11.004] [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: 04/13/2023] [Revised: 10/27/2023] [Accepted: 11/13/2023] [Indexed: 12/02/2023]
Abstract
The aryl hydrocarbon receptor (AhR) is a cytosolic transcription factor that can be activated by endogenous or xenobiotic ligands. Upon activation, the AhR translocates to the nucleus, dimerizes with the AhR nuclear translator (ARNT), and binds to specific DNA sequences called xenobiotic response elements (XRE) to promote target gene transcription, including cytochrome P450 (e.g., CYP1A1) expression. In addition to mRNA, the AhR may also regulate long non-coding RNA (lncRNA) expression. lncRNA are transcripts more than 200 nucleotides in length that do not encode a protein. Herein, we tested whether AhR activation regulates the expression of lncRNA in response to benzo[a]pyrene (B[a]P) using RNA sequencing (RNA-seq). We found that many lncRNA (e.g., SATB1-AS1, MIR4290HG, AC008969.1, LINC01533, VIPR1-AS1) and protein-coding RNA (e.g., CYP1A1, BX005266.2, AQP3, BTG2, DCX, and AhRR) were differentially expressed (DE) in A549 cells treated with B[a]P; many of these genes were dependent on AhR expression including CYP1A1, CYP1B1 and TiPARP. GO analyses indicated that DE protein-coding RNAs in A549WT cells are associated with distinct molecular functions compared to A549KO cells. KEGG analyses showed the hsa01100 pathway was associated with DE lncRNA only in A549WT cells. A549KO cells treated with B[a]P exhibited a distinct set of differentially-regulated lncRNA including upregulation of HOTAIR. We further confirmed that despite AhR activation in A549WT cells, B[a]P did not alter the expression of many well-characterized lncRNA including NEAT1, HOTTIP, SOX2OT, MALAT1, H19, and Linc00673. Thus, there is control over select lncRNA expression in A549 cells exposed to B[a]P, a finding which could yield insight into the molecular function of the AhR.
Collapse
Affiliation(s)
- Aeshah Alluli
- Meakins-Christie Laboratories, McGill University, Montreal, Canada; Translational Research in Respiratory Diseases Program at the Research Institute of the McGill University Health Centre, Montreal, Canada; Department of Pathology, McGill University, Montreal, Canada
| | - Gregory Fonseca
- Meakins-Christie Laboratories, McGill University, Montreal, Canada; Translational Research in Respiratory Diseases Program at the Research Institute of the McGill University Health Centre, Montreal, Canada; Department of Medicine, McGill University, Montreal, Canada
| | - Jason Matthews
- Department of Nutrition, University of Oslo, Oslo, Norway; Department of Pharmacology & Toxicology, University of Toronto, Toronto, Canada
| | - David H Eidelman
- Meakins-Christie Laboratories, McGill University, Montreal, Canada; Department of Medicine, McGill University, Montreal, Canada
| | - Carolyn J Baglole
- Meakins-Christie Laboratories, McGill University, Montreal, Canada; Translational Research in Respiratory Diseases Program at the Research Institute of the McGill University Health Centre, Montreal, Canada; Department of Pathology, McGill University, Montreal, Canada; Department of Medicine, McGill University, Montreal, Canada; Department of Pharmacology and Therapeutics, McGill University, Montreal, Canada.
| |
Collapse
|
5
|
Cieplińska K, Niedziela E, Kowalska A. Immunological Processes in the Orbit and Indications for Current and Potential Drug Targets. J Clin Med 2023; 13:72. [PMID: 38202079 PMCID: PMC10780108 DOI: 10.3390/jcm13010072] [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: 11/12/2023] [Revised: 12/17/2023] [Accepted: 12/20/2023] [Indexed: 01/12/2024] Open
Abstract
Thyroid eye disease (TED) is an extrathyroidal manifestation of Graves' disease (GD). Similar to GD, TED is caused by an autoimmune response. TED is an autoimmune inflammatory disorder of the orbit and periorbital tissues, characterized by upper eyelid retraction, swelling, redness, conjunctivitis, and bulging eyes. The pathophysiology of TED is complex, with the infiltration of activated T lymphocytes and activation of orbital fibroblasts (OFs) and autoantibodies against the common autoantigen of thyroid and orbital tissues. Better understanding of the multifactorial pathogenesis of TED contributes to the development of more effective therapies. In this review, we present current and potential drug targets. The ideal treatment should slow progression of the disease with as little interference with patient immunity as possible. In the future, TED treatment will target the immune mechanism involved in the disease and will be based on a strategy of restoring tolerance to autoantigens.
Collapse
Affiliation(s)
| | - Emilia Niedziela
- Collegium Medicum, Jan Kochanowski University in Kielce, 25-317 Kielce, Poland; (E.N.); (A.K.)
- Department of Endocrinology, Holy Cross Cancer Center, 25-734 Kielce, Poland
| | - Aldona Kowalska
- Collegium Medicum, Jan Kochanowski University in Kielce, 25-317 Kielce, Poland; (E.N.); (A.K.)
- Department of Endocrinology, Holy Cross Cancer Center, 25-734 Kielce, Poland
| |
Collapse
|
6
|
Cheng L, Hu J, Zhang L, Shen N, Chen H, Zhang F. Repurposing Lenvatinib as A Potential Therapeutic Agent against Thyroid Eye Disease by Suppressing Adipogenesis in Orbital Adipose Tissues. Pharmaceuticals (Basel) 2022; 15:1305. [PMID: 36355477 PMCID: PMC9696555 DOI: 10.3390/ph15111305] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 10/19/2022] [Accepted: 10/19/2022] [Indexed: 08/22/2023] Open
Abstract
Thyroid eye disease (TED) is the most common orbital disease in adults. Targeting expanded orbital adipose tissue (OAT) removed by surgery has therapeutic potential. However, drugs targeting OAT are unavailable because of the lack of deciphering features of OAT. Here, we aimed to investigate the mechanism underlying OAT expansion and identify a drug targeting OAT in TED. We found an increasing number of adipocytes with smaller size in TED-derived OATs as compared with controls, indicating that hyperplasia rather than hypertrophy contributed to OAT enlargement in TED. Typically smaller-sized adipocytes in TED patient-derived OATs were noted to localize surrounding vessels. RNA sequencing revealed enriched vascular endothelial growth factor receptor (VEGFR) genes in adipocytes differentiated from preadipocytes of TED-derived stromal vascular fraction (SVF). Similarly, OATs in patients with TED also expressed a higher level of VEGFR-1 and -2. We induced adipogenesis in TED-derived SVF with or without Lenvatinib, an FDA-approved small-molecule VEGFR inhibitor. Lenvatinib significantly suppressed lipid accumulation in a dose-dependent manner. In conclusion, our study revealed the potential anti-adipogenic effect of Lenvatinib on the OAT of TED-affected patients. In addition to proposing a drug for TED treatment, this study shows the therapeutic potential of anti-adipogenesis drugs targeting the VEGF pathway.
Collapse
Affiliation(s)
- Lu Cheng
- National Clinical Research Center for Eye Diseases, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
- Shanghai Key Laboratory of Fundus Diseases, Shanghai 200080, China
| | - Jing Hu
- National Clinical Research Center for Eye Diseases, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
- Shanghai Key Laboratory of Fundus Diseases, Shanghai 200080, China
| | - Ling Zhang
- National Clinical Research Center for Eye Diseases, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
- Shanghai Key Laboratory of Fundus Diseases, Shanghai 200080, China
| | - Ning Shen
- National Clinical Research Center for Eye Diseases, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
- Shanghai Key Laboratory of Fundus Diseases, Shanghai 200080, China
| | - Hui Chen
- National Clinical Research Center for Eye Diseases, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
- Shanghai Key Laboratory of Fundus Diseases, Shanghai 200080, China
- Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai 200080, China
- Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai 200080, China
| | - Fang Zhang
- National Clinical Research Center for Eye Diseases, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
- Shanghai Key Laboratory of Fundus Diseases, Shanghai 200080, China
- Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai 200080, China
- Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai 200080, China
| |
Collapse
|
7
|
Gupta V, Hammond CL, Roztocil E, Gonzalez MO, Feldon SE, Woeller CF. Thinking inside the box: Current insights into targeting orbital tissue remodeling and inflammation in thyroid eye disease. Surv Ophthalmol 2022; 67:858-874. [PMID: 34487739 PMCID: PMC8891393 DOI: 10.1016/j.survophthal.2021.08.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Revised: 08/24/2021] [Accepted: 08/30/2021] [Indexed: 12/21/2022]
Abstract
Thyroid eye disease (TED) is an autoimmune disorder that manifests in the orbit. In TED, the connective tissue behind the eye becomes inflamed and remodels with increased fat accumulation and/or increased muscle and scar tissue. As orbital tissue expands, patients develop edema, exophthalmos, diplopia, and optic neuropathy. In severe cases vision loss may occur secondary to corneal scarring from exposure or optic nerve compression. Currently there is no cure for TED, and treatments are limited. A major breakthrough in TED therapy occurred with the FDA approval of teprotumumab, a monoclonal insulin-like growth factor 1 receptor (IGF1R) blocking antibody. Yet, teprotumumab therapy has limitations, including cost, infusion method of drug delivery, variable response, and relapse. We describe approaches to target orbital fibroblasts and the complex pathophysiology that underlies tissue remodeling and inflammation driving TED. Further advances in the elucidation of the mechanisms of TED may lead to prophylaxis based upon early biomarkers as well as lead to more convenient, less expensive therapies.
Collapse
Affiliation(s)
- Vardaan Gupta
- Flaum Eye Institute, University of Rochester, 210 Crittenden Boulevard, Rochester, New York 14642, USA
| | - Christine L Hammond
- Flaum Eye Institute, University of Rochester, 210 Crittenden Boulevard, Rochester, New York 14642, USA
| | - Elisa Roztocil
- Flaum Eye Institute, University of Rochester, 210 Crittenden Boulevard, Rochester, New York 14642, USA
| | - Mithra O Gonzalez
- Flaum Eye Institute, University of Rochester, 210 Crittenden Boulevard, Rochester, New York 14642, USA
| | - Steven E Feldon
- Flaum Eye Institute, University of Rochester, 210 Crittenden Boulevard, Rochester, New York 14642, USA
| | - Collynn F Woeller
- Flaum Eye Institute, University of Rochester, 210 Crittenden Boulevard, Rochester, New York 14642, USA.
| |
Collapse
|
8
|
Teprotumumab in Thyroid-Associated Ophthalmopathy: Rationale for Therapeutic Insulin-Like Growth Factor-I Receptor Inhibition. J Neuroophthalmol 2021; 40:74-83. [PMID: 32040069 DOI: 10.1097/wno.0000000000000890] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Thyroid-associated ophthalmopathy (TAO) is an autoimmune component of Graves' disease for which no currently available medical therapy provides reliable and safe benefit. Based on insights generated experimentally over the past several decades, the insulin-like growth factor-I receptor (IGF-IR) has been implicated in the pathogenesis of TAO. Furthermore, an IGF-IR inhibitor, teprotumumab, has emerged from 2 clinical trials as a promising treatment for active, moderate to severe TAO. This brief review intends to provide an overview of the rationale underlying the development of teprotumumab for this disease. It is possible that teprotumumab will soon take its place in our therapeutic armamentarium for active TAO.
Collapse
|
9
|
Fernando R, Smith TJ. Slit2 Regulates Hyaluronan & Cytokine Synthesis in Fibrocytes: Potential Relevance to Thyroid-Associated Ophthalmopathy. J Clin Endocrinol Metab 2021; 106:e20-e33. [PMID: 32968816 PMCID: PMC7765649 DOI: 10.1210/clinem/dgaa684] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 09/18/2020] [Indexed: 02/06/2023]
Abstract
CONTEXT CD34+ fibrocytes have been implicated in development of thyroid-associated ophthalmopathy (TAO), a consequential autoimmune manifestation of Graves disease (GD). In TAO, CD34+ fibrocytes appear to masquerade as CD34+ orbital fibroblasts mixed with CD34- OF (collectively, GD-OF). Slit2, an axon guidance glycoprotein, is expressed by CD34- OF and attenuates GD-OF gene expression. Cardinal features of TAO include hyaluronan (HA) accumulation and cytokine-driven inflammation. OBJECTIVE Compare expression of HA synthase isoenzymes (HAS1-3), UDP-glucose dehydrogenase (UGDH), synthesis of HA, interleukin-6 (IL-6), and tumor necrosis factor-α (TNF-α) in fibrocytes and GD-OF. Determine whether Slit2 alters gene expression patterns. DESIGN/SETTING/PARTICIPANTS Patients with TAO and healthy donors were recruited from an academic practice. MAIN OUTCOME MEASURES Real-time polymerase chain reaction, HA, IL-6, and TNF-α immunoassays. RESULTS HA synthesis and release from fibrocytes is substantially lower than in GD-OF. HAS1 expression dominates in fibrocytes while HAS2 in GD-OF. In contrast, HAS2 and UGDH expression dominate GD-OF and localize to CD34- OF. Recombinant human Slit2 (rhSlit2) substantially upregulates HA synthesis and HAS2 expression in fibrocytes but attenuates IL-6 and TNF-α production in these cells. In contrast, knocking down Slit2 in GD-OF reduces HA synthesis and HAS2 and UGDH expression while upregulating IL-6 and TNF-α. CONCLUSION The dramatic differences in HA, IL-6, and TNF-α production, and HAS and UGDH expression found in fibrocytes and GD-OF appear, at least in part, to be attributable to Slit2. These findings provide novel insight into the differences in gene expression exhibited by CD34+ fibrocytes and CD34+ OF and therefore reveal important aspects of disease pathogenesis.
Collapse
Affiliation(s)
- Roshini Fernando
- Department of Ophthalmology and Visual Sciences, Kellogg Eye Center, University of Michigan Medical School, Ann Arbor, Michigan
- Division of Metabolism, Endocrinology and Diabetes, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan
| | - Terry J Smith
- Department of Ophthalmology and Visual Sciences, Kellogg Eye Center, University of Michigan Medical School, Ann Arbor, Michigan
- Division of Metabolism, Endocrinology and Diabetes, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan
| |
Collapse
|
10
|
Hirata Y, Kariya S, Kanai K, Fujiwara T, Makihara SI, Omichi R, Higaki T, Haruna T, Oka A, Nishizaki K, Okano M. Effect of Prostaglandin D 2 on mRNA Expression of Three Isoforms of Hyaluronic Acid Synthase in Nasal Polyp Fibroblasts. Am J Rhinol Allergy 2020; 35:44-51. [PMID: 32551924 DOI: 10.1177/1945892420932781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
BACKGROUND Hyaluronan is one of the major extracellular matrixes in chronic rhinosinusitis (CRS) associated with tissue remodeling. Prostaglandin D2 (PGD2) is also associated with the pathogenesis of CRS. However, little is known about whether PGD2 regulates hyaluronan production by human airway fibroblasts. OBJECTIVE We sought to determine the effect of PGD2 on the mRNA expression of three isoforms of membrane-bound hyaluronic acid synthase (HAS1, HAS2 and HAS3) in fibroblasts, the major source of hyaluronan production, derived from CRS patients. METHODS Nasal polyp-derived fibroblasts (NPDF) and uncinate tissue-derived fibroblasts (UTDF) were established from CRS patients with nasal polyps and those without, respectively. These fibroblasts were stimulated with PGD2 or PGD2 receptor (DP/CRTH2)-selective agonists in the presence or absence of receptor-selective antagonists. mRNA levels for HAS1, HAS2 and HAS3 were determined by real-time quantitative PCR. RESULTS PGD2 (1 µM) significantly enhanced HAS1 but not HAS2 or HAS3 mRNA expression by NPDF. Enhanced HAS1 mRNA expression was also obtained by stimulation with a DP receptor-selective agonist, but not with a CRTH2 receptor-selective agonist. In addition, PGD2-induced HAS1 mRNA expression was significantly inhibited by pre-treatment with DP receptor-selective antagonists. Similar induction of PGD2-induced HAS1 mRNA expression was seen in UTDF. CONCLUSION PGD2 selectively stimulates HAS1 mRNA expression in local fibroblasts in CRS via DP, but not CRTH2, receptors.
Collapse
Affiliation(s)
- Yuji Hirata
- Department of Otolaryngology-Head & Neck Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan.,Department of Otorhinolaryngology, Kagawa Prefectural Central Hospital, Takamatsu, Japan
| | - Shin Kariya
- Department of Otolaryngology-Head & Neck Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Kengo Kanai
- Department of Otolaryngology-Head & Neck Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan.,Department of Otorhinolaryngology, Kagawa Prefectural Central Hospital, Takamatsu, Japan
| | - Tazuko Fujiwara
- Department of Otolaryngology-Head & Neck Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | | | - Ryotaro Omichi
- Department of Otolaryngology-Head & Neck Surgery, The University of Iowa, Iowa City, Iowa
| | - Takaya Higaki
- Department of Otolaryngology-Head & Neck Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Takenori Haruna
- Department of Otorhinolaryngology, Himeji St. Mary's Hospital, Himeji, Japan
| | - Aiko Oka
- Department of Otorhinolaryngology, International University of Health and Welfare School of Medicine, Narita, Japan
| | - Kazunori Nishizaki
- Department of Otolaryngology-Head & Neck Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Mitsuhiro Okano
- Department of Otolaryngology-Head & Neck Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan.,Department of Otorhinolaryngology, International University of Health and Welfare School of Medicine, Narita, Japan
| |
Collapse
|
11
|
Roztocil E, Hammond CL, Gonzalez MO, Feldon SE, Woeller CF. The aryl hydrocarbon receptor pathway controls matrix metalloproteinase-1 and collagen levels in human orbital fibroblasts. Sci Rep 2020; 10:8477. [PMID: 32439897 PMCID: PMC7242326 DOI: 10.1038/s41598-020-65414-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Accepted: 05/04/2020] [Indexed: 12/13/2022] Open
Abstract
Thyroid eye disease (TED) affects 25–50% of patients with Graves’ Disease. In TED, collagen accumulation leads to an expansion of the extracellular matrix (ECM) which causes destructive tissue remodeling. The purpose of this study was to investigate the therapeutic potential of activating the aryl hydrocarbon receptor (AHR) to limit ECM accumulation in vitro. The ability of AHR to control expression of matrix metalloproteinase-1 (MMP1) was analyzed. MMP1 degrades collagen to prevent excessive ECM. Human orbital fibroblasts (OFs) were treated with the pro-scarring cytokine, transforming growth factor beta (TGFβ) to induce collagen production. The AHR ligand, 6-formylindolo[3,2b]carbazole (FICZ) was used to activate the AHR pathway in OFs. MMP1 protein and mRNA levels were analyzed by immunosorbent assay, Western blotting and quantitative PCR. MMP1 activity was detected using collagen zymography. AHR and its transcriptional binding partner, ARNT were depleted using siRNA to determine their role in activating expression of MMP1. FICZ induced MMP1 mRNA, protein expression and activity. MMP1 expression led to a reduction in collagen 1A1 levels. Furthermore, FICZ-induced MMP1 expression required both AHR and ARNT, demonstrating that the AHR-ARNT transcriptional complex is necessary for expression of MMP1 in OFs. These data show that activation of the AHR by FICZ increases MMP1 expression while leading to a decrease in collagen levels. Taken together, these studies suggest that AHR activation could be a promising target to block excessive collagen accumulation and destructive tissue remodeling that occurs in fibrotic diseases such as TED.
Collapse
Affiliation(s)
- Elisa Roztocil
- Flaum Eye Institute, University of Rochester, Rochester, New York, 14642, USA
| | - Christine L Hammond
- Flaum Eye Institute, University of Rochester, Rochester, New York, 14642, USA
| | - Mithra O Gonzalez
- Flaum Eye Institute, University of Rochester, Rochester, New York, 14642, USA
| | - Steven E Feldon
- Flaum Eye Institute, University of Rochester, Rochester, New York, 14642, USA
| | - Collynn F Woeller
- Flaum Eye Institute, University of Rochester, Rochester, New York, 14642, USA. .,Department of Environmental Medicine School of Medicine and Dentistry, University of Rochester, Rochester, New York, 14642, USA.
| |
Collapse
|
12
|
Immunological Aspects of Graves' Ophthalmopathy. BIOMED RESEARCH INTERNATIONAL 2019; 2019:7453260. [PMID: 31781640 PMCID: PMC6875285 DOI: 10.1155/2019/7453260] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Accepted: 08/30/2019] [Indexed: 12/13/2022]
Abstract
The body's autoimmune process is involved in the development of Graves' disease (GD), which is manifested by an overactive thyroid gland. In some patients, autoreactive inflammatory reactions contribute to the development of symptoms such as thyroid ophthalmopathy, and the subsequent signs and symptoms are derived from the expansion of orbital adipose tissue and edema of extraocular muscles within the orbit. The autoimmune process, production of antibodies against self-antigens such as TSH receptor (TSHR) and IGF-1 receptor (IGF-1R), inflammatory infiltration, and accumulation of glycosaminoglycans (GAG) lead to edematous-infiltrative changes in periocular tissues. As a consequence, edema exophthalmos develops. Orbital fibroblasts seem to play a crucial role in orbital inflammation, tissue expansion, remodeling, and fibrosis because of their proliferative activity as well as their capacity to differentiate into adipocytes and myofibroblasts and production of GAG. In this paper, based on the available medical literature, the immunological mechanism of GO pathogenesis has been summarized. Particular attention was paid to the role of orbital fibroblasts and putative autoantigens. A deeper understanding of the pathomechanism of the disease and the involvement of immunological processes may give rise to the introduction of new, effective, and safe methods of treatment or monitoring of the disease activity.
Collapse
|
13
|
Wang ZM, Wang ZY, Lu Y. The role of cell mediated immunopathogenesis in thyroid-associated ophthalmopathy. Int J Ophthalmol 2019; 12:1209-1214. [PMID: 31341815 DOI: 10.18240/ijo.2019.07.24] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Accepted: 05/21/2019] [Indexed: 12/31/2022] Open
Abstract
Currently, thyroid-associated ophthalmopathy (TAO) lacks effective treatment due to our lack of clarity in its immunopathogenesis. Orbital fibroblasts play a key role in altering inflammation and immune response in TAO, and are considered as the key target and effector cells in its pathogenesis. The orbit infiltrating CD34+ fibrocytes add on to the process by expressing high levels of autoantigens and inflammatory cytokines, while also differentiating into myofibroblasts or adipocytes. This review focuses on the role of orbital fibroblasts and CD34+ fibrocytes in the pathogenesis of TAO, highlighting the basis of emerging treatments.
Collapse
Affiliation(s)
- Zhen-Mao Wang
- Joint Shantou International Eye Center of Shantou University and the Chinese University of Hong Kong, Shantou 515000, Guangdong Province, China
| | - Zheng-Yan Wang
- The People's Hospital of Xintai, Xintai 271200, Shandong Province, China
| | - Yan Lu
- Department of Ophthalmology, Jinling Hospital, School of Medicine, Nanjing University, Nanjing 210002, Jiangsu Province, China
| |
Collapse
|
14
|
Carcinogenesis: the cancer cell–mast cell connection. Inflamm Res 2018; 68:103-116. [DOI: 10.1007/s00011-018-1201-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Revised: 11/14/2018] [Accepted: 11/14/2018] [Indexed: 12/20/2022] Open
|
15
|
Giménez‐Arnau A, Curto‐Barredo L, Nonell L, Puigdecanet E, Yelamos J, Gimeno R, Rüberg S, Santamaria‐Babi L, Pujol R. Transcriptome analysis of severely active chronic spontaneous urticaria shows an overall immunological skin involvement. Allergy 2017; 72:1778-1790. [PMID: 28407332 DOI: 10.1111/all.13183] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/10/2017] [Indexed: 02/02/2023]
Abstract
BACKGROUND The knowledge about chronic spontaneous urticaria (CSU) phenotypes is based on its clinical characteristics, associated comorbidities, course of the disease, and its response to the available effective drugs. Genotype expression and its further correlation with CSU phenotypes are still unknown. We describe the cutaneous transcriptome of patients suffering a severely active CSU refractory to antihistamine treatment. METHODS Through the bioinformatic analysis of the whole Human Genome with Oligo Microarrays and quantitative real-time polymerase chain reaction (qPCR), relevant genes expressed in nonlesional (NLS-CSU) and lesional skin (LS-CSU) and peripheral blood were identified in 20 patients suffering from severely active CSU and 10 healthy controls (HCs). RESULTS From 39 genes differentially expressed in NLS-CSU when compared with HCs, 31 (79.48%) were confirmed by qPCR corresponding to genes involved in epidermal homeostasis and dermal repair. From the analysis comparing LS-CSU with NLS-CSU, a selection of 142 genes was studied with qPCR, and 103 (72.53%) were confirmed. Differentially expressed genes in the phenomenon of wheal development are involved in a variety of biological functions as, epidermal differentiation, intracellular signal function, transcriptional factors cell cycle differentiation, inflammation, or coagulation. Differentially expressed genes that uniformly increase or decrease along the skin worsening until the wheal appearance is shown. CONCLUSION The skin of CSU patients with a severely active disease shows an overall immunological skin involvement showing a peculiar gene profile.
Collapse
Affiliation(s)
- A. Giménez‐Arnau
- Department of Dermatology Hospital del Mar IMIM Universitat Autònoma de Barcelona Barcelona Spain
| | - L. Curto‐Barredo
- Department of Dermatology Hospital del Mar IMIM Universitat Autònoma de Barcelona Barcelona Spain
| | - L. Nonell
- Microarray Analysis Service Institut Hospital del Mar d'Investigacions Mèdiques (IMIM) Barcelona Spain
| | - E. Puigdecanet
- Microarray Analysis Service Institut Hospital del Mar d'Investigacions Mèdiques (IMIM) Barcelona Spain
| | - J. Yelamos
- Immunology Department Institut Hospital del Mar d'Investigacions Mèdiques (IMIM) Barcelona Spain
| | - R. Gimeno
- Immunology Department Institut Hospital del Mar d'Investigacions Mèdiques (IMIM) Barcelona Spain
| | - S. Rüberg
- Microarray Analysis Service Milteny Biotec GmbH Teterow Germany
| | - L. Santamaria‐Babi
- Translational Immunology, Department of Physiology and Immunology Universitat de Barcelona Barcelona Spain
| | - R.M. Pujol
- Department of Dermatology Hospital del Mar IMIM Universitat Autònoma de Barcelona Barcelona Spain
| |
Collapse
|
16
|
Xia N, Ye X, Hu X, Song S, Xu H, Niu M, Wang H, Wang J. Simultaneous induction of Graves' hyperthyroidism and Graves' ophthalmopathy by TSHR genetic immunization in BALB/c mice. PLoS One 2017; 12:e0174260. [PMID: 28319174 PMCID: PMC5358867 DOI: 10.1371/journal.pone.0174260] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2016] [Accepted: 03/06/2017] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Graves' disease is the most common form of autoimmune thyroid disorder, characterized by hyperthyroidism due to circulating autoantibodies. To address the pathological features and establish a therapeutic approach of this disease, an animal model carrying the phenotype of Graves' disease (GD) in concert with Graves' Ophthalmopathy (GO) will be very important. However, there are no ideal animal models that are currently available. The aim of the present study is to establish an animal model of GD and GO disease, and its pathological features were further characterized. METHODS A recombinant plasmid pcDNA3.1- T289 was constructed by inserting the TSHR A-subunit gene into the expression vector pcDNA3.1, and genetic immunization was successfully performed by intramuscular injection of the plasmid pcDNA3.1-T289 on female 8-week-old BALB/c mice. Each injection was immediately followed by in vivo electroporation using ECM830 square wave electroporator. Morphological changes of the eyes were examined using 7.0T MRI scanner. Levels of serum T4 and TSHR antibodies (TRAb) were assessed by ELISA. The pathological changes of the thyroid and orbital tissues were examined by histological staining such as H&E staining and Alcian blue staining. RESULTS More than 90% of the immunized mice spontaneously developed goiter, and about 80% of the immunized mice manifested increased serum T4 and TRAb levels, combined with hypertrophy and hyperplasia of thyroid follicles. A significantly increased synthesis of hyaluronic acid was detected in in the immunized mice compared with the control groups. CONCLUSION We have successfully established an animal model manifesting Graves' hyperthyroidism and ophthalmopathy, which provides a useful tool for future study of the pathological features and the development of novel therapies of the diseases.
Collapse
Affiliation(s)
- Nan Xia
- Department of Endocrinology, Jingling Hospital, Medical School of Nanjing University, Nanjing, P.R. China
- Center for Translational Medicine and Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, P.R. China
- Department of Endocrinology, Mingci Cardiovascular Hospital, Wuxi, P.R. China
| | - Xiaozhen Ye
- Department of Endocrinology, Jingling Hospital, Medical School of Nanjing University, Nanjing, P.R. China
| | - Xiaohao Hu
- Department of Endocrinology, Jingling Hospital, Medical School of Nanjing University, Nanjing, P.R. China
| | - Shiyu Song
- Center for Translational Medicine and Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, P.R. China
| | - Hui Xu
- Center for Translational Medicine and Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, P.R. China
| | - Mengyuan Niu
- Center for Translational Medicine and Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, P.R. China
| | - Hongwei Wang
- Center for Translational Medicine and Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, P.R. China
| | - Jian Wang
- Department of Endocrinology, Jingling Hospital, Medical School of Nanjing University, Nanjing, P.R. China
| |
Collapse
|
17
|
Fusco M, Skaper SD, Coaccioli S, Varrassi G, Paladini A. Degenerative Joint Diseases and Neuroinflammation. Pain Pract 2017; 17:522-532. [DOI: 10.1111/papr.12551] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Revised: 12/17/2016] [Accepted: 12/17/2016] [Indexed: 12/14/2022]
Affiliation(s)
- Mariella Fusco
- Scientific Information and Documentation Center; Epitech Group; Padua Italy
| | - Stephen D. Skaper
- Department of Pharmaceutical and Pharmacological Sciences; University of Padua; Padua Italy
| | - Stefano Coaccioli
- Department of Internal Medicine and Rheumatology; Santa Maria Hospital; University of Perugia; Terni Italy
| | - Giustino Varrassi
- Department of Anesthesiology and Pain Medicine; School of Dentistry; LUdeS University; La Valletta Malta
- Paolo Procacci Foundation and European League Against Pain; Rome Italy
| | | |
Collapse
|
18
|
Di Paola R, Fusco R, Impellizzeri D, Cordaro M, Britti D, Morittu VM, Evangelista M, Cuzzocrea S. Adelmidrol, in combination with hyaluronic acid, displays increased anti-inflammatory and analgesic effects against monosodium iodoacetate-induced osteoarthritis in rats. Arthritis Res Ther 2016; 18:291. [PMID: 27955699 PMCID: PMC5153857 DOI: 10.1186/s13075-016-1189-5] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2016] [Accepted: 11/21/2016] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Osteoarthritis (OA) is a degenerative joint disease produced by a cascade of events that can ultimately lead to joint damage. The aim of this study was to evaluate the effect of adelmidrol, a synthetic palmitoylethanolamide analogue, combined with hyaluronic acid on pain severity and modulation of the inflammatory response in a rat model of monosodium iodoacetate (MIA)-induced osteoarthritis. METHODS OA was induced by intra-articular injection of MIA in the knee joint. On day 21 post-MIA administration, the knee joint was analyzed. Rats subjected to OA were treated by intra-articular injection of adelmidrol in combination with sodium hyaluronate at different doses and time points after MIA induction. Limb nociception was assessed by the paw withdrawal latency and threshold measurement. Samples were examined macroscopically, histologically, and by immunohistochemistry. RESULTS At day 21 post-MIA injection, the MIA + solvent and MIA + 1.0% sodium hyaluronate groups showed irregularities and fibrillation in the surface layer, a decrease in blood cells and multilayering in transition and radial zones, no pannus formation, and modified Mankin scores significantly higher than sham knees. The combination of hyaluronic acid and adelmidrol dose-dependently (adelmidrol 0.6% + 1.0% sodium hyaluronate and adelmidrol 2% + 1.0% sodium hyaluronate) reduced the histological alterations induced by MIA. Moreover, degeneration of articular cartilage, mast cell infiltration, and pro-inflammatory cytokine and chemokine plasma levels were significantly downregulated by treatment with a combination of hyaluronic acid and adelmidrol at the above doses. CONCLUSIONS Our results clearly demonstrate that the combination of hyaluronic acid and adelmidrol improves the signs of OA induced by MIA.
Collapse
Affiliation(s)
- Rosanna Di Paola
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D'Alcontres, n 31, Messina, 98166, Italy
| | - Roberta Fusco
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D'Alcontres, n 31, Messina, 98166, Italy
| | - Daniela Impellizzeri
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D'Alcontres, n 31, Messina, 98166, Italy
| | - Marika Cordaro
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D'Alcontres, n 31, Messina, 98166, Italy
| | - Domenico Britti
- Department of Health Science, University of Catanzaro, Viale Europa, Campus S. Venuta, Germaneto, Catanzaro, 88100, Italy
| | - Valeria Maria Morittu
- Department of Health Science, University of Catanzaro, Viale Europa, Campus S. Venuta, Germaneto, Catanzaro, 88100, Italy
| | - Maurizio Evangelista
- Institute of Anaesthesiology and Reanimation, Catholic University of the Sacred Heart, Rome, Italy
| | - Salvatore Cuzzocrea
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D'Alcontres, n 31, Messina, 98166, Italy. .,Department of Pharmacological and Physiological Science, Saint Louis University School of Medicine, 1402 South Grand Blvd, St. Louis, MO, 63104, USA.
| |
Collapse
|
19
|
Krieger CC, Place RF, Bevilacqua C, Marcus-Samuels B, Abel BS, Skarulis MC, Kahaly GJ, Neumann S, Gershengorn MC. TSH/IGF-1 Receptor Cross Talk in Graves' Ophthalmopathy Pathogenesis. J Clin Endocrinol Metab 2016; 101:2340-7. [PMID: 27043163 PMCID: PMC4891793 DOI: 10.1210/jc.2016-1315] [Citation(s) in RCA: 95] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
CONTEXT The TSH receptor (TSHR) is considered the main target of stimulatory autoantibodies in the pathogenesis of Graves' ophthalmopathy (GO); however, it has been suggested that stimulatory IGF-1 receptor (IGF-1R) autoantibodies also play a role. OBJECTIVE We previously demonstrated that a monoclonal stimulatory TSHR antibody, M22, activates TSHR/IGF-1R cross talk in orbital fibroblasts/preadipocytes obtained from patients with GO (GO fibroblasts [GOFs]). We show that cross talk between TSHR and IGF-1R, not direct IGF-1R activation, is involved in the mediation of GO pathogenesis stimulated by Graves' autoantibodies. DESIGN/SETTING/PARTICIPANTS Immunoglobulins were purified from the sera of 57 GO patients (GO-Igs) and tested for their ability to activate TSHR and/or IGF-1R directly and TSHR/IGF-1R cross talk in primary cultures of GOFs. Cells were treated with M22 or GO-Igs with or without IGF-1R inhibitory antibodies or linsitinib, an IGF-1R kinase inhibitor. MAIN OUTCOME MEASURES Hyaluronan (hyaluronic acid [HA]) secretion was measured as a major biological response for GOF stimulation. IGF-1R autophosphorylation was used as a measure of direct IGF-1R activation. TSHR activation was determined through cAMP production. RESULTS A total of 42 out of 57 GO-Ig samples stimulated HA secretion. None of the GO-Ig samples exhibited evidence for IGF-1R autophosphorylation. Both anti-IGF-1R antibodies completely inhibited IGF-1 stimulation of HA secretion. By contrast, only 1 IGF-1R antibody partially blocked HA secretion stimulated by M22 or GO-Igs in a manner similar to linsitinib, whereas the other IGF-1R antibody had no effect on M22 or GO-Ig stimulation. These findings show that the IGF-1R is involved in GO-Igs stimulation of HA secretion without direct activation of IGF-1R. CONCLUSIONS IGF-1R activation by GO-Igs occurs via TSHR/IGF-1R cross talk rather than direct binding to IGF-1R, and this cross talk is important in the pathogenesis of GO.
Collapse
Affiliation(s)
- Christine C Krieger
- Laboratory of Endocrinology and Receptor Biology (C.C.K., R.F.P., C.B., B.M.-S., S.N., M.C.G.), National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland 20892; Nova Therapeutics LLC (R.F.P.), Pasadena, California; Diabetes, Endocrinology, and Obesity Branch (B.S.A., M.C.S.), National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland 20892; and Johannes Gutenberg University Medical Center (G.J.K.), Mainz, Germany
| | - Robert F Place
- Laboratory of Endocrinology and Receptor Biology (C.C.K., R.F.P., C.B., B.M.-S., S.N., M.C.G.), National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland 20892; Nova Therapeutics LLC (R.F.P.), Pasadena, California; Diabetes, Endocrinology, and Obesity Branch (B.S.A., M.C.S.), National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland 20892; and Johannes Gutenberg University Medical Center (G.J.K.), Mainz, Germany
| | - Carmine Bevilacqua
- Laboratory of Endocrinology and Receptor Biology (C.C.K., R.F.P., C.B., B.M.-S., S.N., M.C.G.), National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland 20892; Nova Therapeutics LLC (R.F.P.), Pasadena, California; Diabetes, Endocrinology, and Obesity Branch (B.S.A., M.C.S.), National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland 20892; and Johannes Gutenberg University Medical Center (G.J.K.), Mainz, Germany
| | - Bernice Marcus-Samuels
- Laboratory of Endocrinology and Receptor Biology (C.C.K., R.F.P., C.B., B.M.-S., S.N., M.C.G.), National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland 20892; Nova Therapeutics LLC (R.F.P.), Pasadena, California; Diabetes, Endocrinology, and Obesity Branch (B.S.A., M.C.S.), National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland 20892; and Johannes Gutenberg University Medical Center (G.J.K.), Mainz, Germany
| | - Brent S Abel
- Laboratory of Endocrinology and Receptor Biology (C.C.K., R.F.P., C.B., B.M.-S., S.N., M.C.G.), National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland 20892; Nova Therapeutics LLC (R.F.P.), Pasadena, California; Diabetes, Endocrinology, and Obesity Branch (B.S.A., M.C.S.), National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland 20892; and Johannes Gutenberg University Medical Center (G.J.K.), Mainz, Germany
| | - Monica C Skarulis
- Laboratory of Endocrinology and Receptor Biology (C.C.K., R.F.P., C.B., B.M.-S., S.N., M.C.G.), National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland 20892; Nova Therapeutics LLC (R.F.P.), Pasadena, California; Diabetes, Endocrinology, and Obesity Branch (B.S.A., M.C.S.), National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland 20892; and Johannes Gutenberg University Medical Center (G.J.K.), Mainz, Germany
| | - George J Kahaly
- Laboratory of Endocrinology and Receptor Biology (C.C.K., R.F.P., C.B., B.M.-S., S.N., M.C.G.), National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland 20892; Nova Therapeutics LLC (R.F.P.), Pasadena, California; Diabetes, Endocrinology, and Obesity Branch (B.S.A., M.C.S.), National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland 20892; and Johannes Gutenberg University Medical Center (G.J.K.), Mainz, Germany
| | - Susanne Neumann
- Laboratory of Endocrinology and Receptor Biology (C.C.K., R.F.P., C.B., B.M.-S., S.N., M.C.G.), National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland 20892; Nova Therapeutics LLC (R.F.P.), Pasadena, California; Diabetes, Endocrinology, and Obesity Branch (B.S.A., M.C.S.), National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland 20892; and Johannes Gutenberg University Medical Center (G.J.K.), Mainz, Germany
| | - Marvin C Gershengorn
- Laboratory of Endocrinology and Receptor Biology (C.C.K., R.F.P., C.B., B.M.-S., S.N., M.C.G.), National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland 20892; Nova Therapeutics LLC (R.F.P.), Pasadena, California; Diabetes, Endocrinology, and Obesity Branch (B.S.A., M.C.S.), National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland 20892; and Johannes Gutenberg University Medical Center (G.J.K.), Mainz, Germany
| |
Collapse
|
20
|
Virakul S, Phetsuksiri T, van Holten-Neelen C, Schrijver B, van Steensel L, Dalm VASH, Paridaens D, van den Bosch WA, van Hagen PM, Dik WA. Histamine induces NF-κB controlled cytokine secretion by orbital fibroblasts via histamine receptor type-1. Exp Eye Res 2016; 147:85-93. [PMID: 27170049 DOI: 10.1016/j.exer.2016.05.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Revised: 04/21/2016] [Accepted: 05/04/2016] [Indexed: 12/19/2022]
Abstract
Mast cells and their products are likely to be involved in regulating orbital fibroblast activity in Graves' Ophthalmopathy (GO). Histamine is abundantly present in granules of mast cells and is released upon mast cell activation. However, the effect of histamine on orbital fibroblasts has not been examined so far. Orbital tissues from GO patients and controls were analyzed for the presence of mast cells using toluidine blue staining and immunohistochemical detection of CD117 (stem cell factor receptor). Orbital fibroblasts were cultured from GO patients and healthy controls, stimulated with histamine and cytokines (IL-6, IL-8, CCL2, CCL5, CCL7, CXCL10 and CXCL11) were measured in culture supernatants. Also hyaluronan levels were measured in culture supernatants and hyaluronan synthase (HAS) and hyaluronidase (HYAL) gene expression levels were determined. In addition, histamine receptor subtype gene expression levels were examined as well as the effect of the histamine receptor-1 (HRH1) antagonist loratadine and NF-κB inhibitor SC-514 on histamine-induced cytokine production. Mast cell numbers were increased in GO orbital tissues. Histamine stimulated the production of IL-6, IL-8 and CCL2 by orbital fibroblasts, while it had no effect on the production of CCL5, CCL7, CXCL10, CXCL11 and hyaluronan. Orbital fibroblasts expressed HRH1 and loratadine and SC-514 both blocked histamine-induced IL-6, IL-8 and CCL2 production by orbital fibroblasts. In conclusion, this study demonstrates that histamine can induce the production of NF-κB controlled-cytokines by orbital fibroblasts, which supports a role for mast cells in GO.
Collapse
Affiliation(s)
- Sita Virakul
- Department of Immunology, Laboratory Medical Immunology, Erasmus MC, Rotterdam, The Netherlands; Internal Medicine, Division of Clinical Immunology, Erasmus MC, Rotterdam, The Netherlands
| | - Tanachaporn Phetsuksiri
- Department of Immunology, Laboratory Medical Immunology, Erasmus MC, Rotterdam, The Netherlands
| | - Conny van Holten-Neelen
- Department of Immunology, Laboratory Medical Immunology, Erasmus MC, Rotterdam, The Netherlands
| | - Benjamin Schrijver
- Department of Immunology, Laboratory Medical Immunology, Erasmus MC, Rotterdam, The Netherlands
| | - Leendert van Steensel
- Department of Immunology, Laboratory Medical Immunology, Erasmus MC, Rotterdam, The Netherlands
| | - Virgil A S H Dalm
- Department of Immunology, Laboratory Medical Immunology, Erasmus MC, Rotterdam, The Netherlands; Internal Medicine, Division of Clinical Immunology, Erasmus MC, Rotterdam, The Netherlands
| | | | | | - P Martin van Hagen
- Department of Immunology, Laboratory Medical Immunology, Erasmus MC, Rotterdam, The Netherlands; Internal Medicine, Division of Clinical Immunology, Erasmus MC, Rotterdam, The Netherlands; Rotterdam Eye Hospital, Rotterdam, The Netherlands
| | - Willem A Dik
- Department of Immunology, Laboratory Medical Immunology, Erasmus MC, Rotterdam, The Netherlands.
| |
Collapse
|
21
|
Dik WA, Virakul S, van Steensel L. Current perspectives on the role of orbital fibroblasts in the pathogenesis of Graves' ophthalmopathy. Exp Eye Res 2016; 142:83-91. [PMID: 26675405 DOI: 10.1016/j.exer.2015.02.007] [Citation(s) in RCA: 114] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2014] [Revised: 02/05/2015] [Accepted: 02/10/2015] [Indexed: 02/06/2023]
Abstract
Graves' ophthalmopathy (GO) is an extra-thyroidal complication of Graves' disease (GD; Graves' hyperthyroidism) characterized by orbital tissue inflammation, expansion, remodeling and fibrosis. Although the initiating trigger of GO is still indistinct, excessive orbital fibroblast activity is at the heart of its pathogenesis. Orbital fibroblasts are activated by cellular interactions with immune cells and the soluble factors they secrete. Orbital fibroblasts, especially from GO patients, express the thyrotropin receptor (TSH-receptor; TSHR), and activation of the orbital fibroblast population by stimulatory autoantibodies directed against the TSHR may provide an important link between GD and GO. Furthermore, stimulatory autoantibodies directed against the insulin-like growth factor-1 receptor have been proposed to contribute to orbital fibroblast activation in GO. Activated orbital fibroblasts produce inflammatory mediators thereby contributing to the orbital inflammatory process in GO. Moreover, orbital fibroblasts exhibit robust proliferative activity and extracellular matrix (especially hyaluronan) synthesizing capacity and can differentiate into adipocytes and myofibroblasts with disease progression, thereby contributing to tissue expansion/remodeling and fibrosis in GO. Orbital fibroblasts, especially those from GO patients, exhibit a hyper-responsive phenotype when compared to fibroblasts from other anatomical regions, which may further contribute to GO pathogenesis. Fibrocytes have been identified as additional source of orbital fibroblasts in GO, where they may contribute to orbital tissue inflammation, adipogenesis and remodeling/fibrosis. This review addresses our current view on the role that orbital fibroblasts fulfill in GO pathogenesis and both established as well as less established not fully crystallized concepts that need future studies will be discussed.
Collapse
Affiliation(s)
- Willem A Dik
- Department of Immunology, Laboratory Medical Immunology, Erasmus MC, University Medical Center Rotterdam, The Netherlands.
| | - Sita Virakul
- Department of Immunology, Laboratory Medical Immunology, Erasmus MC, University Medical Center Rotterdam, The Netherlands
| | - Leendert van Steensel
- Department of Immunology, Laboratory Medical Immunology, Erasmus MC, University Medical Center Rotterdam, The Netherlands
| |
Collapse
|
22
|
Abstract
Thyroid-associated ophthalmopathy (TAO) is a vexing and undertreated ocular component of Graves disease in which orbital tissues undergo extensive remodelling. My colleagues and I have introduced the concept that fibrocytes expressing the haematopoietic cell antigen CD34 (CD34(+) fibrocytes), which are precursor cells of bone-marrow-derived monocyte lineage, express the TSH receptor (TSHR). These cells also produce several other proteins whose expression was traditionally thought to be restricted to the thyroid gland. TSHR-expressing fibrocytes in which the receptor is activated by its ligand generate extremely high levels of several inflammatory cytokines. Acting in concert with TSHR, the insulin-like growth factor 1 receptor (IGF-1R) expressed by orbital fibroblasts and fibrocytes seems to be necessary for TSHR-dependent cytokine production, as anti-IGF-1R blocking antibodies attenuate these proinflammatory actions of TSH. Furthermore, circulating fibrocytes are highly abundant in patients with TAO and seem to infiltrate orbital connective tissues, where they might transition to CD34(+) fibroblasts. My research group has postulated that the infiltration of fibrocytes into the orbit, their unique biosynthetic repertoire and their proinflammatory and profibrotic phenotype account for the characteristic properties exhibited by orbital connective tissues that underlie susceptibility to TAO. These insights, which have emerged in the past few years, might be of use in therapeutically targeting pathogenic orbit-infiltrating fibrocytes selectively by utilizing novel biologic agents that interfere with TSHR and IGF-1R signalling.
Collapse
Affiliation(s)
- Terry J Smith
- Department of Ophthalmology and Visual Sciences, Room 7112, Brehm Tower, Kellogg Eye Center, University of Michigan Medical School, 1000 Wall Street, Ann Arbor, MI 48105, USA
| |
Collapse
|
23
|
Siiskonen H, Oikari S, Pasonen-Seppänen S, Rilla K. Hyaluronan synthase 1: a mysterious enzyme with unexpected functions. Front Immunol 2015; 6:43. [PMID: 25699059 PMCID: PMC4318391 DOI: 10.3389/fimmu.2015.00043] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2014] [Accepted: 01/22/2015] [Indexed: 11/13/2022] Open
Abstract
Hyaluronan synthase 1 (HAS1) is one of three isoenzymes responsible for cellular hyaluronan synthesis. Interest in HAS1 has been limited because its role in hyaluronan production seems to be insignificant compared to the two other isoenzymes, HAS2 and HAS3, which have higher enzymatic activity. Furthermore, in most cell types studied so far, the expression of its gene is low and the enzyme requires high concentrations of sugar precursors for hyaluronan synthesis, even when overexpressed in cell cultures. Both expression and activity of HAS1 are induced by pro-inflammatory factors like interleukins and cytokines, suggesting its involvement in inflammatory conditions. Has1 is upregulated in states associated with inflammation, like atherosclerosis, osteoarthritis, and infectious lung disease. In addition, both full length and splice variants of HAS1 are expressed in malignancies like bladder and prostate cancers, multiple myeloma, and malignant mesothelioma. Interestingly, immunostainings of tissue sections have demonstrated the role of HAS1 as a poor predictor in breast cancer, and is correlated with high relapse rate and short overall survival. Utilization of fluorescently tagged proteins has revealed the intracellular distribution pattern of HAS1, distinct from other isoenzymes. In all cell types studied so far, a high proportion of HAS1 is accumulated intracellularly, with a faint signal detected on the plasma membrane and its protrusions. Furthermore, the pericellular hyaluronan coat produced by HAS1 is usually thin without induction by inflammatory agents or glycemic stress and depends on CD44–HA interactions. These specific interactions regulate the organization of hyaluronan into a leukocyte recruiting matrix during inflammatory responses. Despite the apparently minor enzymatic activity of HAS1 under normal conditions, it may be an important factor under conditions associated with glycemic stress like metabolic syndrome, inflammation, and cancer.
Collapse
Affiliation(s)
- Hanna Siiskonen
- Department of Dermatology, Kuopio University Hospital, University of Eastern Finland , Kuopio , Finland
| | - Sanna Oikari
- Institute of Biomedicine, University of Eastern Finland , Kuopio , Finland
| | | | - Kirsi Rilla
- Institute of Biomedicine, University of Eastern Finland , Kuopio , Finland
| |
Collapse
|
24
|
Wang Y, Smith TJ. Current concepts in the molecular pathogenesis of thyroid-associated ophthalmopathy. Invest Ophthalmol Vis Sci 2014; 55:1735-48. [PMID: 24651704 DOI: 10.1167/iovs.14-14002] [Citation(s) in RCA: 157] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Graves' disease (GD) is a common autoimmune condition. At its core, stimulatory autoantibodies are directed at the thyroid-stimulating hormone receptor (TSHR), resulting in dysregulated thyroid gland activity and growth. Closely associated with GD is the ocular condition known as thyroid-associated ophthalmopathy (TAO). The pathogenesis of TAO remains enigmatic as do the connections between the thyroid and orbit. This review highlights the putative molecular mechanisms involved in TAO and suggests how these insights provide future directions for identifying therapeutic targets. Genetic, epigenetic, and environmental factors have been suggested as contributory to the development of GD and TAO. Thyroid-stimulating hormone receptor and insulin-like growth factor receptor (IGF-1R) are expressed at higher levels in the orbital connective tissue from individuals with TAO than in healthy tissues. Together, they form a functional complex and appear to promote signaling relevant to GD and TAO. Orbital fibroblasts display an array of cell surface receptors and generate a host of inflammatory molecules that may participate in T and B cell infiltration. Recently, a population of orbital fibroblasts has been putatively traced to bone marrow-derived progenitor cells, known as fibrocytes, as they express CD45, CD34, CXCR4, collagen I, functional TSHR, and thyroglobulin (Tg). Fibrocytes become more numerous in GD and we believe traffic to the orbit in TAO. Numerous attempts at developing complete animal models of GD have been largely unsuccessful, because they lack fidelity with the ocular manifestations seen in TAO. Better understanding of the pathogenesis of TAO and development of improved animal models should greatly accelerate the identification of medical therapy for this vexing medical problem.
Collapse
Affiliation(s)
- Yao Wang
- Department of Ophthalmology and Visual Sciences and Division of Metabolic and Endocrine Disease, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan
| | | |
Collapse
|
25
|
Kuriyan AE, Woeller CF, O'Loughlin CW, Phipps RP, Feldon SE. Orbital fibroblasts from thyroid eye disease patients differ in proliferative and adipogenic responses depending on disease subtype. Invest Ophthalmol Vis Sci 2013; 54:7370-7. [PMID: 24135759 DOI: 10.1167/iovs.13-12741] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
PURPOSE Thyroid eye disease (TED) patients are classified as type I (predominantly fat compartment enlargement) or type II (predominantly extraocular muscle enlargement) based on orbital imaging. Orbital fibroblasts (OFs) can be driven to proliferate or differentiate into adipocytes in vitro. We tested the hypothesis that type I OFs undergo more adipogenesis than type II OFs, whereas type II OFs proliferate more than type I OFs. We also examined the effect of cyclooxygenase (COX) inhibitors on OF adipogenesis and proliferation. METHODS Type I, type II, and non-TED OFs were treated with transforming growth factor-beta (TGFβ) to induce proliferation and with 15-deoxy-Δ(-12,14)-prostaglandin J2 (15d-PGJ2) to induce adipogenesis. Proliferation was measured using the [(3)H]thymidine assay, and adipogenesis was measured using the AdipoRed assay, Oil Red O staining, and flow cytometry. The effect of COX inhibition on adipogenesis and proliferation was also studied. RESULTS Type II OFs incorporated 1.7-fold more [(3)H]thymidine than type I OFs (P < 0.05). Type I OFs accumulated 4.8-fold more lipid than type II OFs (P < 0.05) and 12.6-fold more lipid than non-TED OFs (P < 0.05). Oil Red O staining and flow cytometry also demonstrated increased adipogenesis in type I OFs compared to type II and non-TED OFs. Cyclooxygenase inhibition significantly decreased proliferation and adipogenesis in type II OFs, but not type I OFs. CONCLUSIONS We have demonstrated that OFs from TED patients have heterogeneous responses to proproliferative and proadipogenic stimulators in vitro in a manner that corresponds to their different clinical manifestations. Furthermore, we demonstrated a differential effect of COX inhibitors on type I and type II OF proliferation and adipogenesis.
Collapse
Affiliation(s)
- Ajay E Kuriyan
- Flaum Eye Institute, University of Rochester School of Medicine and Dentistry, Rochester, New York
| | | | | | | | | |
Collapse
|
26
|
Neumann S, Pope A, Geras-Raaka E, Raaka BM, Bahn RS, Gershengorn MC. A drug-like antagonist inhibits thyrotropin receptor-mediated stimulation of cAMP production in Graves' orbital fibroblasts. Thyroid 2012; 22:839-43. [PMID: 22784331 PMCID: PMC3407388 DOI: 10.1089/thy.2011.0520] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
BACKGROUND Fibroblasts (FIBs) within the retro-orbital space of patients with Graves' disease (GOFs) express thyrotropin receptors (TSHRs) and are thought to be an orbital target of TSHR-stimulating autoantibodies in Graves' ophthalmopathy (GO). Recently, we developed a low molecular weight, drug-like TSHR antagonist (NCGC00229600) that inhibited TSHR activation in a model cell system overexpressing TSHRs and in normal human thyrocytes expressing endogenous TSHRs. Herein, we test the hypothesis that NCGC00229600 will inhibit activation of TSHRs endogenously expressed in GOFs. METHODS Three strains of GOFs, previously obtained from patients with GO, were studied as undifferentiated FIBs and after differentiation into adipocytes (ADIPs), and another seven strains were studied only as FIBs. ADIP differentiation was monitored by morphology and measurement of adiponectin mRNA. FIBs and ADIPs were treated with the TSH- or TSHR-stimulating antibody M22 in the absence or presence of NCGC00229600 and TSHR activation was monitored by cAMP production. RESULTS FIBs contained few if any lipid vesicles and undetectable levels of adiponectin mRNA, whereas ADIPs exhibited abundant lipid vesicles and levels of adiponectin mRNA more than 250,000 times greater than FIBs; TSHR mRNA levels were 10-fold higher in ADIPs than FIBs. FIBs exhibited higher absolute levels of basal and forskolin-stimulated cAMP production than ADIPs. Consistent with previous findings, TSH stimulated cAMP production in the majority of ADIP strains and less consistently in FIBs. Most importantly, NCGC00229600 reduced both TSH- and M22-stimulated cAMP production in GOFs. CONCLUSIONS These data confirm previous findings that TSHR activation may cause increased cAMP production in GOFs and show that NCGC00229600 can inhibit TSHR activation in GOFs. These findings suggest that drug-like TSHR antagonists may have a role in treatment of GO.
Collapse
Affiliation(s)
- Susanne Neumann
- Laboratory of Endocrinology and Receptor Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland
| | - Arthur Pope
- Laboratory of Endocrinology and Receptor Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland
| | - Elizabeth Geras-Raaka
- Laboratory of Endocrinology and Receptor Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland
| | - Bruce M. Raaka
- Laboratory of Endocrinology and Receptor Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland
| | - Rebecca S. Bahn
- Division of Endocrinology, Diabetes, Metabolism, and Nutrition, Mayo Clinic, Rochester, Minnesota
| | - Marvin C. Gershengorn
- Laboratory of Endocrinology and Receptor Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
27
|
van Steensel L, Paridaens D, van Meurs M, van Hagen PM, van den Bosch WA, Kuijpers RWAM, Drexhage HA, Hooijkaas H, Dik WA. Orbit-infiltrating mast cells, monocytes, and macrophages produce PDGF isoforms that orchestrate orbital fibroblast activation in Graves' ophthalmopathy. J Clin Endocrinol Metab 2012; 97:E400-8. [PMID: 22238384 DOI: 10.1210/jc.2011-2697] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
PURPOSE Platelet-derived growth factors (PDGF) are regulators of fibroblast activity that may be involved in the pathophysiology of Graves' ophthalmopathy (GO). We unraveled the expression and origin of PDGF family members in GO orbital tissue and investigated the effect of PDGF isoforms on IL-6 and hyaluronan production and proliferation by orbital fibroblasts. METHODS PDGF-A, PDGF-B, PDGF-C, PDGF-D, PDGF-Rα, and PDGF-Rβ expression was determined by real-time quantitative PCR and PDGF-A and PDGF-B protein expression was determined by Western blot in orbital tissues. Orbital tissues were immunohistochemically stained for PDGF-A and PDGF-B expression, together with stainings for T cells, monocytes, B cells, macrophages, and mast cells. Effects of PDGF-AA, PDGF-AB, and PDGF-BB on orbital fibroblast proliferation and IL-6 and hyaluronan production were examined. Finally, effects of PDGF-BB- and PDGF-AA-neutralizing antibodies on IL-6 and hyaluronan production in GO whole orbital tissue cultures were tested. RESULTS GO orbital tissue showed increased PDGF-A and PDGF-B mRNA and protein levels. Increased numbers of PDGF-A- and PDGF-B-positive monocytes, macrophages, and mast cells were present in GO orbital tissue. PDGF-BB stimulated proliferation and hyaluronan and IL-6 production by orbital fibroblasts the most, followed by PDGF-AB and PDGF-AA. Finally, in particular imatinib mesylate and PDGF-BB-neutralizing antibodies reduced IL-6 and hyaluronan production by whole orbital tissue cultures from GO patients. CONCLUSIONS In GO, mast cells, monocytes, and macrophages may activate orbital fibroblasts via secretion of especially PDGF-AB and PDGF-BB. Preclinical studies with whole orbital tissue cultures show that blocking PDGF-B chain containing isoforms can be a promising treatment for GO.
Collapse
Affiliation(s)
- L van Steensel
- Department of Immunology, Erasmus Medical Center, University Medical Center, and Rotterdam Eye Hospital, P.O. Box 2040, 3000 CA, Rotterdam, The Netherlands
| | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Zhang L, Grennan-Jones F, Lane C, Rees DA, Dayan CM, Ludgate M. Adipose tissue depot-specific differences in the regulation of hyaluronan production of relevance to Graves' orbitopathy. J Clin Endocrinol Metab 2012; 97:653-62. [PMID: 22162480 DOI: 10.1210/jc.2011-1299] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
CONTEXT Graves' orbitopathy (GO) is associated with Graves' disease, in which anti-TSH receptor (TSHR) autoantibodies (thyroid-stimulating antibodies) increase cAMP causing hyperthyroidism. Excess adipogenesis and hyaluronan (HA) overproduction [HA synthase 2 (HAS2) is the major source] expand the orbital contents causing GO. TSHR activation participates in both processes but an anti-TSHR monoclonal without TSAB activity also increased HA, suggesting the involvement of other cascades. OBJECTIVE AND PATIENTS STUDIED: We investigated using in vitro models in which preadipocytes/fibroblasts from human orbital (n = 12) and sc (n = 10) adipose tissues were treated with IGF-I (to probe the pAkt pathway, recently identified as a positive regulator of HAS2), TSH, and/or various inhibitors. Changes in HA during in vitro-induced adipogenesis were also evaluated. MAIN OUTCOME AND RESULTS Adipogenesis in orbital preadipocytes was accompanied by significantly increased HAS2 transcripts and HA accumulation in contrast to sc cells in which differentiation significantly decreased HAS2 mRNA and secreted HA. Surprisingly, IGF-I alone did not increase HAS2 levels, despite significantly increasing the ratio of phosphorylated to total Akt; furthermore, an Akt inhibitor increased orbital (but not sc) HAS2 transcripts. A stimulatory effect of IGF-I on HAS2 transcripts was revealed by addition of rapamycin in sc but by a MAPK kinase inhibitor in orbital fibroblasts. CONCLUSIONS The results have several possible explanations including a phosphorylation-dependent repressor of HAS2 transcript accumulation, exclusively in the orbit. The difference in control of HAS2 expression allows the activation of one of the mechanisms underlying GO, adipogenesis, to be linked biologically with the second, HA overproduction.
Collapse
Affiliation(s)
- Lei Zhang
- Centre for Endocrine and Diabetes Sciences, School of Medicine, Cardiff University, Cardiff, UK
| | | | | | | | | | | |
Collapse
|
29
|
Woodward DF, Jones RL, Narumiya S. International Union of Basic and Clinical Pharmacology. LXXXIII: classification of prostanoid receptors, updating 15 years of progress. Pharmacol Rev 2011; 63:471-538. [PMID: 21752876 DOI: 10.1124/pr.110.003517] [Citation(s) in RCA: 332] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
It is now more than 15 years since the molecular structures of the major prostanoid receptors were elucidated. Since then, substantial progress has been achieved with respect to distribution and function, signal transduction mechanisms, and the design of agonists and antagonists (http://www.iuphar-db.org/DATABASE/FamilyIntroductionForward?familyId=58). This review systematically details these advances. More recent developments in prostanoid receptor research are included. The DP(2) receptor, also termed CRTH2, has little structural resemblance to DP(1) and other receptors described in the original prostanoid receptor classification. DP(2) receptors are more closely related to chemoattractant receptors. Prostanoid receptors have also been found to heterodimerize with other prostanoid receptor subtypes and nonprostanoids. This may extend signal transduction pathways and create new ligand recognition sites: prostacyclin/thromboxane A(2) heterodimeric receptors for 8-epi-prostaglandin E(2), wild-type/alternative (alt4) heterodimers for the prostaglandin FP receptor for bimatoprost and the prostamides. It is anticipated that the 15 years of research progress described herein will lead to novel therapeutic entities.
Collapse
Affiliation(s)
- D F Woodward
- Dept. of Biological Sciences RD3-2B, Allergan, Inc., 2525 Dupont Dr., Irvine, CA 92612, USA.
| | | | | |
Collapse
|
30
|
Tsui S, Fernando R, Chen B, Smith TJ. Divergent Sp1 protein levels may underlie differential expression of UDP-glucose dehydrogenase by fibroblasts: role in susceptibility to orbital Graves disease. J Biol Chem 2011; 286:24487-99. [PMID: 21576248 DOI: 10.1074/jbc.m111.241166] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
UDP-glucose dehydrogenase (UGDH) catalyzes the formation of UDP-glucuronate. Glucuronate represents an integral component of the glycosaminoglycan, hyaluronan, which accumulates in orbital Graves disease. Here we report that orbital fibroblasts express higher levels of UGDH than do those from skin. This is a consequence of greater UGDH gene promoter activity and more abundant steady-state UGDH mRNA. Six Sp1 sites located in the proximal 550 bp of the UGDH gene promoter appear to determine basal promoter activity, as does a previously unrecognized 49-bp sequence spanning -1436 nucleotides (nt) and -1388 nt that negatively affects activity. Nuclear Sp1 protein is more abundant in orbital fibroblasts, and its binding to specific sites on DNA is greater than that in dermal fibroblasts. Mutating each of these Sp1 sites in a UGDH gene promoter fragment, extending from -1387 to +71 nt and fused to a luciferase reporter, results in divergent activities when transfected in orbital and dermal fibroblasts. Reducing Sp1 attenuated UGDH gene promoter activity, lowered steady-state UGDH mRNA levels, and reduced UGDH enzyme activity. Targeting Sp1 and UGDH with specific siRNAs also lowered hyaluronan synthase-1 (HAS-1) and HAS-2 levels and reduced hyaluronan accumulation in orbital fibroblasts. These findings suggest that orbital fibroblasts express high levels of UGDH in an anatomic-specific manner, apparently the result of greater constitutive Sp1. These high UGDH levels may underlie susceptibility of the orbit to localized overproduction of hyaluronan in Graves disease.
Collapse
Affiliation(s)
- Shanli Tsui
- Division of Molecular Medicine, Department of Medicine, Harbor-UCLA Medical Center, Torrance, California 90502, USA
| | | | | | | |
Collapse
|
31
|
Guo N, Woeller CF, Feldon SE, Phipps RP. Peroxisome proliferator-activated receptor gamma ligands inhibit transforming growth factor-beta-induced, hyaluronan-dependent, T cell adhesion to orbital fibroblasts. J Biol Chem 2011; 286:18856-67. [PMID: 21454487 DOI: 10.1074/jbc.m110.179317] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Thyroid eye disease is characterized by the infiltration of leukocytes and accumulation of hyaluronan (HA) in orbital tissue. Inflamed orbital tissue expands in size due to excessive HA and to the formation of scar tissue (fibrosis) and/or adipose accumulation. Transforming growth factor β (TGF-β) acts as a key inducer of fibrosis by enhancing extracellular matrix production. Treatment of primary human orbital fibroblasts with TGF-β led to significant increases in both HA synthesis and secretion. TGF-β also strongly induced hyaluronan synthase 1 (HAS1) and HAS2 mRNA levels, which increased 50- and 6-fold, respectively. Remarkably, the addition of the peroxisome proliferator-activated receptor (PPARγ) ligands pioglitazone (Pio) or rosiglitazone (Rosi) to TGF-β-treated orbital fibroblasts attenuated HA synthesis and reduced HAS1 and HAS2 mRNA levels. The attenuation of TGF-β function by Pio and Rosi was independent of PPARγ activity. Furthermore, Pio and Rosi treatment inhibited TGF-β-induced T cell adhesion to orbital fibroblasts. Our findings demonstrate that TGF-β plays an important role in HA synthesis and in the inflammatory response by enhancing or facilitating inflammatory cell infiltration and adhesion to orbital tissue. Pio and Rosi exhibit anti-fibrotic and anti-inflammatory activity and may be useful in treating thyroid eye disease.
Collapse
Affiliation(s)
- Naxin Guo
- Flaum Eye Institute, University of Rochester School of Medicine and Dentistry, Rochester, New York 14642, USA
| | | | | | | |
Collapse
|