1
|
Liguori L, Luciano A, Polcaro G, Ottaiano A, Cascella M, Perri F, Pepe S, Sabbatino F. Prior Anti-Angiogenic TKI-Based Treatment as Potential Predisposing Factor to Nivolumab-Mediated Recurrent Thyroid Disorder Adverse Events in mRCC Patients: A Case Series. Biomedicines 2023; 11:2974. [PMID: 38001973 PMCID: PMC10669217 DOI: 10.3390/biomedicines11112974] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 10/26/2023] [Accepted: 11/02/2023] [Indexed: 11/26/2023] Open
Abstract
Immune checkpoint inhibitors (ICIs) targeting programmed cell death 1 (PD-1) or its ligand 1 (PD-L1) have revolutionized the management of many types of solid tumors, including metastatic renal cell carcinoma (mRCC). Both sequential and combinatorial therapeutic strategies utilizing anti-PD-1 monoclonal antibodies (mAbs) and anti-angiogenic tyrosine kinase inhibitors (TKIs) have demonstrated to improve the survival of patients with mRCC as compared to standard therapies. On the other hand, both ICIs and TKIs are well known to potentially cause thyroid disorder adverse events (TDAEs). However, in the context of sequential therapeutic strategy, it is not clear whether prior anti-angiogenic TKI may increase the risk and/or the severity of ICI-related TDAEs. In this work, by describing and analyzing a case series of mRCC patients treated sequentially with prior TKIs and then with ICIs, we investigated the role of prior anti-angiogenic TKI-based treatment as a potential predisposing factor to anti-PD-1-mediated recurrent TDAEs, as well as its potential impact on the clinical characteristics of nivolumab-mediated recurrent TDAEs. Fifty mRCC patients were included in the analysis. TKI-mediated TDAEs were reported in ten out of fifty patients. TKI-mediated TDAEs were characterized by hypothyroidism in all ten patients. Specifically, 40%, 40% and 20% of patients presented grade 1, 2 and 3 hypothyroidisms, respectively. Following tumor progression and during anti-PD-1 nivolumab treatment, five out of ten patients developed anti-PD-1 nivolumab-mediated recurrent TDAEs. Anti-PD-1 nivolumab-mediated recurrent TDAEs were characterized by an early transient phase of thyrotoxicosis and a late phase of hypothyroidism in all five patients. The TDAEs were grade 1 and 2 in four and one patients, respectively. Prior anti-angiogenic TKI did not modify the clinical characteristics of nivolumab-mediated recurrent TDAEs. However, all five patients required an increased dosage of levothyroxine replacement therapy. In conclusion, our work suggests that prior anti-angiogenic TKI-based treatment significantly increases the risk of ICI-mediated recurrent TDAEs in patients with mRCC without modifying their clinical characteristics. The most relevant effect for these patients is the need to increase the dosage of lifelong levothyroxine replacement therapy.
Collapse
Affiliation(s)
- Luigi Liguori
- Oncology Unit, Department of Clinical Medicine and Surgery, University of Naples “Federico II”, 80131 Naples, Italy; (L.L.); (A.L.)
- Oncology Unit, Department of Medicine, Surgery and Dentistry, University of Salerno, 84081 Baronissi, Italy; (G.P.); (S.P.)
| | - Angelo Luciano
- Oncology Unit, Department of Clinical Medicine and Surgery, University of Naples “Federico II”, 80131 Naples, Italy; (L.L.); (A.L.)
- Oncology Unit, Department of Medicine, Surgery and Dentistry, University of Salerno, 84081 Baronissi, Italy; (G.P.); (S.P.)
| | - Giovanna Polcaro
- Oncology Unit, Department of Medicine, Surgery and Dentistry, University of Salerno, 84081 Baronissi, Italy; (G.P.); (S.P.)
| | - Alessandro Ottaiano
- SSD Innovative Therapies for Abdominal Metastases, Abdominal Oncology, INT IRCCS Foundation “G. Pascale”, 80131 Naples, Italy;
| | - Marco Cascella
- Unit of Anesthesiology, Intensive Care Medicine, and Pain, Department of Medicine, Surgery and Dentistry Medicine, University of Salerno, 84081 Baronissi, Italy;
| | - Francesco Perri
- Medical and Experimental Head and Neck Oncology Unit, INT IRCCS Foundation “G. Pascale”, 80131 Naples, Italy;
| | - Stefano Pepe
- Oncology Unit, Department of Medicine, Surgery and Dentistry, University of Salerno, 84081 Baronissi, Italy; (G.P.); (S.P.)
| | - Francesco Sabbatino
- Oncology Unit, Department of Medicine, Surgery and Dentistry, University of Salerno, 84081 Baronissi, Italy; (G.P.); (S.P.)
| |
Collapse
|
2
|
Yang M, Lai Y, Gan D, Liu Q, Wang Y, He X, An Y, Gao T. Possible molecular exploration of herbal pair Haizao-Kunbu in the treatment of Graves' disease by network pharmacology, molecular docking, and molecular dynamic analysis. Front Endocrinol (Lausanne) 2023; 14:1236549. [PMID: 37859983 PMCID: PMC10583570 DOI: 10.3389/fendo.2023.1236549] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 09/22/2023] [Indexed: 10/21/2023] Open
Abstract
Objective To promote the development and therapeutic application of new medications, it is crucial to conduct a thorough investigation into the mechanism by which the traditional Chinese herb pair of Haizao-Kunbu (HK) treats Graves' disease (GD). Materials and methods Chemical ingredients of HK, putative target genes, and GD-associated genes were retrieved from online public databases. Using Cytoscape 3.9.1, a compound-gene target network was established to explore the association between prosperous ingredients and targets. STRING, Gene Ontology, and Kyoto Encyclopedia of Genes and Genomes pathway analyses visualized core targets and disease pathways. Additionally, we conducted a refined analysis of the binding interactions between active ingredients and their respective targets. To visualize these findings, we employed precise molecular docking techniques. Furthermore, we carried out molecular dynamics simulations to gain insights into the formation of more tightly bound complexes. Results We found that there were nine key active ingredients in HK, which mainly acted on 21 targets. These targets primarily regulated several biological processes such as cell population proliferation, protein phosphorylation, and regulation of kinase activity, and acted on PI3K-AKT and MAPK pathways to treat GD. Analysis of the molecular interaction simulation under computer technology revealed that the key targets exhibited strong binding activity to active ingredients, and Fucosterol-AKT1 and Isofucosterol-AKT1 complexes were highly stable in humans. Conclusion This study demonstrates that HK exerts therapeutic effects on GD in a multi-component, multi-target, and multi-pathway manner by regulating cell proliferation, differentiation, inflammation, and immunomodulatory-related targets. This study provides a theoretical foundation for further investigation into GD.
Collapse
Affiliation(s)
- Mengfei Yang
- Graduate School, Liaoning University of Traditional Chinese Medicine, Shenyang, Liaoning, China
| | - Yiwen Lai
- Department of Endocrinology, The Affiliated Hospital of Liaoning University of Traditional Chinese Medicine, Shenyang, Liaoning, China
| | - Di Gan
- Graduate School, Liaoning University of Traditional Chinese Medicine, Shenyang, Liaoning, China
| | - Qingyang Liu
- Department of Endocrinology, The Affiliated Hospital of Liaoning University of Traditional Chinese Medicine, Shenyang, Liaoning, China
| | - Yingna Wang
- Department of Endocrinology, The Affiliated Hospital of Liaoning University of Traditional Chinese Medicine, Shenyang, Liaoning, China
| | - Xinyong He
- Insititute of Laboratory Medicine, Liaoning University of Traditional Chinese Medicine, Shenyang, Liaoning, China
| | - Yi An
- Department of Obstetrics, The People’s Hospital of Liaoning, Shenyang, Liaoning, China
| | - Tianshu Gao
- Department of Endocrinology, The Affiliated Hospital of Liaoning University of Traditional Chinese Medicine, Shenyang, Liaoning, China
| |
Collapse
|
3
|
Chartoumpekis DV, Ziros PG, Georgakopoulos-Soares I, Smith AAT, Marques AC, Ibberson M, A. Kopp P, Habeos I, Trougakos IP, Khoo NKH, Sykiotis GP. The Transcriptomic Response of the Murine Thyroid Gland to Iodide Overload and the Role of the Nrf2 Antioxidant System. Antioxidants (Basel) 2020; 9:antiox9090884. [PMID: 32961913 PMCID: PMC7555824 DOI: 10.3390/antiox9090884] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 09/16/2020] [Accepted: 09/16/2020] [Indexed: 12/23/2022] Open
Abstract
Background: Thyroid follicular cells have physiologically high levels of reactive oxygen species because oxidation of iodide is essential for the iodination of thyroglobulin (Tg) during thyroid hormone synthesis. Thyroid follicles (the functional units of the thyroid) also utilize incompletely understood autoregulatory mechanisms to defend against exposure to excess iodide. To date, no transcriptomic studies have investigated these phenomena in vivo. Nuclear erythroid factor 2 like 2 (Nrf2 or Nfe2l2) is a transcription factor that regulates the expression of numerous antioxidant and other cytoprotective genes. We showed previously that the Nrf2 pathway regulates the antioxidant defense of follicular cells, as well as Tg transcription and Tg iodination. We, thus, hypothesized that Nrf2 might be involved in the transcriptional response to iodide overload. Methods: C57BL6/J wild-type (WT) or Nrf2 knockout (KO) male mice were administered regular water or water supplemented with 0.05% sodium iodide for seven days. RNA from their thyroids was prepared for next-generation RNA sequencing (RNA-Seq). Gene expression changes were assessed and pathway analyses were performed on the sets of differentially expressed genes. Results: Analysis of differentially expressed messenger RNAs (mRNAs) indicated that iodide overload upregulates inflammatory-, immune-, fibrosis- and oxidative stress-related pathways, including the Nrf2 pathway. Nrf2 KO mice showed a more pronounced inflammatory–autoimmune transcriptional response to iodide than WT mice. Compared to previously published datasets, the response patterns observed in WT mice had strong similarities with the patterns typical of Graves’ disease and papillary thyroid carcinoma (PTC). Long non-coding RNAs (lncRNAs) and microRNAs (miRNAs) also responded to iodide overload, with the latter targeting mRNAs that participate mainly in inflammation pathways. Conclusions: Iodide overload induces the Nrf2 cytoprotective response and upregulates inflammatory, immune, and fibrosis pathways similar to autoimmune hyperthyroidism (Graves’ disease) and PTC.
Collapse
Affiliation(s)
- Dionysios V. Chartoumpekis
- Service of Endocrinology and Diabetology, Lausanne University Hospital, and Faculty of Biology and Medicine, University of Lausanne, 1011 Lausanne, Switzerland; (D.V.C.); (P.G.Z.); (P.A.K.)
- Division of Endocrinology, Department of Internal Medicine, University of Patras, 26504 Patras, Greece;
| | - Panos G. Ziros
- Service of Endocrinology and Diabetology, Lausanne University Hospital, and Faculty of Biology and Medicine, University of Lausanne, 1011 Lausanne, Switzerland; (D.V.C.); (P.G.Z.); (P.A.K.)
| | - Ilias Georgakopoulos-Soares
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, CA 94158, USA;
- Institute for Human Genetics, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Adam A. T. Smith
- Department of Computational Biology, University of Lausanne, 1015 Lausanne, Switzerland; (A.A.T.S.); (A.C.M.)
| | - Ana Claudia Marques
- Department of Computational Biology, University of Lausanne, 1015 Lausanne, Switzerland; (A.A.T.S.); (A.C.M.)
| | - Mark Ibberson
- Vital-IT Group, Swiss Institute of Bioinformatics, 1015 Lausanne, Switzerland;
| | - Peter A. Kopp
- Service of Endocrinology and Diabetology, Lausanne University Hospital, and Faculty of Biology and Medicine, University of Lausanne, 1011 Lausanne, Switzerland; (D.V.C.); (P.G.Z.); (P.A.K.)
| | - Ioannis Habeos
- Division of Endocrinology, Department of Internal Medicine, University of Patras, 26504 Patras, Greece;
| | - Ioannis P. Trougakos
- Department of Cell Biology and Biophysics, Faculty of Biology, National and Kapodistrian University of Athens, 15784 Athens, Greece;
| | - Nicholas K. H. Khoo
- Department of Pharmacology & Chemical Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA;
| | - Gerasimos P. Sykiotis
- Service of Endocrinology and Diabetology, Lausanne University Hospital, and Faculty of Biology and Medicine, University of Lausanne, 1011 Lausanne, Switzerland; (D.V.C.); (P.G.Z.); (P.A.K.)
- Correspondence: ; Tel.: +41-21-314-0606
| |
Collapse
|
4
|
Tsai CH, Yang PS, Lee JJ, Liu TP, Kuo CY, Cheng SP. Effects of Preoperative Iodine Administration on Thyroidectomy for Hyperthyroidism: A Systematic Review and Meta-analysis. Otolaryngol Head Neck Surg 2019; 160:993-1002. [PMID: 30721111 DOI: 10.1177/0194599819829052] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
OBJECTIVE The current guidelines recommend that potassium iodide be given in the immediate preoperative period for patients with Graves' disease who are undergoing thyroidectomy. Nonetheless, the evidence behind this recommendation is tenuous. The purpose of this study is to clarify the benefits of preoperative iodine administration from published comparative studies. DATA SOURCES We searched PubMed, Embase, Cochrane, and CINAHL from 1980 to June 2018. REVIEW METHODS Studies were included that compared preoperative iodine administration and no premedication before thyroidectomy. For the meta-analysis, studies were pooled with the random-effects model. RESULTS A total of 510 patients were divided into the iodine (n = 223) and control (n = 287) groups from 9 selected studies. Preoperative iodine administration was significantly associated with decreased thyroid vascularity and intraoperative blood loss. Significant heterogeneity was present among studies. We found no significant difference in thyroid volume or operative time. Furthermore, the meta-analysis showed no difference in the risk of postoperative complications, including vocal cord palsy, hypoparathyroidism/hypocalcemia, and hemorrhage or hematoma after thyroidectomy. CONCLUSION Preoperative iodine administration decreases thyroid vascularity and intraoperative blood loss. Nonetheless, it does not translate to more clinically meaningful differences in terms of operative time and postoperative complications.
Collapse
Affiliation(s)
- Chung-Hsin Tsai
- 1 Department of Surgery, MacKay Memorial Hospital and Mackay Medical College, Taipei, Taiwan.,2 Mackay Junior College of Medicine, Nursing, and Management, Taipei, Taiwan
| | - Po-Sheng Yang
- 1 Department of Surgery, MacKay Memorial Hospital and Mackay Medical College, Taipei, Taiwan
| | - Jie-Jen Lee
- 1 Department of Surgery, MacKay Memorial Hospital and Mackay Medical College, Taipei, Taiwan
| | - Tsang-Pai Liu
- 1 Department of Surgery, MacKay Memorial Hospital and Mackay Medical College, Taipei, Taiwan.,2 Mackay Junior College of Medicine, Nursing, and Management, Taipei, Taiwan
| | - Chi-Yu Kuo
- 1 Department of Surgery, MacKay Memorial Hospital and Mackay Medical College, Taipei, Taiwan
| | - Shih-Ping Cheng
- 1 Department of Surgery, MacKay Memorial Hospital and Mackay Medical College, Taipei, Taiwan.,3 Department of Pharmacology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
5
|
Wang Q, Shen Y, Ye B, Hu H, Fan C, Wang T, Zheng Y, Lv J, Ma Y, Xiang M. Gene expression differences between thyroid carcinoma, thyroid adenoma and normal thyroid tissue. Oncol Rep 2018; 40:3359-3369. [PMID: 30272326 PMCID: PMC6196629 DOI: 10.3892/or.2018.6717] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2018] [Accepted: 09/03/2018] [Indexed: 12/14/2022] Open
Abstract
To identify differences in gene expression profiles of infected cells between thyroid carcinoma (C), thyroid adenoma (A) and normal thyroid (N) epithelial cells, differentially expressed genes were identified using three pairwise comparisons with the GEO2R online tool. Gene ontology and Kyoto Encyclopedia of Genes and Genomes pathway enrichment analysis were used to classify them at the functional level. The most significant cluster in the N vs. A pairwise comparison had four hub genes: Insulin-like growth factor 2, Von Willebrand factor (VWF), multimerin 1 (MMRN1) and complement factor D (CFD). In N vs. C, the most significant cluster had 19 genes: IGF2, early growth response 2, transcription factor 3, KIT proto‑oncogene receptor tyrosine kinase, SMAD family member 9, MLLT3 super elongation complex subunit, runt related transcription factor 1, CFD, actinin α 1, SWI/SNF related matrix associated actin dependent regulator of chromatin subfamily a member 4, JunD proto‑oncogene AP‑1 transcription factor subunit, serum response factor (SRF), FosB proto‑oncogene, AP‑1 transcription factor subunit, connective tissue growth factor (CTGF), SRC proto‑oncogene, non‑receptor tyrosine kinase, MMRN1, SRY‑box 9, early growth response 3 and ETS variant 4. In A vs. C, the most significant cluster had 14 genes: BCL2-like 1, galectin 3, MCL1 BCL2 family apoptosis regulator, DNA damage inducible transcript 3, BCL2 apoptosis regulator, CTGF, matrix metallopeptidase 7, early growth response 1, kinase insert domain receptor, TIMP metallopeptidase inhibitor 1, apolipoprotein E, VWF, cyclin D1 and placental growth factor. Histological evidence was presented to confirm the makeup of the hubs prior to logistic regression analysis to differentiate benign and malignant neoplasms. The results of the present study may aid in the search for novel potential biomarkers for the differential diagnosis, prognosis and development of drug targets of thyroid neoplasm.
Collapse
Affiliation(s)
- Quan Wang
- Department of Otolaryngology & Head and Neck Surgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, P.R. China
| | - Yilin Shen
- Department of Otolaryngology & Head and Neck Surgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, P.R. China
| | - Bin Ye
- Department of Otolaryngology & Head and Neck Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, P.R. China
| | - Haixia Hu
- Department of Otolaryngology & Head and Neck Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, P.R. China
| | - Cui Fan
- Department of Otolaryngology & Head and Neck Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, P.R. China
| | - Tan Wang
- Department of Otolaryngology & Head and Neck Surgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, P.R. China
| | - Yuqin Zheng
- Department of Otolaryngology & Head and Neck Surgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, P.R. China
| | - Jingrong Lv
- Department of Otolaryngology & Head and Neck Surgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, P.R. China
| | - Yan Ma
- Department of Otolaryngology & Head and Neck Surgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, P.R. China
| | - Mingliang Xiang
- Department of Otolaryngology & Head and Neck Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, P.R. China
| |
Collapse
|
6
|
Effect of the micronutrient iodine in thyroid carcinoma angiogenesis. Aging (Albany NY) 2017; 8:3180-3184. [PMID: 27997357 PMCID: PMC5270662 DOI: 10.18632/aging.101143] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Accepted: 12/15/2016] [Indexed: 11/25/2022]
Abstract
Iodide is a micronutrient essential for thyroid hormone production. The uptake and metabolism of iodide by thyrocytes is crucial to proper thyroid function. Iodide ions are drawn into the thyroid follicular cell via the sodium-iodide symporter (NIS) in the cell membrane and become integrated into tyrosyl residues to ultimately form thyroid hormones. We sought to learn how an abnormal concentration of iodide within thyrocyte can have significant effects on the thyroid, specifically the surrounding vascular network. Insufficient levels of iodide can lead to increased expression or activity of several pathways, including vascular endothelial growth factor (VEGF). The VEGF protein fuel vessel growth (angiogenesis) and therefore enhances the nutrients available to surrounding cells. Alternatively, normal/surplus iodide levels can have inhibitory effects on angiogenesis. Varying levels of iodide in the thyroid can influence thyroid carcinoma cell proliferation and angiogenesis via regulation of the hypoxia inducible factor-1 (HIF-1) and VEGF-dependent pathway. We have reviewed a number of studies to investigate how the effect of iodide on angiogenic and oxidative stress regulation can affect the viability of thyroid carcinoma cells. The various studies outlined give key insights to the role of iodide in thyroid follicles function and vascular growth, generally highlighting that insufficient levels of iodide stimulate pathways resulting in vascular growth, and viceversa normal/surplus iodide levels inhibit such pathways. Intriguingly, TSH and iodine levels differentially regulate the expression levels of angiogenic factors. All cells, including carcinoma cells, increase uptake of blood nutrients, meaning the vascular profile is influential to tumor growth and progression. Importantly, variation in the iodine concentrations also influence BRAFV600E-mediated oncogenic activity and might deregulate tumor proliferation. Although the mechanisms are not well eluted, iodine concentrations and metabolism might have a crucial influence on thyroid carcinoma cell viability via regulation of different molecular pathways, including angiogenesis regulatory autocrine and microenvironment-mediated signals.
Collapse
|
7
|
Bos MM, Smit RAJ, Trompet S, van Heemst D, Noordam R. Thyroid Signaling, Insulin Resistance, and 2 Diabetes Mellitus: A Mendelian Randomization Study. J Clin Endocrinol Metab 2017; 102:1960-1970. [PMID: 28323940 DOI: 10.1210/jc.2016-2816] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Accepted: 03/02/2017] [Indexed: 02/10/2023]
Abstract
Context Increasing evidence suggests an association between thyroid-stimulating hormone (TSH), free thyroxine (fT4), and deiodinases with insulin resistance and type 2 diabetes mellitus (T2D). Objective We examined whether TSH and fT4 levels and deiodinases are causally associated with insulin resistance and T2D, using Mendelian randomization. Methods We selected 20 genetic variants for TSH level and four for fT4 level (identified in a genome-wide association study (GWAS) meta-analysis of European-ancestry cohorts) as instrumental variables for TSH and fT4 levels, respectively. We used summary data from GWASs on the outcomes T2D [Diabetes, Genetics Replication and Meta-analysis (DIAGRAM), n = 12,171 cases and n = 56,862 control subjects] and glycemic traits in patients without diabetes [Meta-Analyses of Glucose and Insulin-Related Traits Consortium (MAGIC), n = 46,186 for fasting glucose and insulin and n = 46,368 for hemoglobin A1c]. To examine whether the associations between TSH/fT4 levels and the study outcomes were causal, we combined the effects of the genetic instruments. Furthermore, we examined the associations among 16 variants in DIO1, DIO2, DIO3, and T2D and glycemic traits. Results We found no evidence for an association between the combined genetic instrumental variables for TSH and fT4 and the study outcomes. For example, we did not observe a genetically determined association between high TSH level and T2D (odds ratio, 0.91 per standard deviation TSH increase; 95% confidence interval, 0.78 to 1.07). Selected genetic variants in DIO1 (e.g., rs7527713) were associated with measures of insulin resistance. Conclusion We found no evidence for a causal association between circulatory levels of TSH and fT4 with insulin resistance and T2D, but we found suggestive evidence that DIO1 affects glucose metabolism.
Collapse
Affiliation(s)
- Maxime M Bos
- Department of Internal Medicine, Section of Gerontology and Geriatrics, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Roelof A J Smit
- Department of Internal Medicine, Section of Gerontology and Geriatrics, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
- Department of Cardiology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Stella Trompet
- Department of Internal Medicine, Section of Gerontology and Geriatrics, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
- Department of Cardiology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Diana van Heemst
- Department of Internal Medicine, Section of Gerontology and Geriatrics, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Raymond Noordam
- Department of Internal Medicine, Section of Gerontology and Geriatrics, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| |
Collapse
|
8
|
Nagasaki S, Fukui M, Asano S, Ono K, Miki Y, Araki SI, Isobe M, Nakashima N, Takahashi K, Sasano H, Sato J. Induction of adrenomedullin 2/intermedin expression by thyroid stimulating hormone in thyroid. Mol Cell Endocrinol 2014; 395:32-40. [PMID: 25102228 DOI: 10.1016/j.mce.2014.07.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2014] [Revised: 07/09/2014] [Accepted: 07/11/2014] [Indexed: 10/24/2022]
Abstract
TSH is the important regulator of thyroid function but detailed molecular mechanisms have not been clarified. We first generated the iodine deficient (ID) rat in which goiter is induced by accelerated endogenous TSH secretion. The result of microarray analysis demonstrated markedly increased levels of adrenomedullin 2/intermedin (AM2/IMD) expression in the ID rat thyroid. AM2/IMD is a potent vasodilator. AM2/IMD mRNA expression was induced by TSH in a rat thyroid follicular cell line FRTL-5. Immunohistochemical analysis in human normal and Graves' disease thyroid revealed that AM2/IMD immunoreactivity was detected in follicular cells and more pronounced in Graves' disease. These results indicated that TSH induced AM2/IMD expression in the rat thyroid gland and it could locally work as a potent vasodilator, resulting in the expansion of thyroid inter-follicular capillaries. AM2/IMD could also contribute to facilitate thyroid hormone synthesis possibly via vasodilation effects and/or cAMP stimulating effects in the human thyroid gland.
Collapse
Affiliation(s)
- Shuji Nagasaki
- Drug Discovery Department, ASKA Pharmaceutical Co., Ltd., Kawasaki, Japan; Department of Pathology, Tohoku University Graduate School of Medicine, Sendai, Japan.
| | - Motoko Fukui
- Safety Research Department, ASKA Pharmaceutical Co., Ltd., Kawasaki, Japan
| | - Satoko Asano
- Drug Discovery Department, ASKA Pharmaceutical Co., Ltd., Kawasaki, Japan
| | - Katsuhiko Ono
- Department of Pathology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Yasuhiro Miki
- Department of Pathology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Sei-ichi Araki
- Safety Research Department, ASKA Pharmaceutical Co., Ltd., Kawasaki, Japan
| | - Mitsui Isobe
- Safety Research Department, ASKA Pharmaceutical Co., Ltd., Kawasaki, Japan
| | - Noriaki Nakashima
- Department of Breast and Endocrine Surgery, Tohoku University Hospital, Sendai, Japan
| | - Kazuhiro Takahashi
- Department of Endocrinology and Applied Medical Science, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Hironobu Sasano
- Department of Pathology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Jun Sato
- Drug Discovery Department, ASKA Pharmaceutical Co., Ltd., Kawasaki, Japan
| |
Collapse
|
9
|
Tyrosine kinase inhibitors induced thyroid dysfunction: a review of its incidence, pathophysiology, clinical relevance, and treatment. BIOMED RESEARCH INTERNATIONAL 2013; 2013:725410. [PMID: 24282820 PMCID: PMC3824811 DOI: 10.1155/2013/725410] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 07/16/2013] [Revised: 09/06/2013] [Accepted: 09/08/2013] [Indexed: 01/07/2023]
Abstract
Tyrosine kinase inhibitors (TKI) belong to a new class of molecular multitargeted anticancer therapy which targets different growth factor receptors and hence attenuates cancer cell survival and growth. Since their introduction as adjunct treatment for renal cell carcinoma and gastrointestinal stromal tumors (GIST), a number of reports have demonstrated that TKI can induce thyroid dysfunction which was especially more common with sunitinib maleate. Many mechanisms with respect to this adverse effect of tyrosine kinase inhibitors have been proposed including their induction of thyroiditis, capillary regression in the thyroid gland, antithyroid peroxidase antibody production, and their ability to decrease iodine uptake by the thyroid gland. Of interest is the observation that TKI-induced thyroid dysfunction may actually be protective as it was shown to improve overall survival, and it was suggested that it may have a prognostic value. Followup on thyroid function tests while patients are maintained on tyrosine kinase inhibitor is strongly recommended. When thyroid dysfunction occurs, appropriate treatment should be individualized depending on patients symptoms and thyroid stimulating hormone level.
Collapse
|
10
|
Porcu E, Medici M, Pistis G, Volpato CB, Wilson SG, Cappola AR, Bos SD, Deelen J, den Heijer M, Freathy RM, Lahti J, Liu C, Lopez LM, Nolte IM, O'Connell JR, Tanaka T, Trompet S, Arnold A, Bandinelli S, Beekman M, Böhringer S, Brown SJ, Buckley BM, Camaschella C, de Craen AJM, Davies G, de Visser MCH, Ford I, Forsen T, Frayling TM, Fugazzola L, Gögele M, Hattersley AT, Hermus AR, Hofman A, Houwing-Duistermaat JJ, Jensen RA, Kajantie E, Kloppenburg M, Lim EM, Masciullo C, Mariotti S, Minelli C, Mitchell BD, Nagaraja R, Netea-Maier RT, Palotie A, Persani L, Piras MG, Psaty BM, Räikkönen K, Richards JB, Rivadeneira F, Sala C, Sabra MM, Sattar N, Shields BM, Soranzo N, Starr JM, Stott DJ, Sweep FCGJ, Usala G, van der Klauw MM, van Heemst D, van Mullem A, H.Vermeulen S, Visser WE, Walsh JP, Westendorp RGJ, Widen E, Zhai G, Cucca F, Deary IJ, Eriksson JG, Ferrucci L, Fox CS, Jukema JW, Kiemeney LA, Pramstaller PP, Schlessinger D, Shuldiner AR, Slagboom EP, Uitterlinden AG, Vaidya B, Visser TJ, Wolffenbuttel BHR, Meulenbelt I, Rotter JI, Spector TD, Hicks AA, Toniolo D, Sanna S, Peeters RP, Naitza S. A meta-analysis of thyroid-related traits reveals novel loci and gender-specific differences in the regulation of thyroid function. PLoS Genet 2013; 9:e1003266. [PMID: 23408906 PMCID: PMC3567175 DOI: 10.1371/journal.pgen.1003266] [Citation(s) in RCA: 167] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2012] [Accepted: 11/12/2012] [Indexed: 12/15/2022] Open
Abstract
Thyroid hormone is essential for normal metabolism and development, and overt abnormalities in thyroid function lead to common endocrine disorders affecting approximately 10% of individuals over their life span. In addition, even mild alterations in thyroid function are associated with weight changes, atrial fibrillation, osteoporosis, and psychiatric disorders. To identify novel variants underlying thyroid function, we performed a large meta-analysis of genome-wide association studies for serum levels of the highly heritable thyroid function markers TSH and FT4, in up to 26,420 and 17,520 euthyroid subjects, respectively. Here we report 26 independent associations, including several novel loci for TSH (PDE10A, VEGFA, IGFBP5, NFIA, SOX9, PRDM11, FGF7, INSR, ABO, MIR1179, NRG1, MBIP, ITPK1, SASH1, GLIS3) and FT4 (LHX3, FOXE1, AADAT, NETO1/FBXO15, LPCAT2/CAPNS2). Notably, only limited overlap was detected between TSH and FT4 associated signals, in spite of the feedback regulation of their circulating levels by the hypothalamic-pituitary-thyroid axis. Five of the reported loci (PDE8B, PDE10A, MAF/LOC440389, NETO1/FBXO15, and LPCAT2/CAPNS2) show strong gender-specific differences, which offer clues for the known sexual dimorphism in thyroid function and related pathologies. Importantly, the TSH-associated loci contribute not only to variation within the normal range, but also to TSH values outside the reference range, suggesting that they may be involved in thyroid dysfunction. Overall, our findings explain, respectively, 5.64% and 2.30% of total TSH and FT4 trait variance, and they improve the current knowledge of the regulation of hypothalamic-pituitary-thyroid axis function and the consequences of genetic variation for hypo- or hyperthyroidism. Levels of thyroid hormones are tightly regulated by TSH produced in the pituitary, and even mild alterations in their concentrations are strong indicators of thyroid pathologies, which are very common worldwide. To identify common genetic variants associated with the highly heritable markers of thyroid function, TSH and FT4, we conducted a meta-analysis of genome-wide association studies in 26,420 and 17,520 individuals, respectively, of European ancestry with normal thyroid function. Our analysis identified 26 independent genetic variants regulating these traits, several of which are new, and confirmed previously detected polymorphisms affecting TSH (within the PDE8B gene and near CAPZB, MAF/LOC440389, and NR3C2) and FT4 (within DIO1) levels. Gender-specific differences in the genetic effects of several variants for TSH and FT4 levels were identified at several loci, which offer clues to understand the known sexual dimorphism in thyroid function and pathology. Of particular clinical interest, we show that TSH-associated loci contribute not only to normal variation, but also to TSH values outside reference range, suggesting that they may be involved in thyroid dysfunction. Overall, our findings add to the developing landscape of the regulation of thyroid homeostasis and the consequences of genetic variation for thyroid related diseases.
Collapse
Affiliation(s)
- Eleonora Porcu
- Istituto di Ricerca Genetica e Biomedica (IRGB), Consiglio Nazionale delle Ricerche, c/o Cittadella Universitaria di Monserrato, Monserrato, Cagliari, Italy
- Dipartimento di Scienze Biomediche, Università di Sassari, Sassari, Italy
| | - Marco Medici
- Department of Internal Medicine, Erasmus MC, Rotterdam, The Netherlands
| | - Giorgio Pistis
- Division of Genetics and Cell Biology, San Raffaele Research Institute, Milano, Italy
- Università degli Studi di Trieste, Trieste, Italy
| | - Claudia B. Volpato
- Center for Biomedicine, European Academy Bozen/Bolzano (EURAC), Bolzano, Italy (Affiliated Institute of the University of Lübeck, Lübeck, Germany)
| | - Scott G. Wilson
- Department of Endocrinology and Diabetes, Sir Charles Gairdner Hospital, Nedlands, Western Australia, Australia
- Department of Twin Research and Genetic Epidemiology, King's College London, London, United Kingdom
- School of Medicine and Pharmacology, University of Western Australia, Crawley, Western Australia, Australia
| | - Anne R. Cappola
- University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Steffan D. Bos
- Leiden University Medical Center, Molecular Epidemiology, Leiden, The Netherlands
- Netherlands Consortium for Healthy Ageing, Leiden, The Netherlands
| | - Joris Deelen
- Leiden University Medical Center, Molecular Epidemiology, Leiden, The Netherlands
- Netherlands Consortium for Healthy Ageing, Leiden, The Netherlands
| | - Martin den Heijer
- Department of Endocrinology, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
- Department of Internal Medicine, Free University Medical Center, Amsterdam, The Netherlands
| | - Rachel M. Freathy
- Genetics of Complex Traits, Peninsula College of Medicine and Dentistry, University of Exeter, Exeter, United Kingdom
| | - Jari Lahti
- Institute of Behavioural Sciences, University of Helsinki, Helsinki, Finland
| | - Chunyu Liu
- Center for Population Studies, National Heart, Lung, and Blood Institute, Framingham, Massachusetts, United States of America
| | - Lorna M. Lopez
- Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, Edinburgh, United Kingdom
- Department of Psychology, University of Edinburgh, Edinburgh, United Kingdom
| | - Ilja M. Nolte
- Unit of Genetic Epidemiology and Bioinformatics, Department of Epidemiology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Jeffrey R. O'Connell
- Department of Medicine, University of Maryland Medical School, Baltimore, Maryland, United States of America
| | - Toshiko Tanaka
- Clinical Research Branch, National Institute on Aging, Baltimore, Maryland, United States of America
| | - Stella Trompet
- Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands
- Department of Gerontology and Geriatrics, Leiden University Medical Center, Leiden, The Netherlands
| | - Alice Arnold
- Cardiovascular Health Research Unit and Department of Medicine, University of Washington, Seattle, Washington, United States of America
| | | | - Marian Beekman
- Leiden University Medical Center, Molecular Epidemiology, Leiden, The Netherlands
- Netherlands Consortium for Healthy Ageing, Leiden, The Netherlands
| | - Stefan Böhringer
- Leiden University Medical Center, Medical Statistics and Bioinformatics, Leiden, The Netherlands
| | - Suzanne J. Brown
- Department of Endocrinology and Diabetes, Sir Charles Gairdner Hospital, Nedlands, Western Australia, Australia
| | - Brendan M. Buckley
- Department of Pharmacology and Therapeutics, University College Cork, Cork, Ireland
| | - Clara Camaschella
- Division of Genetics and Cell Biology, San Raffaele Research Institute, Milano, Italy
- Vita e Salute University, San Raffaele Scientific Institute, Milano, Italy
| | - Anton J. M. de Craen
- Department of Gerontology and Geriatrics, Leiden University Medical Center, Leiden, The Netherlands
| | - Gail Davies
- Department of Psychology, University of Edinburgh, Edinburgh, United Kingdom
| | - Marieke C. H. de Visser
- Department for Health Evidence, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Ian Ford
- Robertson Center for Biostatistics, University of Glasgow, Glasgow, United Kingdom
| | - Tom Forsen
- Department of Chronic Disease Prevention, National Institute for Health and Welfare, Helsinki, Finland
- Department of General Practice and Primary Health Care, University of Helsinki, Helsinki, Finland
- Helsinki University Central Hospital, Unit of General Practice, Helsinki, Finland
- Vaasa Health Care Centre, Diabetes Unit, Vaasa, Finland
| | - Timothy M. Frayling
- Genetics of Complex Traits, Peninsula College of Medicine and Dentistry, University of Exeter, Exeter, United Kingdom
| | - Laura Fugazzola
- Endocrine Unit, Fondazione Ca' Granda Policlinico and Department of Clinical Sciences and Community Health, University of Milan, Milano, Italy
| | - Martin Gögele
- Center for Biomedicine, European Academy Bozen/Bolzano (EURAC), Bolzano, Italy (Affiliated Institute of the University of Lübeck, Lübeck, Germany)
| | - Andrew T. Hattersley
- Peninsula NIHR Clinical Research Facility, Peninsula College of Medicine and Dentistry, University of Exeter, Exeter, United Kingdom
| | - Ad R. Hermus
- Department of Endocrinology, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
| | - Albert Hofman
- Department of Epidemiology, Erasmus MC, Rotterdam, The Netherlands
- Netherlands Genomics Initiative (NGI)–sponsored Netherlands Consortium for Healthy Aging (NCHA), Rotterdam, The Netherlands
| | | | - Richard A. Jensen
- Cardiovascular Health Research Unit and Department of Medicine, University of Washington, Seattle, Washington, United States of America
| | - Eero Kajantie
- Department of Chronic Disease Prevention, National Institute for Health and Welfare, Helsinki, Finland
- Hospital for Children and Adolescents, Helsinki University Central Hospital and University of Helsinki, Helsinki, Finland
| | - Margreet Kloppenburg
- Department of Clinical Epidemiology and Rheumatology, Leiden University Medical Center, Leiden, The Netherlands
| | - Ee M. Lim
- Department of Endocrinology and Diabetes, Sir Charles Gairdner Hospital, Nedlands, Western Australia, Australia
- Pathwest Laboratory Medicine WA, Nedlands, Western Australia, Australia
| | - Corrado Masciullo
- Division of Genetics and Cell Biology, San Raffaele Research Institute, Milano, Italy
| | - Stefano Mariotti
- Dipartimento di Scienze Mediche, Università di Cagliari, c/o Cittadella Universitaria di Monserrato, Monserrato, Cagliari, Italy
| | - Cosetta Minelli
- Center for Biomedicine, European Academy Bozen/Bolzano (EURAC), Bolzano, Italy (Affiliated Institute of the University of Lübeck, Lübeck, Germany)
| | - Braxton D. Mitchell
- Department of Medicine, University of Maryland Medical School, Baltimore, Maryland, United States of America
| | - Ramaiah Nagaraja
- Laboratory of Genetics, National Institute on Aging, Baltimore, Maryland, United States of America
| | - Romana T. Netea-Maier
- Department of Endocrinology, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
| | - Aarno Palotie
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Cambridge, United Kingdom
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
- Department of Medical Genetics, University of Helsinki and University Central Hospital, Helsinki, Finland
| | - Luca Persani
- Department of Clinical Sciences and Community Health, University of Milan, Milano, Italy
- Division of Endocrinology and Metabolic Diseases, IRCCS Ospedale San Luca, Milan, Italy
| | - Maria G. Piras
- Istituto di Ricerca Genetica e Biomedica (IRGB), Consiglio Nazionale delle Ricerche, c/o Cittadella Universitaria di Monserrato, Monserrato, Cagliari, Italy
| | - Bruce M. Psaty
- Cardiovascular Health Research Unit, Departments of Medicine, Epidemiology, and Health Services, University of Washington, Seattle, Washington, United States of America
- Group Health Research Institute, Group Health Cooperative, Seattle, Washington, United States of America
| | - Katri Räikkönen
- Institute of Behavioural Sciences, University of Helsinki, Helsinki, Finland
| | - J. Brent Richards
- Department of Twin Research and Genetic Epidemiology, King's College London, London, United Kingdom
- Department of Medicine, Jewish General Hospital, McGill University, Montréal, Québec, Canada
- Departments of Human Genetics, Epidemiology, and Biostatistics, Jewish General Hospital, Lady Davis Institute, McGill University, Montréal, Québec
| | - Fernando Rivadeneira
- Department of Internal Medicine, Erasmus MC, Rotterdam, The Netherlands
- Department of Epidemiology, Erasmus MC, Rotterdam, The Netherlands
- Netherlands Genomics Initiative (NGI)–sponsored Netherlands Consortium for Healthy Aging (NCHA), Rotterdam, The Netherlands
| | - Cinzia Sala
- Division of Genetics and Cell Biology, San Raffaele Research Institute, Milano, Italy
| | - Mona M. Sabra
- Memorial Sloan Kettering Cancer Center, Medicine-Endocrinology, New York, New York, United States of America
| | - Naveed Sattar
- BHF Glasgow Cardiovascular Research Centre, Faculty of Medicine, Glasgow, United Kingdom
| | - Beverley M. Shields
- Peninsula NIHR Clinical Research Facility, Peninsula College of Medicine and Dentistry, University of Exeter, Exeter, United Kingdom
| | - Nicole Soranzo
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Cambridge, United Kingdom
| | - John M. Starr
- Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, Edinburgh, United Kingdom
- Alzheimer Scotland Dementia Research Centre, University of Edinburgh, Edinburgh, United Kingdom
| | - David J. Stott
- Academic Section of Geriatric Medicine, Faculty of Medicine, University of Glasgow, Glasgow, United Kingdom
| | - Fred C. G. J. Sweep
- Department of Laboratory Medicine, Radboud University Nijmegen Medical Center, Nijmegen, The Netherlands
| | - Gianluca Usala
- Istituto di Ricerca Genetica e Biomedica (IRGB), Consiglio Nazionale delle Ricerche, c/o Cittadella Universitaria di Monserrato, Monserrato, Cagliari, Italy
| | - Melanie M. van der Klauw
- LifeLines Cohort Study, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
- Department of Endocrinology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Diana van Heemst
- Leiden University Medical Center, Gerontology and Geriatrics, Leiden, The Netherlands
| | - Alies van Mullem
- Department of Internal Medicine, Erasmus MC, Rotterdam, The Netherlands
| | - Sita H.Vermeulen
- Department for Health Evidence, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - W. Edward Visser
- Department of Internal Medicine, Erasmus MC, Rotterdam, The Netherlands
| | - John P. Walsh
- Department of Endocrinology and Diabetes, Sir Charles Gairdner Hospital, Nedlands, Western Australia, Australia
- School of Medicine and Pharmacology, University of Western Australia, Crawley, Western Australia, Australia
| | - Rudi G. J. Westendorp
- Leiden University Medical Center, Gerontology and Geriatrics, Leiden, The Netherlands
| | - Elisabeth Widen
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
| | - Guangju Zhai
- Department of Twin Research and Genetic Epidemiology, King's College London, London, United Kingdom
- Discipline of Genetics, Faculty of Medicine, Memorial University of Newfoundland, St. Johns, Newfoundland, Canada
| | - Francesco Cucca
- Istituto di Ricerca Genetica e Biomedica (IRGB), Consiglio Nazionale delle Ricerche, c/o Cittadella Universitaria di Monserrato, Monserrato, Cagliari, Italy
- Dipartimento di Scienze Biomediche, Università di Sassari, Sassari, Italy
| | - Ian J. Deary
- Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, Edinburgh, United Kingdom
- Department of Psychology, University of Edinburgh, Edinburgh, United Kingdom
| | - Johan G. Eriksson
- Department of Chronic Disease Prevention, National Institute for Health and Welfare, Helsinki, Finland
- Department of General Practice and Primary Health Care, University of Helsinki, Helsinki, Finland
- Helsinki University Central Hospital, Unit of General Practice, Helsinki, Finland
- Folkhalsan Research Centre, Helsinki, Finland
- Vasa Central Hospital, Vasa, Finland
| | - Luigi Ferrucci
- Clinical Research Branch, National Institute on Aging, Baltimore, Maryland, United States of America
| | - Caroline S. Fox
- Division of Intramural Research, National Heart, Lung, and Blood Institute, Framingham, Massachusetts, United States of America
- Division of Endocrinology, Hypertension, and Metabolism, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
| | - J. Wouter Jukema
- Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands
- Durrer Center for Cardiogenetic Research, Amsterdam, The Netherlands
- Interuniversity Cardiology Institute of the Netherlands, Utrecht, The Netherlands
| | - Lambertus A. Kiemeney
- Department for Health Evidence, Radboud University Medical Centre, Nijmegen, The Netherlands
- Department of Urology, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Peter P. Pramstaller
- Center for Biomedicine, European Academy Bozen/Bolzano (EURAC), Bolzano, Italy (Affiliated Institute of the University of Lübeck, Lübeck, Germany)
- Department of Neurology, General Central Hospital, Bolzano, Italy
- Department of Neurology, University of Lübeck, Lübeck, Germany
| | - David Schlessinger
- Laboratory of Genetics, National Institute on Aging, Baltimore, Maryland, United States of America
| | - Alan R. Shuldiner
- Department of Medicine, University of Maryland Medical School, Baltimore, Maryland, United States of America
- Geriatric Research and Education Clinical Center, Veterans Administration Medical Center, Baltimore, Maryland, United States of America
| | - Eline P. Slagboom
- Leiden University Medical Center, Molecular Epidemiology, Leiden, The Netherlands
- Netherlands Consortium for Healthy Ageing, Leiden, The Netherlands
| | - André G. Uitterlinden
- Department of Internal Medicine, Erasmus MC, Rotterdam, The Netherlands
- Department of Epidemiology, Erasmus MC, Rotterdam, The Netherlands
- Netherlands Genomics Initiative (NGI)–sponsored Netherlands Consortium for Healthy Aging (NCHA), Rotterdam, The Netherlands
| | - Bijay Vaidya
- Diabetes, Endocrinology and Vascular Health Centre, Royal Devon and Exeter NHS Foundation Trust, Exeter, United Kingdom
| | - Theo J. Visser
- Department of Internal Medicine, Erasmus MC, Rotterdam, The Netherlands
| | - Bruce H. R. Wolffenbuttel
- LifeLines Cohort Study, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
- Department of Endocrinology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Ingrid Meulenbelt
- Leiden University Medical Center, Molecular Epidemiology, Leiden, The Netherlands
- Netherlands Consortium for Healthy Ageing, Leiden, The Netherlands
| | - Jerome I. Rotter
- Medical Genetics Institute, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
| | - Tim D. Spector
- Department of Twin Research and Genetic Epidemiology, King's College London, London, United Kingdom
| | - Andrew A. Hicks
- Center for Biomedicine, European Academy Bozen/Bolzano (EURAC), Bolzano, Italy (Affiliated Institute of the University of Lübeck, Lübeck, Germany)
| | - Daniela Toniolo
- Division of Genetics and Cell Biology, San Raffaele Research Institute, Milano, Italy
- Institute of Molecular Genetics–CNR, Pavia, Italy
| | - Serena Sanna
- Istituto di Ricerca Genetica e Biomedica (IRGB), Consiglio Nazionale delle Ricerche, c/o Cittadella Universitaria di Monserrato, Monserrato, Cagliari, Italy
- * E-mail: (S Sanna); (RP Peeters); (S Naitza)
| | - Robin P. Peeters
- Department of Internal Medicine, Erasmus MC, Rotterdam, The Netherlands
- * E-mail: (S Sanna); (RP Peeters); (S Naitza)
| | - Silvia Naitza
- Istituto di Ricerca Genetica e Biomedica (IRGB), Consiglio Nazionale delle Ricerche, c/o Cittadella Universitaria di Monserrato, Monserrato, Cagliari, Italy
- * E-mail: (S Sanna); (RP Peeters); (S Naitza)
| |
Collapse
|
11
|
Aparicio-Gallego G, Blanco M, Figueroa A, García-Campelo R, Valladares-Ayerbes M, Grande-Pulido E, Antón-Aparicio L. New insights into molecular mechanisms of sunitinib-associated side effects. Mol Cancer Ther 2011; 10:2215-23. [PMID: 22161785 DOI: 10.1158/1535-7163.mct-10-1124] [Citation(s) in RCA: 90] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The introduction of targeted therapy represents a major advance in the treatment of tumor progression. Targeted agents are a novel therapeutic approach developed to disrupt different cellular signaling pathways. The tyrosine kinase inhibitor sunitinib specifically blocks multiple tyrosine kinase receptors that are involved in the progression of many tumors. Sunitinib is the current standard of care in first-line treatment of advanced renal cell carcinoma, and it is approved in imatinib-intolerant and imatinib-refractory gastrointestinal stromal tumors. However, it is increasingly evident that sunitinib may display collateral effects on other proteins beyond its main target receptors, eliciting undesirable and unexpected adverse events. A better understanding of the molecular mechanisms underlying these undesirable sunitinib-associated side effects will help physicians to maximize efficacy of sunitinib and minimize adverse events. Here, we focus on new insights into molecular mechanisms that may mediate sunitinib-associated adverse events.
Collapse
Affiliation(s)
- Guadalupe Aparicio-Gallego
- Clinical Oncology Department and Biomedical Research Institute (INIBIC), A Coruña University Hospital, A Coruña, Spain
| | | | | | | | | | | | | |
Collapse
|
12
|
Brown RL. Tyrosine kinase inhibitor-induced hypothyroidism: incidence, etiology, and management. Target Oncol 2011; 6:217-26. [DOI: 10.1007/s11523-011-0197-2] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2011] [Accepted: 10/19/2011] [Indexed: 11/29/2022]
|
13
|
Abstract
It is well known that radiation significantly impacts the morbidity of thyroid cancer and that is why Belarus has the highest incidence of the malignancy. Author describes statistical data, classification of angiogenesis, and typical pathological features of malignant thyroid diseases with regard to the vascular network.
Collapse
Affiliation(s)
- Matvey Vladimir Sprindzuk
- United Institute of Informatics Problems, National Academy of Sciences of Belarus, Minsk. Belarus, 220040, Minsk, Bogdanovicha lane, 112/38. ,
| |
Collapse
|
14
|
Suzuki K, Kimura H, Wu H, Kudo N, Kim WB, Suzuki S, Yoshida A, Caturegli P, Kohn LD. Excess iodide decreases transcription of NIS and VEGF genes in rat FRTL-5 thyroid cells. Biochem Biophys Res Commun 2010; 393:286-90. [PMID: 20132794 DOI: 10.1016/j.bbrc.2010.01.123] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2010] [Accepted: 01/29/2010] [Indexed: 12/20/2022]
Abstract
Although it is well known that an excess of iodide suppresses thyroid function and blood flow in vivo, the underlying molecular mechanisms are not fully known. The functional effect of iodide occurs at multiple steps, which include inhibition of sodium/iodide symporter (NIS) expression, transient block of organification, and inhibition of hormonal release. The vascular effect likely involves suppression of the vascular endothelial growth factor (VEGF) gene. In this report, we show that excess iodide coordinately suppresses the expression of the NIS and VEGF genes in FRTL-5 thyroid cells. We also demonstrate that the mechanism of iodide suppression of NIS gene expression is transcriptional, which is synergized by the addition of thyroglobulin. Based on the findings of reporter gene assays and electrophoretic gel mobility shift analysis, we also report two novel DNA binding proteins that responded specifically to iodide and modulated NIS promoter activity. The results suggest that excess iodide affects thyroid vascular function in addition to iodide uptake. This study provides additional insights into the mechanism of action of excess iodide on thyroid function.
Collapse
Affiliation(s)
- Koichi Suzuki
- Leprosy Research Center, National Institute of Infectious Diseases, Tokyo, Japan.
| | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Yamazaki K, Tanigawa K, Suzuki K, Yamada E, Yamada T, Takano K, Obara T, Sato K. Iodide-induced chemokines and genes related to immunological function in cultured human thyroid follicles in the presence of thyrotropin. Thyroid 2010; 20:67-76. [PMID: 20025541 DOI: 10.1089/thy.2009.0242] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
BACKGROUND It is well known that iodide exacerbates thyroid function in subclinical hypothyroid patients with autoimmune thyroiditis. To investigate the immunological mechanism of iodine-induced thyroid dysfunction, we studied the effect of iodide in cultured human thyroid follicles, which respond to physiological concentrations of human thyrotropin (TSH) (0.3-10 microU/mL) and maintain the Wolff-Chaikoff effect. MATERIALS AND METHODS Thyroid follicles obtained from Graves' patients at subtotal thyroidectomy were precultured in medium containing 0.5% fetal calf serum and 10(-8) M iodide for 5 days, and then cultured with the medium containing bovine TSH (30 microU/mL) and low (10(-8)M) or high (10(-5)M) concentrations of iodide. After 3-72 hours of culture, the effect of iodide on thyroid cell mRNA expression was analyzed by microarray and reverse transcriptase-polymerase chain reaction. RESULTS After 48 hours of culture, iodide nearly doubled the mRNA expression levels of the immunity-associated genes (intercellular adhesion molecule-1, transforming growth factor beta 1-induced protein, early growth response gene 1, guanylate-binding protein 1, and annexin A1) and decreased the mRNA expression of sodium-iodide symporter to less than 20%. Further, the mRNA expression levels of chemokines (CCL2, CXCL8, and CXCL14) increased nearly twofold, whereas their receptors did not show any significant response. Real-time polymerase chain reaction analyses confirmed that iodide increased the mRNA expression levels of these genes in a time- and concentration-dependent manner. Immunohistochemical studies revealed that the chemokines were expressed mainly in the thyroid follicular cells in addition to the immune cells. The iodide-induced increase in CCL2 was greater in thyroid follicles obtained from thyroid gland that had been moderately infiltrated with the immunocompetent cells. CONCLUSION We have demonstrated that iodide stimulates thyroid follicular cells to produce chemokines, particularly CCL2, CXCL8, and CXCL14. These chemokines and intercellular adhesion molecule-1 would attract immunocompetent cells into thyroid gland. These in vitro findings suggest that iodide at high concentrations may induce thyroid dysfunction through not only biochemical but also immunological mechanisms, particularly in patients with autoimmune thyroid disorders.
Collapse
|
16
|
Illouz F, Laboureau-Soares S, Dubois S, Rohmer V, Rodien P. Tyrosine kinase inhibitors and modifications of thyroid function tests: a review. Eur J Endocrinol 2009; 160:331-6. [PMID: 19103722 DOI: 10.1530/eje-08-0648] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Tyrosine kinase inhibitors (TKI) belong to new molecular multi-targeted therapies that are approved for the treatment of haematological and solid tumours. They interact with a large variety of protein tyrosine kinases involved in oncogenesis. In 2005, the first case of hypothyroidism was described and since then, some data have been published and have confirmed that TKI can affect the thyroid function tests (TFT). This review analyses the present clinical and fundamental findings about the effects of TKI on the thyroid function. Various hypotheses have been proposed to explain the effect of TKI on the thyroid function but those are mainly based on clinical observations. Moreover, it appears that TKI could alter the thyroid hormone regulation by mechanisms that are specific to each molecule. The present propositions for the management of TKI-induced hypothyroidism suggest that we assess the TFT of the patients regularly before and during the treatment by TKI. Thus, a better approach of patients with TKI-induced hypothyroidism could improve their quality of life.
Collapse
Affiliation(s)
- Frédéric Illouz
- CHU d'Angers, Département d'Endocrinologie Diabétologie Nutrition, Angers Cedex, France.
| | | | | | | | | |
Collapse
|
17
|
Opitz R, Schmidt F, Braunbeck T, Wuertz S, Kloas W. Perchlorate and ethylenethiourea induce different histological and molecular alterations in a non-mammalian vertebrate model of thyroid goitrogenesis. Mol Cell Endocrinol 2009; 298:101-14. [PMID: 18801409 DOI: 10.1016/j.mce.2008.08.020] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2008] [Accepted: 08/19/2008] [Indexed: 11/20/2022]
Abstract
Despite evidence for a conserved role of thyroid-stimulating hormone (TSH) in regulating vertebrate thyroid function, molecular data on thyroid responses to TSH are mainly limited to mammalian species. In this study, we examined histological and molecular changes in the thyroid of Xenopus laevis tadpoles during a 12-day treatment with 20mg/l perchlorate (PER) and 50mg/l ethylenethiourea (ETU). Inhibition of thyroid hormone (TH) synthesis by PER and ETU was evident from developmental retardation, reduced expression of TH-regulated genes and up-regulation of tshb-A mRNA. Thyroid histopathology revealed goiters with strikingly different follicular morphologies following PER and ETU treatment. Using real-time PCR, we analyzed thyroids sampled on day 12 for differential expression of 60 candidate genes. Further temporal analyses were performed for a subset of 14 genes. Relative to the control, PER and ETU treatment modulated the expression of 51 and 49 transcripts, respectively. Particularly genes related to TH synthesis and protein metabolism were similarly affected by PER and ETU. However, several genes were differentially expressed in PER- and ETU-treated tadpoles. Specifically, goiter formation in the PER treatment was associated with low expression of genes related to DNA replication but high expression of negative growth regulators. Results from this work provide for the first time a characterization of gene expression profiles during goitrogenesis in a non-mammalian vertebrate model. Overall, our data suggest that, in addition to TSH over-stimulation, further mechanisms related to the mode of goitrogen action contribute to the regulation of thyroid gene expression.
Collapse
Affiliation(s)
- R Opitz
- Department of Inland Fisheries, Leibniz-Institute of Freshwater Ecology and Inland Fisheries, Berlin, Germany.
| | | | | | | | | |
Collapse
|
18
|
Nakajima K, Umino KI, Azuma Y, Kosaka S, Takano K, Obara T, Sato K. Stimulating parathyroid cell proliferation and PTH release with phosphate in organ cultures obtained from patients with primary and secondary hyperparathyroidism for a prolonged period. J Bone Miner Metab 2009; 27:224-33. [PMID: 19194773 DOI: 10.1007/s00774-008-0032-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2008] [Accepted: 07/08/2008] [Indexed: 10/21/2022]
Abstract
The pathogenesis of primary hyperparathyroidism (I degrees -HPT) and secondary hyperparathyroidism (II degrees -HPT) remains to be elucidated. To characterize their pathophysiology, we investigated the effects of calcium and phosphate on cell proliferation and PTH release in an organ culture of parathyroid tissues. Dissected parathyroid tissues obtained from patients with I degrees -HPT (adenoma) or II degrees -HPT (nodular hyperplasia) were precultured on a collagen-coated membrane for 1-4 week. After changing the medium for one containing various concentrations of phosphate, PTH release and [(3)H]thymidine incorporation were studied. In contrast to dispersed parathyroid cells cultured in a monolayer, calcium decreased PTH release in a concentration-dependent manner in parathyroid tissues. Furthermore, when parathyroid tissues obtained from II degrees -HPT were precultured for 1-4 weeks, PTH release and parathyroid cell proliferation were significantly increased in high-phosphate medium. These phosphate effects were also observed to a lesser extent in parathyroid tissues obtained from I degrees -HPT, but there was no significant difference between I degrees -HPT and II degrees -HPT. Microarray analyses revealed that mRNA levels of PTH, CaSR, and VDR were well preserved, and several growth factors (e.g. TGF-beta1-induced protein) were abundantly expressed in II degrees -HPT. Using organ cultures of hyperparathyroid tissues, in which PTH release and CaSR are well preserved for a prolonged period, we have demonstrated that phosphate stimulates parathyroid cell proliferation not only in II degrees -HPT but also in I degrees -HPT. Although the mechanism responsible for phosphate-induced cell proliferation remains to be elucidated, our in vitro findings suggest that both parathyroid tissues preserve to some extent a physiological response system to hyperphosphatemia as observed in normal parathyroid cells.
Collapse
Affiliation(s)
- Kishiko Nakajima
- Department of Medicine, Institute of Clinical Endocrinology, Tokyo Women's Medical University, Kawada-cho 8-1, Shinjuku-ku, Tokyo 162-8666, Japan
| | | | | | | | | | | | | |
Collapse
|
19
|
Yabuta T, Ito Y, Hirokawa M, Fukushima M, Inoue H, Tomoda C, Higashiyama T, Kihara M, Uruno T, Takamura Y, Kobayashi K, Miya A, Matsuzuka F, Miyauchi A. Preoperative administration of excess iodide increases thyroid volume of patients with Graves' disease. Endocr J 2009; 56:371-5. [PMID: 19139595 DOI: 10.1507/endocrj.k08e-240] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Preoperative excess iodide administration for patients with Graves' disease has been widely adopted by surgeons to perform surgery safely, because it decreases blood flow in the thyroid. However, surgeons often encounter the enlargement of thyroid volume after iodide administration, which makes surgery even more difficult. In this study, we retrospectively investigated the change in thyroid volume in Graves' disease that was evaluated on ultrasonography between before and after iodide administration. Eighty-nine patients who received iodide administered (KI(+) patients) and 24 in whom iodide was not administrated (KI(-) patients) before surgery for Graves' disease were enrolled in the study. The level of free T4 (FT4) significantly decreased and that of thyroid stimulating hormone (TSH) significantly increased after iodide administration. Average thyroid volume also significantly increased for KI(+) patients after iodide administration and 17% of these patients showed a 30% or more increase in thyroid volume. In KI(-) patients who were preoperatively treated only by anti-thyroid drugs, thyroid volume did not change before surgery. Preoperative TSH levels remained below measurement sensitivity in 37 of KI(+) patients, but the average thyroid volume also significantly increased after iodide administration. These findings suggest that thyroid volume in Graves' disease can increase with iodide administration not only due to TSH stimulation but also due to reasons other than TSH. Surgeons should be careful when preoperatively iodide administering to patients with Graves' disease, especially when the goiter is large.
Collapse
|
20
|
Onoda N, Kato Y, Seki T, Kurimoto M, Takano K, Ito Y, Sato K. Increased thyroid blood flow in the hypoechoic lesions in patients with recurrent, painful Hashimoto's thyroiditis at the time of acute exacerbation. Endocr J 2009; 56:65-72. [PMID: 18840927 DOI: 10.1507/endocrj.k08e-194] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
We report two cases with painful Hashimoto's thyroiditis, who developed recurrent fever and painful thyroid. Glucocorticoid treatment was transiently successful but tenderness in the thyroid gland and fever developed when glucocorticoid was tapered. One patient underwent total thyroidectomy uneventfully. As is well known, it is frequently difficult to make differential diagnosis between painful Hashimoto's thyroiditis and subacute thyroiditis particularly at the initial phase. Interestingly, color flow doppler sonography of patient 1 revealed an increased thyroid blood flow in the hypoechoic lesions at the time of acute exacerbation although the serum level of TSH was suppressed. In the other patient, thyroid blood flow was also increased mainly in the hypoechoic lesions when the serum level of TSH was moderately increased, and it disappeared completely after supplementation of prednisolone and L-T4. Since thyroid blood flow in subacute thyroiditis is always decreased, such an increased blood flow in the hypoechoic lesion may be one of clinical characteristics of painful Hashimoto's thyroiditis, and useful for differential diagnosis from subacute thyroiditis.
Collapse
Affiliation(s)
- Noritaka Onoda
- Department of Medicine, Institute of Clinical Endocrinology, Tokyo Women's Medical University, Japan
| | | | | | | | | | | | | |
Collapse
|
21
|
Hassan I, Danila R, Aljabri H, Hoffmann S, Wunderlich A, Karakas E, Zielke A. Is rapid preparation for thyroidectomy in severe Graves' disease beneficial? The relationship between clinical and immunohistochemical aspects. Endocrine 2008; 33:189-95. [PMID: 18493879 DOI: 10.1007/s12020-008-9076-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2008] [Revised: 04/10/2008] [Accepted: 04/25/2008] [Indexed: 10/22/2022]
Abstract
BACKGROUND To evaluate the effect of Lugol's Iodine (LI) on angiogenic characteristics and postthyroidectomy morbidity in patients with Graves' Disease (GD). METHODS Medical records of 153 patients, who underwent thyroidectomies for GD between 1987 and 2006 were reviewed. LI in addition to antithyroid drugs (ATD) was administered to 16 toxic Graves' cases (group I), while 137 received ATD only (group II). In order to evaluate thyroid vascularity, immunohistochemical staining (IHS) for VEGF and CD31 for assessing microvessel density (MVD) were performed in group I, 20 random cases of group II, and 7 stochastic patients with non-toxic goiter (group III). RESULTS Early postoperative complications were significantly higher in group I compared to group II: (transient recurrent nerve palsy (P < 0.04), transient hypocalcemia (P < 0.02), and postoperative bleeding (P < 0.003)). Meanwhile permanent complications were not significantly different. VEGF-IHS revealed ten patients of group I (62.5%) to have positive staining compared to only 3 (15%) of group II. All patients of group III stained positive. The expression of VEGF was more intense in group I as compared to group II (P < 0.02). However, no significant difference of MVD among all groups was recorded. CONCLUSION Preoperative LI treatment may be associated with increased early postoperative morbidity and did not affect thyroid vascularity.
Collapse
Affiliation(s)
- Iyad Hassan
- Department of Visceral-, Thoracic- and Vascular Surgery, Philipps University of Marburg, 35033, Marburg, Germany.
| | | | | | | | | | | | | |
Collapse
|
22
|
Sato K, Omi Y, Kodama H, Obara T, Yamazaki K, Yamada E, Seki T, Takano K, Shiga T, Kasanuki H. Differential diagnosis and appropriate treatment of four thyrotoxic patients with Graves' disease required to take amiodarone due to life-threatening arrhythmia. Intern Med 2008; 47:757-62. [PMID: 18421194 DOI: 10.2169/internalmedicine.47.0843] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
We report the treatment of four thyrotoxic patients. Two were cases of type I amiodarone-induced thyrotoxicosis (AIT) treated with methimazole. The third Graves' disease patient, who became hypothyroid 25 years after subtotal thyroidectomy, developed type II AIT. Furthermore, one case with heart failure and ventricular tachycardia, who developed an adverse reaction to antithyroid agents and was prescribed amiodarone, underwent total thyroidectomy. The clinical course was uneventful, and the patient is doing well. Since amiodarone contains a large amount of iodine, it is frequently difficult to make a differential diagnosis. Surgical treatment of Graves' disease patients is recommended when immediate control of hyperthyroidism and heart failure is required.
Collapse
Affiliation(s)
- Kanji Sato
- Department of Medicine, Institute of Clinical Endocrinology, Tokyo Women's Medical University, Tokyo.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Yamazaki K, Mitsuhashi T, Yamada E, Yamada T, Kosaka S, Takano K, Obara T, Sato K. Amiodarone reversibly decreases sodium-iodide symporter mRNA expression at therapeutic concentrations and induces antioxidant responses at supraphysiological concentrations in cultured human thyroid follicles. Thyroid 2007; 17:1189-200. [PMID: 18020914 DOI: 10.1089/thy.2007.0215] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
CONTEXT Amiodarone, a potent antiarrhythmic, iodine-containing agent, is a highly active oxidant exerting cytotoxic effects on thyrocytes at pharmacological concentrations. Patients receiving amiodarone usually remain euthyroid, but occasionally develop thyroid dysfunction. Although there is a general consensus that amiodarone-associated hypothyroidism is iodine induced, the destructive mechanism of thyroid follicles in amiodarone-induced thyrotoxicosis remains unknown. OBJECTIVE To elucidate the mechanism by which amiodarone elicits thyroid dysfunction. DESIGN Human thyroid follicles were cultured with thyroid-stimulating hormone (TSH) and amiodarone at therapeutic (1-2 microM) and pharmacological (10-20 microM) concentrations, and the drug-induced effect on whole human gene expression was analyzed by cDNA microarray. Microarray data were confirmed by real-time PCR and Western blot. MAIN OUTCOMES Amiodarone at 1-2 muM decreased the expression level of the sodium-iodide symporter (NIS) to nearly half, but did not affect genes participating in thyroid hormonogenesis (thyroid peroxidase, thyroglobulin, pendrin, and NADPH oxidase). Higher concentrations (10-20 microM) decreased the expression of all these genes, accompanied by increased expression of antioxidant proteins such as heme oxygenase 1 and ferritin. When thyroid follicles obtained from a patient with Graves' disease who had been treated with amiodarone were cultured in amiodarone-free medium, TSH-induced thyroid function was intact, suggesting that amiodarone at a maintenance dose did not elicit any cytotoxic effect on thyrocytes. The ultrastructural features of cultured thyroid follicles were compatible with these in vitro findings. CONCLUSION These in vitro and ex vivo findings suggest that patients taking maintenance doses of amiodarone usually remain euthyroid, probably due to escape from the Wolff-Chaikoff effect mediated by decreased expression of NIS mRNA. Further, amiodarone is not cytotoxic for thyrocytes at therapeutic concentrations but elicits cytotoxicity through oxidant activity at supraphysiological concentrations. We speculate that when amiodarone-induced prooxidant activity somehow exceeds the endogenous antioxidant capacity, the thyroid follicles will be destroyed and amiodarone-induced destructive thyrotoxicosis may develop.
Collapse
|
24
|
Klubo-Gwiezdzinska J, Junik R, Kopczynska E, Juraniec O, Kardymowicz H. The comparison of serum vascular endothelial growth factor levels between patients with metastatic and non-metastatic thyroid cancer, and patients with nontoxic multinodular goiter. Eur J Endocrinol 2007; 157:521-7. [PMID: 17893268 DOI: 10.1530/eje-07-0252] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
BACKGROUND One of the important proangiogenic factors involved in the growth of normal and neoplastic tissues is vascular endothelial growth factor (VEGF). AIM We hypothesized that serum VEGF concentration would differ between patients with metastatic and non-metastatic thyroid cancer, multinodular goiter, and healthy subjects. We also hypothesized that endogenous TSH stimulation would affect serum VEGF level. SUBJECTS AND METHODS The study group consisted of 71 patients (62 females and 9 males), aged 44.9 +/- 12.3 years, with differentiated thyroid cancer (50 papillary, 17 follicular, and 4 oxyphilic), treated in our department during the years 2003-2006. All patients had undergone total or near-total thyroidectomy and radioactive iodine treatment, that had resulted in remission in 59 patients and persistent/recurrent disease in 12 patients. The study included two control groups: 30 patients with nontoxic multinodular goiter and 30 healthy subjects. RESULTS Serum VEGF concentrations were significantly higher in patients with distant metastases than those in remission or healthy patients. (423.4 vs 217.6 vs 235.55 pg/ml respectively, P < 0.05). This was not observed in patients with locoregional metastases. During endogenous TSH stimulation, VEGF decreased significantly (215.3 vs 169.6 pg/ml, P < 0.05). Patients with multinodular goiter showed significantly lower VEGF concentrations than the remaining study groups. CONCLUSIONS Serum VEGF concentration might be used as an additional marker of thyroid cancer with distant metastases, but its interpretation should be undertaken very cautiously. Endogenous TSH stimulation decreases VEGF levels in patients either with or without thyroid tissue, suggesting that its regulatory effects are through receptors located outside the thyrocytes.
Collapse
Affiliation(s)
- Joanna Klubo-Gwiezdzinska
- Department of Endocrinology and Diabetology, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, ul. M. Sklodowskiej-Curie 9, 85-094 Bydgoszcz, Poland.
| | | | | | | | | |
Collapse
|
25
|
Jebreel A, England J, Bedford K, Murphy J, Karsai L, Atkin S. Vascular endothelial growth factor (VEGF), VEGF receptors expression and microvascular density in benign and malignant thyroid diseases. Int J Exp Pathol 2007; 88:271-7. [PMID: 17696908 PMCID: PMC2517322 DOI: 10.1111/j.1365-2613.2007.00533.x] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Angiogenesis is critical for the growth and metastatic spread of tumours. Vascular endothelial growth factor (VEGF) is the most potent inducer of neovasculature, and its increased expression has been related to a worse clinical outcome in many diseases. The purpose of this study was to evaluate the relation between VEGF, its receptors (VEGFR-1 and VEGFR-2) and microvessel density (MVD) in thyroid diseases. Immunostaining for VEGF and VEGF receptors was performed in 66 specimens of thyroid tissue, comprising 17 multinodular goitre (MNG), 14 Graves' disease, 10 follicular adenoma, 8 Hashimoto's thyroiditis, 7 papillary carcinoma and 10 normal thyroid specimens. Thyrocyte positivity for VEGF and VEGF receptors was scored 0-3. Immunohistochemistry for CD31, and CD34 on the same sections was performed to evaluate MVD. Immunohistochemical staining of VEGF in thyrocytes was positive in 92% of all the thyroid tissues studied. Using an immunostaining intensity cut off of 2, increased thyrocyte staining was seen in follicular adenoma specimens, MNG and normal thyroids compared with Hashimoto's thyroiditis and Graves' disease (P < 0.05). Similarly, VEGF thyrocyte expression in Graves' disease was less than other pathologies (P < 0.05). VEGFR-1 expression and the average MVD score did not differ between the different thyroid pathologies. VEGF expression was lower in autoimmune pathologies compared to autonomous growth processes. Conversely, both VEGFR-1 and VEGFR-2 were widely expressed in benign and neoplastic thyroid disease, suggesting that the up-regulation of VEGF and not its receptors occurs as tissue becomes autonomous. There was no clear relationship between MVD measurement and thyroid pathology.
Collapse
|
26
|
Yamazaki K, Suzuki K, Yamada E, Yamada T, Takeshita F, Matsumoto M, Mitsuhashi T, Obara T, Takano K, Sato K. Suppression of iodide uptake and thyroid hormone synthesis with stimulation of the type I interferon system by double-stranded ribonucleic acid in cultured human thyroid follicles. Endocrinology 2007; 148:3226-35. [PMID: 17395700 DOI: 10.1210/en.2006-1638] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Although viral infection is thought to be associated with subacute thyroiditis and probably with autoimmune thyroid disease, possible changes in thyroid function during the prodromal period of infection or subclinical infection remain largely unknown. Recently, it was shown that pathogen-associated molecular patterns stimulate Toll-like receptors (TLR) and activate innate immune responses by producing type I interferons (IFN). Using a human thyroid follicle culture system, in which de novo synthesized thyroid hormones are released into the culture medium under physiological concentrations of human TSH, we studied the effects of polyinosinic-polycytidylic acid [Poly(I:C)], a chemical analog of viral double-stranded RNA (dsRNA), on TSH-induced thyroid function. Thyrocytes expressed ligands for dsRNA (TLR 3, CD14, and retinoic-acid-inducible protein-1) comparable with the TSH receptor. DNA microarray and real-time PCR analyses revealed that dsRNA increased the expression of mRNA for TLR3, IFN-beta, IFN-regulating factors, proinflammatory cytokines, and class I major histocompatibility complex (MHC), whereas genes associated with thyroid hormonogenesis (sodium/iodide symporter, peroxidase, deiodinases) were suppressed. In accordance to these data, Poly(I:C) suppressed TSH-induced 125I uptake and hormone synthesis dose dependently, accompanied by a decrease in the ratio of 125I-T3/125I-T4 released into the culture medium, whereas peptidoglycan, lipopolysaccharides, or unmethylated CpG DNA, ligands for TLR2, TLR4, and TLR9, respectively, had no significant effect. These inhibitory effects of Poly(I:C) were not blocked by a neutralizing antibody against TLR3 and an anti-IFN alpha/beta receptor antibody. These in vitro findings suggest that when thyrocytes are infected with certain viruses, dsRNA formed intracellularly in thyrocytes may be a cause for thyroid dysfunction, leading to development of autoimmune thyroiditis.
Collapse
Affiliation(s)
- Kazuko Yamazaki
- Thyroid Disease Institute, Kanaji Hospital, Tokyo, and Department of Molecular Biodefense Research, Yokohama City University Graduate School of Medicine, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Erbil Y, Ozluk Y, Giriş M, Salmaslioglu A, Issever H, Barbaros U, Kapran Y, Ozarmağan S, Tezelman S. Effect of lugol solution on thyroid gland blood flow and microvessel density in the patients with Graves' disease. J Clin Endocrinol Metab 2007; 92:2182-9. [PMID: 17389702 DOI: 10.1210/jc.2007-0229] [Citation(s) in RCA: 75] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
CONTEXT Although some endocrine surgeons administer Lugol solution to decrease thyroid gland vascularity, there is still not an agreement on its effectiveness. OBJECTIVE The aims of this clinical trial are to evaluate thyroid blood flow and microvessel density in patients with Graves' disease who received Lugol solution treatment preoperatively. DESIGN This was a prospective clinical trial. SETTING This clinical trial took place at a tertiary referral center. METHOD Thirty-six patients were randomly assigned to receive either preoperative treatment with Lugol solution (group 1, n = 17) or no preoperative treatment with Lugol solution (group 2, n = 19). MAIN OUTCOME MEASURES Blood flow through the thyroid arteries of patients with Graves' disease was measured by color flow Doppler ultrasonography. The microvessel density (MVD) was assessed by immunohistochemical and Western blot analysis of the level of expression of CD-34 in thyroid tissue. The weight and blood loss of the thyroid gland were measured in all patients. RESULTS The mean blood flow, MVD, CD-34 expression, and blood loss in group 1 patients were significantly lower than those in group 2 patients. There was a negative correlation between Lugol solution treatment and blood flow (r(s) = -0.629; P = 0.0001), blood loss (r(s) = -0.621; P = 0.0001), MVD (r(s) = -0.865; P = 0.0001), and CD-34 expression (r(s) = -0.865; P = 0.0001). According to logistic regression analysis, Lugol solution treatment resulted in a 9.33-fold decreased rate of intraoperative blood loss. CONCLUSION Preoperative Lugol solution treatment decreased the rate of blood flow, thyroid vascularity, and intraoperative blood loss during thyroidectomy.
Collapse
Affiliation(s)
- Yeşim Erbil
- Department of General Surgery, Istanbul Medical Faculty, Istanbul University, 34093 Capa/Istanbul, Turkey.
| | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Dedecjus M, Kołomecki K, Brzeziński J, Adamczewski Z, Tazbir J, Lewiński A. Influence of L-thyroxine administration on poor-platelet plasma VEGF concentrations in patients with induced short-term hypothyroidism, monitored for thyroid carcinoma. Endocr J 2007; 54:63-9. [PMID: 17090953 DOI: 10.1507/endocrj.k05-112] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Angiogenesis is a process of new blood vessel development from pre-existing vasculature. It is a crucial process in normal physiology, as well as in several pathological conditions. The vascular endothelial growth factor (VEGF) represents a family of specific endothelial cell mitogens, involved in normal angiogenesis and in tumour development. The aim of the present study was to estimate the influence of L-thyroxine (L-T4) administration on poor-platelet plasma (P-PP) VEGF concentrations in patients with induced short-term hypothyroidism, monitored for differentiated thyroid carcinoma. In the present study, P-PP concentrations of VEGF, thyroglobulin, thyrotropin and free thyroid hormones were investigated in a population of 24 hypothyroid patients, who were withdrawn from L-T4 treatment for 5 weeks and studied before and after 2 months of L-T4 therapy. Only healthy female patients with no evidence of metastasis in whole body scintigraphy were included in the study. They were then compared with 20 healthy control subjects, matched for age, sex and body mass index (BMI). The patients had significantly lower plasma VEGF concentrations before treatment with L-T4 than after administration of that hormone. There was no significant difference in plasma VEGF levels, either between the patients treated with L-T4, and the controls, or between the patients untreated with L-T4, and the controls. Even short-time changes in thyrometabolic profile exert an important influence on P-PP VEGF concentrations, even if there is no thyroid tissue.
Collapse
Affiliation(s)
- Marek Dedecjus
- Department of Endocrine Surgery, Medical University of Lodz, Polish Mother's Memorial Hospital-Research Institute
| | | | | | | | | | | |
Collapse
|