1
|
Ji RL, Tao YX. Biased signaling in drug discovery and precision medicine. Pharmacol Ther 2025; 268:108804. [PMID: 39904401 DOI: 10.1016/j.pharmthera.2025.108804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 01/10/2025] [Accepted: 01/21/2025] [Indexed: 02/06/2025]
Abstract
Receptors are crucial for converting chemical and environmental signals into cellular responses, making them prime targets in drug discovery, with about 70% of drugs targeting these receptors. Biased signaling, or functional selectivity, has revolutionized drug development by enabling precise modulation of receptor signaling pathways. This concept is more firmly established in G protein-coupled receptor and has now been applied to other receptor types, including ion channels, receptor tyrosine kinases, and nuclear receptors. Advances in structural biology have further refined our understanding of biased signaling. This targeted approach enhances therapeutic efficacy and potentially reduces side effects. Numerous biased drugs have been developed and approved as therapeutics to treat various diseases, demonstrating their significant therapeutic potential. This review provides a comprehensive overview of biased signaling in drug discovery and disease treatment, highlighting recent advancements and exploring the therapeutic potential of these innovative modulators across various diseases.
Collapse
Affiliation(s)
- Ren-Lei Ji
- Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL 36849, United States.
| | - Ya-Xiong Tao
- Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL 36849, United States.
| |
Collapse
|
2
|
Razavi SA, Kalari M, Haghzad T, Haddadi F, Nasiri S, Hedayati M. Exploring the potential of myo-inositol in thyroid disease management: focus on thyroid cancer diagnosis and therapy. Front Endocrinol (Lausanne) 2024; 15:1418956. [PMID: 39329107 PMCID: PMC11424451 DOI: 10.3389/fendo.2024.1418956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Accepted: 08/05/2024] [Indexed: 09/28/2024] Open
Abstract
Thyroid cancer (TC) is a malignancy that is increasing in prevalence on a global scale, necessitating the development of innovative approaches for both diagnosis and treatment. Myo-inositol (MI) plays a crucial role in a wide range of physiological and pathological functions within human cells. To date, studies have investigated the function of MI in thyroid physiology as well as its potential therapeutic benefits for hypothyroidism and autoimmune thyroiditis. However, research in the field of TC is very restricted. Metabolomics studies have highlighted the promising diagnostic capabilities of MI, recognizing it as a metabolic biomarker for identifying thyroid tumors. Furthermore, MI can influence therapeutic characteristics by modulating key cellular pathways involved in TC. This review evaluates the potential application of MI as a naturally occurring compound in the management of thyroid diseases, including hypothyroidism, autoimmune thyroiditis, and especially TC. The limited number of studies conducted in the field of TC emphasizes the critical need for future research to comprehend the multifaceted role of MI in TC. A significant amount of research and clinical trials is necessary to understand the role of MI in the pathology of TC, its diagnostic and therapeutic potential, and to pave the way for personalized medicine strategies in managing this intricate disease.
Collapse
Affiliation(s)
- S. Adeleh Razavi
- Cellular and Molecular Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohadeseh Kalari
- Department of Biochemistry, Semnan University of Medical Sciences, Semnan, Iran
| | - Tahereh Haghzad
- Department of Biology, Faculty of Sciences, University of Guilan, Rasht, Iran
| | - Fatemeh Haddadi
- Medicinal Plants and Drugs Research Institute, Shahid Beheshti University, Tehran, Iran
| | - Shirzad Nasiri
- Department of Surgery, Shariati Hospital, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mehdi Hedayati
- Cellular and Molecular Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
3
|
Yavari M, Akbari M, Ramezani Ahmadi A, Siavash Dastjerdi M, Hashemi MS. Investigating the effect of combined use of selenium and Myo-inositol supplements on thyroid function and autoimmune characteristics in thyroid disorders: a systematic review and meta-analysis. Expert Rev Endocrinol Metab 2024; 19:269-277. [PMID: 38147023 DOI: 10.1080/17446651.2023.2295487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Accepted: 11/13/2023] [Indexed: 12/27/2023]
Abstract
BACKGROUND This study aimed to systematically review the effect of selenium and inositol combination on thyroid function, autoimmune characteristics in thyroid diseases. RESEARCH DESIGN AND METHODS To identify eligible studies, a systematic search was conducted in the PubMed/MEDLINE, Science-Direct, CINHAL, EMBASE, SCOPUS, Psychinfo, Cochrane, ProQuest, and Web of Science were searched using the main concepts, and all English-written articles that were published between 2007 and 2022 and had an available full text were examined. RESULTS The data analysis of this research revealed that after the simultaneous use of selenium and inositol supplements, the level of Triiodothyronine(T3) increased by 0.105 in patients with thyroid disorders although this increase was not significant (P-value: 0.228). The level of Thyroxine (T4) significantly increased by 0.06 (P-value: 0.04). Anti-Thyroid Peroxidase Antibody (TPOAb) titer decreased by 119.36%, which was not significant (P-value: 0.070). Finally, the level of Thyroid-stimulating hormone (TSH) decreased by 1.45%, which was a significant change (P-value: 0.001). CONCLUSION It was observed that simultaneous use of selenium and inositol supplements did not change the T3 and TPOAb titer levels; however, it leads to a decrease in TSH and increase in T4 levels. Further studies are required due to the limited number of studies.
Collapse
Affiliation(s)
- Maryam Yavari
- Isfahan Endocrine and Metabolism Research Centre (IEMRC), Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mojtaba Akbari
- Isfahan Endocrine and Metabolism Research Centre (IEMRC), Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Amirhossein Ramezani Ahmadi
- Isfahan Endocrine and Metabolism Research Centre (IEMRC), Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mansour Siavash Dastjerdi
- Endocrinology & Metabolism, Isfahan Endocrine and Metabolism Research Center (IEMRC), Isfahan University of Medical Sciences, Isfahan, Iran
| | - Maryam Sadat Hashemi
- Nursing & Midwifery Care Research Center, Department of critical care, Faculty of Nursing and Midwifery, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
4
|
Makkonen K, Jännäri M, Crisóstomo L, Kuusi M, Patyra K, Melnyk V, Linnossuo V, Ojala J, Ravi R, Löf C, Mäkelä JA, Miettinen P, Laakso S, Ojaniemi M, Jääskeläinen J, Laakso M, Bossowski F, Sawicka B, Stożek K, Bossowski A, Kleinau G, Scheerer P, FinnGen F, Reeve MP, Kero J. Mechanisms of thyrotropin receptor-mediated phenotype variability deciphered by gene mutations and M453T-knockin model. JCI Insight 2024; 9:e167092. [PMID: 38194289 PMCID: PMC11143923 DOI: 10.1172/jci.insight.167092] [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: 11/10/2022] [Accepted: 01/05/2024] [Indexed: 01/10/2024] Open
Abstract
The clinical spectrum of thyrotropin receptor-mediated (TSHR-mediated) diseases varies from loss-of-function mutations causing congenital hypothyroidism to constitutively active mutations (CAMs) leading to nonautoimmune hyperthyroidism (NAH). Variation at the TSHR locus has also been associated with altered lipid and bone metabolism and autoimmune thyroid diseases. However, the extrathyroidal roles of TSHR and the mechanisms underlying phenotypic variability among TSHR-mediated diseases remain unclear. Here we identified and characterized TSHR variants and factors involved in phenotypic variability in different patient cohorts, the FinnGen database, and a mouse model. TSHR CAMs were found in all 16 patients with NAH, with 1 CAM in an unexpected location in the extracellular leucine-rich repeat domain (p.S237N) and another in the transmembrane domain (p.I640V) in 2 families with distinct hyperthyroid phenotypes. In addition, screening of the FinnGen database revealed rare functional variants as well as distinct common noncoding TSHR SNPs significantly associated with thyroid phenotypes, but there was no other significant association between TSHR variants and more than 2,000 nonthyroid disease endpoints. Finally, our TSHR M453T-knockin model revealed that the phenotype was dependent on the mutation's signaling properties and was ameliorated by increased iodine intake. In summary, our data show that TSHR-mediated disease risk can be modified by variants at the TSHR locus both inside and outside the coding region as well as by altered TSHR-signaling and dietary iodine, supporting the need for personalized treatment strategies.
Collapse
Affiliation(s)
- Kristiina Makkonen
- Department of Clinical Sciences, Faculty of Medicine, and
- Integrative Physiology and Pharmacology, Institute of Biomedicine, University of Turku, Turku, Finland
| | - Meeri Jännäri
- Department of Clinical Sciences, Faculty of Medicine, and
- Integrative Physiology and Pharmacology, Institute of Biomedicine, University of Turku, Turku, Finland
| | - Luís Crisóstomo
- Integrative Physiology and Pharmacology, Institute of Biomedicine, University of Turku, Turku, Finland
| | - Matilda Kuusi
- Integrative Physiology and Pharmacology, Institute of Biomedicine, University of Turku, Turku, Finland
| | - Konrad Patyra
- Department of Clinical Sciences, Faculty of Medicine, and
- Integrative Physiology and Pharmacology, Institute of Biomedicine, University of Turku, Turku, Finland
| | | | - Veli Linnossuo
- Department of Clinical Sciences, Faculty of Medicine, and
| | - Johanna Ojala
- Department of Clinical Sciences, Faculty of Medicine, and
| | - Rowmika Ravi
- Department of Clinical Sciences, Faculty of Medicine, and
| | - Christoffer Löf
- Integrative Physiology and Pharmacology, Institute of Biomedicine, University of Turku, Turku, Finland
| | - Juho-Antti Mäkelä
- Integrative Physiology and Pharmacology, Institute of Biomedicine, University of Turku, Turku, Finland
| | - Päivi Miettinen
- New Children’s Hospital, Helsinki University Hospital, Helsinki, Finland
| | - Saila Laakso
- New Children’s Hospital, Helsinki University Hospital, Helsinki, Finland
| | - Marja Ojaniemi
- Department of Pediatrics and Adolescence, PEDEGO Research Unit and Medical Research Center, University and University Hospital of Oulu, Oulu, Finland
| | | | - Markku Laakso
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland, Kuopio, Finland
| | - Filip Bossowski
- Department of Pediatrics, Endocrinology, Diabetes with a Cardiology Unit, Medical University in Białystok, Bialystok, Poland
| | - Beata Sawicka
- Department of Pediatrics, Endocrinology, Diabetes with a Cardiology Unit, Medical University in Białystok, Bialystok, Poland
| | - Karolina Stożek
- Department of Pediatrics, Endocrinology, Diabetes with a Cardiology Unit, Medical University in Białystok, Bialystok, Poland
| | - Artur Bossowski
- Department of Pediatrics, Endocrinology, Diabetes with a Cardiology Unit, Medical University in Białystok, Bialystok, Poland
| | - Gunnar Kleinau
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, and
- Humboldt - Universität zu Berlin, Institute of Medical Physics, Biophysics, Group Structural Biology of Cellular Signaling, Berlin, Germany
| | - Patrick Scheerer
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, and
- Humboldt - Universität zu Berlin, Institute of Medical Physics, Biophysics, Group Structural Biology of Cellular Signaling, Berlin, Germany
| | - FinnGen FinnGen
- Institute for Molecular Medicine Finland, HiLIFE, University of Helsinki, Helsinki, Finland
- FinnGen is detailed in Supplemental Acknowledgments
| | - Mary Pat Reeve
- Institute for Molecular Medicine Finland, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Jukka Kero
- Department of Clinical Sciences, Faculty of Medicine, and
- Department of Pediatrics and Adolescent Medicine, Turku University Hospital, Turku, Finland
| |
Collapse
|
5
|
Yang LK, Zhang J, Liu D, Han TY, Qin QS, Wang AQ, Dong B. Ancestral glycoprotein hormone and its cognate receptor present in primitive chordate ascidian: Molecular identification and functional characterization. Int J Biol Macromol 2023; 229:401-412. [PMID: 36592853 DOI: 10.1016/j.ijbiomac.2022.12.297] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 12/26/2022] [Indexed: 12/31/2022]
Abstract
The glycoprotein hormone (GPH) system is fundamentally significant in regulating the physiology of chordates, such as thyroid activity and gonadal function. However, the knowledge of the GPH system in the primitive chordate ascidian species is largely lacking. Here, we reported an ancestral GPH system in the ascidian (Styela clava), which consists of GPH α subunit (Sc-GPA2), GPH β subunit (Sc-GPB5), and the cognate leucine-rich repeat-containing G protein-coupled receptor (Sc-GPHR). Comparative structure analysis revealed that distinct from vertebrate GPH β subunits, Sc-GPB5 was less conserved, showing an atypical N-terminal sequence with a type II transmembrane domain instead of a typical signal peptide. By investigating the presence of recombinant Sc-GPA2 and Sc-GPB5 in cell lysates and culture media of HEK293T cells, we confirmed that these two subunits could be secreted out of the cells via distinct secretory pathways. The deglycosylation experiments demonstrated that N-linked glycosylation only occurred on the conserved cysteine residue (N78) of Sc-GPA2, whereas Sc-GPB5 was non-glycosylated. Although Sc-GPB5 exhibited distinct topology and biochemical properties in contrast to its chordate counterparts, it could still interact with Sc-GPA2 to form a heterodimer. The Sc-GPHR was then confirmed to be activated by tethered Sc-GPA2/GPB5 heterodimer on the Gs-cAMP pathway, suggesting that Sc-GPA2/GPB5 heterodimer-initiated Gs-cAMP signaling pathway is evolutionarily conserved in chordates. Furthermore, in situ hybridization and RT-PCR results revealed the co-expression patterns of Sc-GPA2 and Sc-GPB5 with Sc-GPHR transcripts, respectively in ascidian larvae and adults, highlighting the potential functions of Sc-GPA2/GPB5 heterodimer as an autocrine/paracrine neurohormone in regulating metamorphosis of larvae and physiological functions of adults. Our study systematically investigated the GPA2/GPB5-GPHR system in ascidian for the first time, which offers insights into understanding the function and evolution of the GPH system within the chordate lineage.
Collapse
Affiliation(s)
- Li-Kun Yang
- Fang Zongxi Centre, MoE Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China
| | - Jin Zhang
- Fang Zongxi Centre, MoE Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China
| | - Di Liu
- Fang Zongxi Centre, MoE Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China
| | - Tong-Ye Han
- Fang Zongxi Centre, MoE Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China
| | - Qi-Shu Qin
- Fang Zongxi Centre, MoE Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China
| | - An-Qi Wang
- Haide College, Ocean University of China, Qingdao 266100, China
| | - Bo Dong
- Fang Zongxi Centre, MoE Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China; Laoshan Laboratory, Qingdao 266237, China; Institute of Evolution & Marine Biodiversity, Ocean University of China, Qingdao 266003, China.
| |
Collapse
|
6
|
Plouffe B, Karamitri A, Flock T, Gallion JM, Houston S, Daly CA, Bonnefond A, Guillaume JL, Le Gouill C, Froguel P, Lichtarge O, Deupi X, Jockers R, Bouvier M. Structural Elements Directing G Proteins and β-Arrestin Interactions with the Human Melatonin Type 2 Receptor Revealed by Natural Variants. ACS Pharmacol Transl Sci 2022; 5:89-101. [PMID: 35846981 PMCID: PMC9281605 DOI: 10.1021/acsptsci.1c00239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
G protein-coupled receptors (GPCRs) can engage distinct subsets of signaling pathways, but the structural determinants of this functional selectivity remain elusive. The naturally occurring genetic variants of GPCRs, selectively affecting different pathways, offer an opportunity to explore this phenomenon. We previously identified 40 coding variants of the MTNR1B gene encoding the melatonin MT2 receptor (MT2). These mutations differently impact the β-arrestin 2 recruitment, ERK activation, cAMP production, and Gαi1 and Gαz activation. In this study, we combined functional clustering and structural modeling to delineate the molecular features controlling the MT2 functional selectivity. Using non-negative matrix factorization, we analyzed the signaling signatures of the 40 MT2 variants yielding eight clusters defined by unique signaling features and localized in distinct domains of MT2. Using computational homology modeling, we describe how specific mutations can selectively affect the subsets of signaling pathways and offer a proof of principle that natural variants can be used to explore and understand the GPCR functional selectivity.
Collapse
Affiliation(s)
- Bianca Plouffe
- Department
of Biochemistry and Molecular Medicine, Université de Montréal, H3T 1J4 Montréal, Québec, Canada,Institute
for Research in Immunology and Cancer, Université
de Montréal, H3T 1J4 Montréal, Québec, Canada,The Wellcome-Wolfson
Institute for Experimental Medicine, Queen’s
University Belfast, BT9 7BL Belfast, U.K.
| | - Angeliki Karamitri
- Université
de Paris, Institut Cochin, INSERM, CNRS, F-75014 Paris, France
| | - Tilman Flock
- Laboratory
of Biomolecular Research, Department of Biology and Chemistry, Paul Scherrer Institute, 5232 Villigen, Switzerland,Department
of Biology, ETH Zürich, 8093 Zürich, Switzerland
| | - Jonathan M. Gallion
- Program
in Structural and Computational Biology and Molecular Biophysics, Baylor College of Medicine, 77030 Houston, Texas, United States
| | - Shane Houston
- The Wellcome-Wolfson
Institute for Experimental Medicine, Queen’s
University Belfast, BT9 7BL Belfast, U.K.
| | - Carole A. Daly
- The Wellcome-Wolfson
Institute for Experimental Medicine, Queen’s
University Belfast, BT9 7BL Belfast, U.K.
| | - Amélie Bonnefond
- Université
de Lille, INSERM/CNRS UMR 1283/8199—EGID, Institut Pasteur
de Lille, CHU de Lille, 59045 Lille, France
| | - Jean-Luc Guillaume
- Université
de Paris, Institut Cochin, INSERM, CNRS, F-75014 Paris, France
| | - Christian Le Gouill
- Institute
for Research in Immunology and Cancer, Université
de Montréal, H3T 1J4 Montréal, Québec, Canada
| | - Phillipe Froguel
- Université
de Lille, INSERM/CNRS UMR 1283/8199—EGID, Institut Pasteur
de Lille, CHU de Lille, 59045 Lille, France
| | - Olivier Lichtarge
- Program
in Structural and Computational Biology and Molecular Biophysics, Baylor College of Medicine, 77030 Houston, Texas, United States,Department
of Molecular and Human Genetics, Baylor
College of Medicine, 77030 Houston, Texas, United States
| | - Xavier Deupi
- Laboratory
of Biomolecular Research, Department of Biology and Chemistry, Paul Scherrer Institute, 5232 Villigen, Switzerland,Condensed
Matter Theory Group, Division of Scientific Computing, Theory, and
Data, Paul Scherrer Institute, 5232 Villigen, Switzerland,. Phone: +41-563103337
| | - Ralf Jockers
- Université
de Paris, Institut Cochin, INSERM, CNRS, F-75014 Paris, France,. Phone: +33-140516434
| | - Michel Bouvier
- Department
of Biochemistry and Molecular Medicine, Université de Montréal, H3T 1J4 Montréal, Québec, Canada,Institute
for Research in Immunology and Cancer, Université
de Montréal, H3T 1J4 Montréal, Québec, Canada,. Phone: 1-514-343-6319
| |
Collapse
|
7
|
The Influence of Oxidative Stress on Thyroid Diseases. Antioxidants (Basel) 2021; 10:antiox10091442. [PMID: 34573074 PMCID: PMC8465820 DOI: 10.3390/antiox10091442] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 09/04/2021] [Accepted: 09/06/2021] [Indexed: 01/19/2023] Open
Abstract
Thyroid diseases, including neoplasms, autoimmune diseases and thyroid dysfunctions, are becoming a serious social problem with rapidly increasing prevalence. The latter is increasingly linked to oxidative stress. There are many methods for determining the biomarkers of oxidative stress, making it possible to evaluate the oxidative profile in patients with thyroid diseases compared to the healthy population. This opens up a new perspective for investigating the role of elevated parameters of oxidative stress and damage in people with thyroid diseases, especially of neoplastic nature. An imbalance between oxidants and antioxidants is observed at different stages and in different types of thyroid diseases. The organ, which is part of the endocrine system, uses free radicals (reactive oxygen species, ROS) to produce hormones. Thyroid cells release enzymes that catalyse ROS generation; therefore, a key role is played by the internal defence system and non-enzymatic antioxidants that counteract excess ROS not utilised to produce thyroid hormones, acting as a buffer to neutralise free radicals and ensure whole-body homeostasis. An excess of free radicals causes structural cell damage, undermining genomic stability. Looking at the negative effects of ROS accumulation, oxidative stress appears to be implicated in both the initiation and progression of carcinogenesis. The aim of this review is to investigate the oxidation background of thyroid diseases and to summarise the links between redox imbalance and thyroid dysfunction and disease.
Collapse
|
8
|
Piras C, Pibiri M, Leoni VP, Balsamo A, Tronci L, Arisci N, Mariotti S, Atzori L. Analysis of metabolomics profile in hypothyroid patients before and after thyroid hormone replacement. J Endocrinol Invest 2021; 44:1309-1319. [PMID: 33025552 DOI: 10.1007/s40618-020-01434-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Accepted: 09/24/2020] [Indexed: 12/12/2022]
Abstract
PURPOSE The serum metabolic changes occurring during the transition from hypothyroidism to euthyroidism are not known. This study aimed to determine the metabolomic profile in hypothyroid patients before (HypoT0) and after (HypoT1) euthyroidism achieved through levothyroxine (L-T4) treatment. METHODS Eighteen patients with overt primary hypothyroidism were recruited for the study. All patients were treated with L-T4 to achieve euthyroidism. Thyrotropin (TSH), free thyroxine (FT4), free triiodothyronine (FT3) and metabolomics profiles were measured before and after 3 months of treatment. The euthyroid control group consisted of 28 healthy volunteers. Metabolomics analysis was performed using Nuclear Magnetic Resonance (NMR) spectroscopy. RESULTS 1H NMR-based metabolomics profiling of patients with newly diagnosed hypothyroidism (HypoT0) showed significantly higher levels of citrate, creatinine, glycerol, myo-inositol and serine, and lower levels of proline and taurine compared to controls. Interestingly, some metabolic changes were persistent three months after pharmacological treatments, despite normal serum TSH and thyroid hormone concentrations (HypoT1). When an Orthogonal Partial Least Squares Discriminant Analysis (OPLS-DA) model was built to evaluate possible differences in the metabolic profile between HypoT0 and HypoT1, the data obtained were not significantly different. CONCLUSION These results suggest that metabolic changes in the patients with hypothyroidism may persist after normalization of serum levels of FT3, FT4, and TSH, which currently represent the gold standard in laboratory testing for diagnosis and evaluation of thyroid pathology. So, the metabolomics approach may contribute to integrate classical hormone assays and to determine the euthyroid status achievement with greater efficacy.
Collapse
Affiliation(s)
- C Piras
- Department of Biomedical Sciences, University of Cagliari, 09042, Monserrato, Italy
| | - M Pibiri
- Department of Biomedical Sciences, University of Cagliari, 09042, Monserrato, Italy
| | - V P Leoni
- Department of Biomedical Sciences, University of Cagliari, 09042, Monserrato, Italy
| | - A Balsamo
- Department of Biomedical Sciences, University of Cagliari, 09042, Monserrato, Italy
| | - L Tronci
- Department of Biomedical Sciences, University of Cagliari, 09042, Monserrato, Italy
| | - N Arisci
- Department of Medical Sciences and Public Health, University of Cagliari, 09042, Monserrato, Italy
| | - S Mariotti
- Department of Medical Sciences and Public Health, University of Cagliari, 09042, Monserrato, Italy.
| | - L Atzori
- Department of Biomedical Sciences, University of Cagliari, 09042, Monserrato, Italy
| |
Collapse
|
9
|
Benvenga S, Nordio M, Laganà AS, Unfer V. The Role of Inositol in Thyroid Physiology and in Subclinical Hypothyroidism Management. Front Endocrinol (Lausanne) 2021; 12:662582. [PMID: 34040582 PMCID: PMC8143049 DOI: 10.3389/fendo.2021.662582] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 04/12/2021] [Indexed: 01/02/2023] Open
Abstract
Myo-Inositol (MYO) is the most abundant stereoisomer of inositols' family, cyclic polyols with 6 hydroxyl groups. Myo-Inositol has a relevant role in thyroid function and autoimmune diseases, as a precursor of phosphoinositides that takes part in the phosphatidylinositol (PI) signal transduction pathway. Among phosphoinositides, phosphatidylinositol 4,5- bisphosphate (PIP2) is the precursor of inositol triphosphates (IP3), second messenger of several hormones including thyroid-stimulating hormone (TSH). As a second messenger in the phospholipase C (PLC)-dependent inositol phosphate Ca2+/DAG pathway, Myo-Inositol is essential to produce H2O2 required for the synthesis of thyroid hormones. Consequently, depletion of Myo-Inositol or impaired inositol dependent TSH signaling pathway may predispose to the development of some thyroid diseases, such as hypothyroidism. Many clinical studies have shown that after treatment with Myo-Inositol plus Selenium (MYO+Se), TSH levels significantly decreased in patients with subclinical hypothyroidism with or without autoimmune thyroiditis. The TSH reduction was accompanied by a decline of antithyroid autoantibodies. Moreover, Myo-Inositol supplementation seemed to be involved also in the management of thyroidal benign nodules, with a possible effect in the size reduction. This review proposes a summary of the role of inositol, especially of Myo-Inositol, in the thyroidal physiology and its contribution on the management of some thyroid diseases.
Collapse
Affiliation(s)
- Salvatore Benvenga
- Department of Clinical and Experimental Medicine, Section of Endocrinology, University of Messina, Messina, Italy
- The Experts Group on Inositol in Basic and Clinical Research (EGOI), Rome, Italy
| | - Maurizio Nordio
- The Experts Group on Inositol in Basic and Clinical Research (EGOI), Rome, Italy
- Department of Experimental Medicine, “Sapienza” Università di Roma, Rome, Italy
| | - Antonio Simone Laganà
- The Experts Group on Inositol in Basic and Clinical Research (EGOI), Rome, Italy
- Department of Obstetrics and Gynecology, “Filippo Del Ponte” Hospital, University of Insubria, Varese, Italy
| | - Vittorio Unfer
- The Experts Group on Inositol in Basic and Clinical Research (EGOI), Rome, Italy
- Systems Biology Group Lab, “Sapienza” Università di Roma, Rome, Italy
| |
Collapse
|
10
|
Asghar MY, Lassila T, Törnquist K. Calcium Signaling in the Thyroid: Friend and Foe. Cancers (Basel) 2021; 13:cancers13091994. [PMID: 33919125 PMCID: PMC8122656 DOI: 10.3390/cancers13091994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 04/19/2021] [Accepted: 04/20/2021] [Indexed: 11/16/2022] Open
Abstract
Simple Summary All cells in our body are activated by several different signals. The calcium ion is one of the most versatile signaling molecules, and regulates a multitude of different events in the cells. These range from activation of muscle contraction, to the regulation of cell movement, just to name a few. In normal thyroid cells, calcium signaling is of importance for the normal physiology of the cells. In thyroid pathologies, e.g., thyroid cancer, calcium is important for the regulation of proliferation and invasion, and may also activate gene transcription programs important for cancer cell survival. In this Commentary, we summarize what is known regarding calcium in the normal thyroid, and highlight the importance of calcium signaling in thyroid pathologies. Abstract Calcium signaling participates in a vast number of cellular processes, ranging from the regulation of muscle contraction, cell proliferation, and mitochondrial function, to the regulation of the membrane potential in cells. The actions of calcium signaling are, thus, of great physiological significance for the normal functioning of our cells. However, many of the processes that are regulated by calcium, including cell movement and proliferation, are important in the progression of cancer. In the normal thyroid, calcium signaling plays an important role, and evidence is also being gathered showing that calcium signaling participates in the progression of thyroid cancer. This review will summarize what we know in regard to calcium signaling in the normal thyroid as, well as in thyroid cancer.
Collapse
Affiliation(s)
- Muhammad Yasir Asghar
- Minerva Foundation Institute for Medical Research, Biomedicum Helsinki 2U, Tukholmankatu 8, 00290 Helsinki, Finland; (M.Y.A.); (T.L.)
| | - Taru Lassila
- Minerva Foundation Institute for Medical Research, Biomedicum Helsinki 2U, Tukholmankatu 8, 00290 Helsinki, Finland; (M.Y.A.); (T.L.)
- Cell Biology, Faculty of Science and Engineering, Åbo Akademi University, Artillerigatan 6, 00250 Turku, Finland
| | - Kid Törnquist
- Minerva Foundation Institute for Medical Research, Biomedicum Helsinki 2U, Tukholmankatu 8, 00290 Helsinki, Finland; (M.Y.A.); (T.L.)
- Cell Biology, Faculty of Science and Engineering, Åbo Akademi University, Artillerigatan 6, 00250 Turku, Finland
- Correspondence:
| |
Collapse
|
11
|
Ma Q, Cao Z, Li H, Wang W, Tian Y, Yan L, Liao Y, Chen X, Chen Y, Shi Y, Tang S, Zhou N. Two naturally occurring mutations of human GPR103 define distinct G protein selection bias. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2021; 1868:119046. [PMID: 33872671 DOI: 10.1016/j.bbamcr.2021.119046] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Revised: 04/12/2021] [Accepted: 04/13/2021] [Indexed: 11/18/2022]
Abstract
The neuropeptide 26RFa plays important roles in the regulation of many physiological functions. 26RFa has been recognized as an endogenous ligand for receptor GPR103. In the present study, we demonstrate that GPR103 dually couples to Gαq and Gαi/o proteins. However, two naturally occurring missense mutations were identified from a young male patient. In the first, Y68H, induction of Ca2+ mobilization was noted without detection of ERK1/2 activation. In the second, R371W, the potential to activate ERK1/2 signaling was retained but with failure to evoke Ca2+ mobilization. Further analysis provides evidence that Gαq, L-type Ca2+ channel and PKCβI and βII are involved in the Y68H-mediated signaling pathway, whereas Gαi/o, Gβγ, and PKCζ are implicated in the R371W-induced signaling. Our results demonstrate that two point mutations, Y68H and R371W, affect the equilibrium between the different receptor conformations, leading to alteration of G protein-coupling preferences. Importantly, these findings provide a foundation for future elucidation of GPCR-mediated biased signaling and the physiological implications of their bias.
Collapse
Affiliation(s)
- Qiang Ma
- Institute of Biochemistry, College of Life Sciences, Zijingang Campus, Zhejiang University, Hangzhou, Zhejiang 310058, China; Department of Neurobiology, NHC and CAMS Key Laboratory of Medical Neurobiology, School of Brain Science and Brian Medicine, MOE Frontier Center of Brain Science and Brain-machine Integration, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
| | - Zheng Cao
- Institute of Biochemistry, College of Life Sciences, Zijingang Campus, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Huanzheng Li
- Wenzhou Key Laboratory of Birth Defects, Wenzhou Central Hospital, Wenzhou, Zhejiang 325000, China
| | - Weiwei Wang
- Institute of Biochemistry, College of Life Sciences, Zijingang Campus, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Yanan Tian
- Institute of Biochemistry, College of Life Sciences, Zijingang Campus, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Lili Yan
- Institute of Biochemistry, College of Life Sciences, Zijingang Campus, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Yuan Liao
- Institute of Biochemistry, College of Life Sciences, Zijingang Campus, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Xiangnan Chen
- Wenzhou Key Laboratory of Birth Defects, Wenzhou Central Hospital, Wenzhou, Zhejiang 325000, China
| | - Yu Chen
- Institute of Biochemistry, College of Life Sciences, Zijingang Campus, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Ying Shi
- Institute of Biochemistry, College of Life Sciences, Zijingang Campus, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Shaohua Tang
- Wenzhou Key Laboratory of Birth Defects, Wenzhou Central Hospital, Wenzhou, Zhejiang 325000, China; School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 32500, China
| | - Naiming Zhou
- Institute of Biochemistry, College of Life Sciences, Zijingang Campus, Zhejiang University, Hangzhou, Zhejiang 310058, China.
| |
Collapse
|
12
|
Yang LK, Hou ZS, Tao YX. Biased signaling in naturally occurring mutations of G protein-coupled receptors associated with diverse human diseases. Biochim Biophys Acta Mol Basis Dis 2021; 1867:165973. [PMID: 32949766 PMCID: PMC7722056 DOI: 10.1016/j.bbadis.2020.165973] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 09/07/2020] [Accepted: 09/14/2020] [Indexed: 12/15/2022]
Abstract
G protein-coupled receptors (GPCRs) play critical roles in transmitting a variety of extracellular signals into the cells and regulate diverse physiological functions. Naturally occurring mutations that result in dysfunctions of GPCRs have been known as the causes of numerous diseases. Significant progresses have been made in elucidating the pathophysiology of diseases caused by mutations. The multiple intracellular signaling pathways, such as G protein-dependent and β-arrestin-dependent signaling, in conjunction with recent advances on biased agonism, have broadened the view on the molecular mechanism of disease pathogenesis. This review aims to briefly discuss biased agonism of GPCRs (biased ligands and biased receptors), summarize the naturally occurring GPCR mutations that cause biased signaling, and propose the potential pathophysiological relevance of biased mutant GPCRs associated with various endocrine diseases.
Collapse
Affiliation(s)
- Li-Kun Yang
- Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL 36849, United States
| | - Zhi-Shuai Hou
- Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL 36849, United States
| | - Ya-Xiong Tao
- Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL 36849, United States.
| |
Collapse
|
13
|
Pace C, Tumino D, Russo M, Le Moli R, Naselli A, Borzì G, Malandrino P, Frasca F. Role of selenium and myo-inositol supplementation on autoimmune thyroiditis progression. Endocr J 2020; 67:1093-1098. [PMID: 32669509 DOI: 10.1507/endocrj.ej20-0062] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Previous reports indicate that selenium supplementation may be useful to reduce cell oxidative stress. In particular, selenium may decrease the level of thyroid autoantibodies in patients with Hashimoto's thyroiditis (HT). Recent studies also indicate that myo-inositol may have beneficial effects on thyroid function in patients with HT. Hence, the aim of the present study is to evaluate whether myo-inositol may enhance the protective effect of selenium on HT progression to hypothyroidism. The study was designed as observational and retrospective. Thyroid hormones were evaluated in patients with HT who were either euthyroid or subclinically hypothyroid. These patients were subdivided into three groups: untreated, treated with selenomethionine alone (Se-meth: 83 μg/day) and treated with Se-meth plus myo-inositol (Se-meth + Myo-I: 83 μg/day + 600 mg/day). Outcome evaluation was performed at baseline and after 6 and 12 months of treatment. High-resolution ultrasound of the thyroid gland was performed to evaluate changes in thyroid echoic pattern during the study. Compared to baseline, levels of thyroid-stimulating hormone (TSH) increased significantly in untreated patients but decreased by 31% and 38%, respectively, in those treated with Se-meth and Se-meth + Myo-I. Moreover, in the latter group the TSH reduction was observed earlier than in the Se-meth-treated group. Densitometric analysis of thyroid ultrasonography showed an echoic pattern improvement in both treated groups compared to untreated patients, although this difference was not statistically significant. Thus, Se-meth treatment is effective in patients with HT and its effect may be improved in combination with Myo-I through earlier achievement of TSH levels closer to physiological concentrations.
Collapse
Affiliation(s)
- Cinzia Pace
- Endocrinology Unit, Department of Clinical and Experimental Medicine, University of Catania, Garibaldi-Nesima Medical Center, Via Palermo 636, 95122 Catania, Italy
| | - Dario Tumino
- Endocrinology Unit, Department of Clinical and Experimental Medicine, University of Catania, Garibaldi-Nesima Medical Center, Via Palermo 636, 95122 Catania, Italy
| | - Marco Russo
- Endocrinology Unit, Department of Clinical and Experimental Medicine, University of Catania, Garibaldi-Nesima Medical Center, Via Palermo 636, 95122 Catania, Italy
| | - Rosario Le Moli
- Endocrinology Unit, Department of Clinical and Experimental Medicine, University of Catania, Garibaldi-Nesima Medical Center, Via Palermo 636, 95122 Catania, Italy
| | - Adriano Naselli
- Endocrinology Unit, Department of Clinical and Experimental Medicine, University of Catania, Garibaldi-Nesima Medical Center, Via Palermo 636, 95122 Catania, Italy
| | - Graziella Borzì
- Endocrinology Unit, Department of Clinical and Experimental Medicine, University of Catania, Garibaldi-Nesima Medical Center, Via Palermo 636, 95122 Catania, Italy
| | - Pasqualino Malandrino
- Endocrinology Unit, Department of Clinical and Experimental Medicine, University of Catania, Garibaldi-Nesima Medical Center, Via Palermo 636, 95122 Catania, Italy
| | - Francesco Frasca
- Endocrinology Unit, Department of Clinical and Experimental Medicine, University of Catania, Garibaldi-Nesima Medical Center, Via Palermo 636, 95122 Catania, Italy
| |
Collapse
|
14
|
Chen J, Chen X, Li S, Jiang Y, Mao H, Zhang R, Ji B, Yan M, Cai X, Wang C. Individual phosphorylation sites at the C-terminus of the apelin receptor play different roles in signal transduction. Redox Biol 2020; 36:101629. [PMID: 32863206 PMCID: PMC7338617 DOI: 10.1016/j.redox.2020.101629] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 06/24/2020] [Accepted: 06/29/2020] [Indexed: 12/16/2022] Open
Abstract
The apelin and Elabela proteins constitute a spatiotemporal double-ligand system that controls apelin receptor (APJ) signal transduction. Phosphorylation of multiple sites within the C-terminus of APJ is essential for the recruitment of β-arrestins. We sought to determine the precise mechanisms by which apelin and Elabela promote APJ phosphorylation, and to elucidate the influence of β-arrestin phosphorylation on G-protein-coupled receptor (GPCR)/β-arrestin-dependent signaling. We used techniques including mass spectrometry (MS), mutation analysis, and bioluminescence resonance energy transfer (BRET) to evaluate the role of phosphorylation sites in APJ-mediated G-protein-dependent and β-dependent signaling. Phosphorylation of APJ occurred at five serine residues in the C-terminal region (Ser335, Ser339, Ser345, Ser348 and Ser369). We also identified two phosphorylation sites in β-arrestin1 and three in β-arrestin2, including three previously identified residues (Ser412, Ser361, and Thr383) and two new sites, Tyr47 in β-arrestin1 and Tyr48 in β-arrestin2. APJ mutations did not affect the phosphorylation of β-arrestins, but it affects the β-arrestin signaling pathway, specifically Ser335 and Ser339. Mutation of Ser335 decreased the ability of the receptor to interact with β-arrestin1/2 and AP2, indicating that APJ affects the β-arrestin signaling pathway by stimulating Elabela. Mutation of Ser339 abolished the capability of the receptor to interact with GRK2 and β-arrestin1/2 upon stimulation with apelin-36, and disrupted receptor internalization and β-arrestin-dependent ERK1/2 activation. Five peptides act on distinct phosphorylation sites at the APJ C-terminus, differentially regulating APJ signal transduction and causing different biological effects. These findings may facilitate screening for drugs to treat cardiovascular and metabolic diseases.
Collapse
Affiliation(s)
- Jing Chen
- Neurobiology Institute, Jining Medical University, Jining, Shandong, China; Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry, CV4 7AL, United Kingdom.
| | - Xiaoyu Chen
- Department of Physiology, Shandong First Medical University (Shandong Academy of Medical Sciences), Taian, Shandong, China
| | - Sheng Li
- Neurobiology Institute, Jining Medical University, Jining, Shandong, China
| | - Yunlu Jiang
- Neurobiology Institute, Jining Medical University, Jining, Shandong, China
| | - Huiling Mao
- Neurobiology Institute, Jining Medical University, Jining, Shandong, China
| | - Rumin Zhang
- Neurobiology Institute, Jining Medical University, Jining, Shandong, China
| | - Bingyuan Ji
- Neurobiology Institute, Jining Medical University, Jining, Shandong, China
| | - Maocai Yan
- School of Pharmacy, Jining Medical University, Shandong, China
| | - Xin Cai
- Department of Physiology, Weifang Medical University, Weifang, Shandong, China
| | - Chunmei Wang
- Neurobiology Institute, Jining Medical University, Jining, Shandong, China
| |
Collapse
|
15
|
Rocco DA, Paluzzi JPV. Expression Profiling, Downstream Signaling, and Inter-subunit Interactions of GPA2/GPB5 in the Adult Mosquito Aedes aegypti. Front Endocrinol (Lausanne) 2020; 11:158. [PMID: 32296389 PMCID: PMC7137729 DOI: 10.3389/fendo.2020.00158] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Accepted: 03/06/2020] [Indexed: 12/22/2022] Open
Abstract
GPA2/GPB5 and its receptor constitute a glycoprotein hormone-signaling system native to the genomes of most vertebrate and invertebrate organisms. Unlike the well-studied gonadotropins and thyrotropin, the exact function of GPA2/GPB5 remains elusive, and whether it elicits its functions as heterodimers, homodimers or as independent monomers remains unclear. Here, the glycoprotein hormone signaling system was investigated in adult mosquitoes, where GPA2 and GPB5 subunit expression was mapped and modes of its signaling were characterized. In adult Aedes aegypti mosquitoes, GPA2 and GPB5 transcripts co-localized to bilateral pairs of neuroendocrine cells, positioned within the first five abdominal ganglia of the central nervous system. Unlike GPA2/GPB5 homologs in human and fly, GPA2/GPB5 subunits in A. aegypti lacked evidence of heterodimerization. Rather, cross-linking analysis to determine subunit interactions revealed A. aegypti GPA2 and GPB5 subunits may form homodimers, although treatments with independent subunits did not demonstrate receptor activity. Since mosquito GPA2/GPB5 heterodimers were not evident by heterologous expression, a tethered fusion construct was generated for expression of the subunits as a single polypeptide chain to mimic heterodimer formation. Our findings revealed A. aegypti LGR1 elicited constitutive activity with elevated levels of cAMP. However, upon treatment with recombinant tethered GPA2/GPB5, an inhibitory G protein (Gi/o) signaling cascade is initiated and forskolin-induced cAMP production is inhibited. These results further support the notion that heterodimerization is a requirement for glycoprotein hormone receptor activation and provide novel insight to how signaling is achieved for GPA2/GPB5, an evolutionary ancient neurohormone.
Collapse
|
16
|
Fallahi P, Ferrari SM, Elia G, Ragusa F, Paparo SR, Caruso C, Guglielmi G, Antonelli A. Myo-inositol in autoimmune thyroiditis, and hypothyroidism. Rev Endocr Metab Disord 2018; 19:349-354. [PMID: 30506520 DOI: 10.1007/s11154-018-9477-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Myo-inositol (Myo-Ins) plays an important role in thyroid function and autoimmunity. Myo-Ins is the precursor for the synthesis of phosphoinositides, which takes part in the phosphatidylinositol (PtdIns) signal transduction pathway, and plays a decisive role in several cellular processes. In the thyroid cells, PtdIns is involved in the intracellular thyroid-stimulating hormone (TSH) signaling, via Phosphatidylinositol (3,4,5)-trisphosphate (PtdIns(3,4,5)P3) (PIP-3). Moreover, the phosphatidyl inositol 3 kinases (PI3K) family of lipid kinases regulates diverse aspects of T, B, and Tregs lymphocyte behaviour. Different mouse models deficient for the molecules involved in the PIP3 pathway suggest that impairment of PIP3 signaling leads to dysregulation of immune responses and, sometimes, autoimmunity. Studies have shown that cytokines modulate Myo-Ins in thyroid cells. Moreover, clinical studies have shown that after treatment with Myo-inositol plus seleniomethionine (Myo-Ins + Se), TSH levels significantly declined in patients with subclinical hypothyroidism due to autoimmune thyroiditis. The treatment was accompanied by a decline of antithyroid autoantibodies. After treatment serum CXCL10 levels declined, confirming the immune-modulatory effect of Myo-Ins. Additional research is necessary in larger population to evaluate the effect on the quality of life, and to study the mechanism of the effect on chemokines.
Collapse
Affiliation(s)
- Poupak Fallahi
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via Savi 10, Pisa, 56126, Italy
| | - Silvia Martina Ferrari
- Department of Clinical and Experimental Medicine, University of Pisa, School of Medicine, Via Savi, 10, I-56126, Pisa, Italy
| | - Giusy Elia
- Department of Clinical and Experimental Medicine, University of Pisa, School of Medicine, Via Savi, 10, I-56126, Pisa, Italy
| | - Francesca Ragusa
- Department of Clinical and Experimental Medicine, University of Pisa, School of Medicine, Via Savi, 10, I-56126, Pisa, Italy
| | - Sabrina Rosaria Paparo
- Department of Clinical and Experimental Medicine, University of Pisa, School of Medicine, Via Savi, 10, I-56126, Pisa, Italy
| | - Claudia Caruso
- Department of Clinical and Experimental Medicine, University of Pisa, School of Medicine, Via Savi, 10, I-56126, Pisa, Italy
| | - Giovanni Guglielmi
- U.O. Medicina Preventiva del Lavoro, Azienda Ospedaliero-Universitaria Pisana, I-56124, Pisa, Italy
| | - Alessandro Antonelli
- Department of Clinical and Experimental Medicine, University of Pisa, School of Medicine, Via Savi, 10, I-56126, Pisa, Italy.
| |
Collapse
|
17
|
Ferrari SM, Elia G, Ragusa F, Paparo SR, Caruso C, Benvenga S, Fallahi P, Antonelli A. The protective effect of myo-inositol on human thyrocytes. Rev Endocr Metab Disord 2018; 19:355-362. [PMID: 30511181 DOI: 10.1007/s11154-018-9476-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Patients affected by autoimmune thyroiditis reached positive effects on indices of thyroid autoimmunity and/or thyroidal function, after following a treatment with selenomethionine (Se) alone, or Se in combination with Myo-inositol (Myo-Ins). Our purpose was to investigate if Myo-Ins alone, or a combination of Se + Myo-Ins, is effective in protecting thyroid cells from the effects given by cytokines, or hydrogen peroxide (H2O2). We assessed the interferon (IFN)-γ-inducible protein 10 (IP-10/CXCL10) secretion by stimulating primary thyrocytes (obtained from Hashimoto's thyroiditis or from control patients) with cytokines in presence/absence of H2O2. Our results confirm: 1) the toxic effect of H2O2 in primary thyrocytes that leads to an increase of the apoptosis, to a decrease of the proliferation, and to a slight reduction of cytokines-induced CXCL10 secretion; 2) the secretion of CXCL10 chemokine induced by IFN-γ + tumor necrosis factor alpha (TNF)-α has been decreased by Myo + Ins, both in presence or absence of H2O2; 3) no effect has been shown by the treatment with Se. Therefore, a protective effect of Myo-Ins on thyroid cells has been suggested by our data, which exact mechanisms are at the basis of this effect need to be furtherly investigated.
Collapse
Affiliation(s)
- Silvia Martina Ferrari
- Department of Clinical and Experimental Medicine, University of Pisa, School of Medicine, Via Savi, 10, I-56126, Pisa, Italy
| | - Giusy Elia
- Department of Clinical and Experimental Medicine, University of Pisa, School of Medicine, Via Savi, 10, I-56126, Pisa, Italy
| | - Francesca Ragusa
- Department of Clinical and Experimental Medicine, University of Pisa, School of Medicine, Via Savi, 10, I-56126, Pisa, Italy
| | - Sabrina Rosaria Paparo
- Department of Clinical and Experimental Medicine, University of Pisa, School of Medicine, Via Savi, 10, I-56126, Pisa, Italy
| | - Claudia Caruso
- Department of Clinical and Experimental Medicine, University of Pisa, School of Medicine, Via Savi, 10, I-56126, Pisa, Italy
| | - Salvatore Benvenga
- Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy
- Master Program on Childhood, Adolescent and Women's Endocrine Health, University of Messina, Messina, Italy
- Interdepartmental Program of Molecular and Clinical Endocrinology and Women's Endocrine Health, Azienda Ospedaliera Universitaria Policlinico 'G. Martino', I-98125, Messina, Italy
| | - Poupak Fallahi
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Alessandro Antonelli
- Department of Clinical and Experimental Medicine, University of Pisa, School of Medicine, Via Savi, 10, I-56126, Pisa, Italy.
| |
Collapse
|
18
|
Sugisawa C, Abe K, Sunaga Y, Taniyama M, Hasegawa T, Narumi S. Identification of compound heterozygous TSHR mutations (R109Q and R450H) in a patient with nonclassic TSH resistance and functional characterization of the mutant receptors. Clin Pediatr Endocrinol 2018; 27:123-130. [PMID: 30083029 PMCID: PMC6073063 DOI: 10.1297/cpe.27.123] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2017] [Accepted: 02/08/2018] [Indexed: 11/05/2022] Open
Abstract
Genetic defects of the TSH receptor (TSHR) signaling pathway cause a form of congenital
hypothyroidism (CH) known as TSH resistance. Consistent with the physiological
understanding that thyroidal iodine uptake is up-regulated by TSHR signaling, most
patients with TSH resistance have low to normal thyroidal 123I uptake
representing the classic TSH resistance. However, paradoxically high 123I
uptake was reported in four molecularly-confirmed patients indicating nonclassic TSH
resistance. Here, we report the fifth patient with the nonclassic phenotype. He was a
12-yr-old CH patient and treated with levothyroxine. At the age 11 yr, he showed slightly
small thyroid gland and elevated thyroidal 123I uptake. Genetic analysis showed
that he was compound heterozygous for two known missense mutations (Arg109Gln and
Arg450His) in the TSHR gene. Further, the signal transduction of Arg109Gln-TSHR was
defective in both Gs- and Gq-coupled pathways, while Arg450His-TSHR showed Gq-dominant
defect. 123I uptake was evaluated earlier in 16 patients with TSH resistance,
and a correlation between TSH levels and 123I uptake was shown in patients with
specific genotypes (Arg450His or Leu653Val). Collectively, we have re-confirmed that the
emergence of the nonclassic phenotype requires two factors: mutant TSHR with Gq-dominant
coupling defect and relatively high levels of serum TSH.
Collapse
Affiliation(s)
- Chiho Sugisawa
- Department of Pediatrics, Keio University School of Medicine, Tokyo, Japan.,Department of Internal Medicine, Showa University Fujigaoka Hospital, Kanagawa, Japan
| | - Kiyomi Abe
- Department of Pediatrics, Keio University School of Medicine, Tokyo, Japan
| | | | - Matsuo Taniyama
- Department of Internal Medicine, Showa University Fujigaoka Hospital, Kanagawa, Japan.,Tokyo Health Service Association, Tokyo, Japan
| | - Tomonobu Hasegawa
- Department of Pediatrics, Keio University School of Medicine, Tokyo, Japan
| | - Satoshi Narumi
- Department of Molecular Endocrinology, National Research Institute for Child Health and Development, Tokyo, Japan
| |
Collapse
|
19
|
Löf C, Patyra K, Kero A, Kero J. Genetically modified mouse models to investigate thyroid development, function and growth. Best Pract Res Clin Endocrinol Metab 2018; 32:241-256. [PMID: 29779579 DOI: 10.1016/j.beem.2018.03.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
The thyroid gland produces thyroid hormones (TH), which are essential regulators for growth, development and metabolism. The thyroid is mainly controlled by the thyroid-stimulating hormone (TSH) that binds to its receptor (TSHR) on thyrocytes and mediates its action via different G protein-mediated signaling pathways. TSH primarily activates the Gs-pathway, and at higher concentrations also the Gq/11-pathway, leading to an increase of intracellular cAMP and Ca2+, respectively. To date, the physiological importance of other G protein-mediated signaling pathways in thyrocytes is unclear. Congenital hypothyroidism (CH) is defined as the lack of TH at birth. In familial cases, high-throughput sequencing methods have facilitated the identification of novel mutations. Nevertheless, the precise etiology of CH yet remains unraveled in a proportion of cases. Genetically modified mouse models can reveal new pathophysiological mechanisms of thyroid diseases. Here, we will present an overview of genetic mouse models for thyroid diseases, which have provided crucial insights into thyroid gland development, function, and growth with a special focus on TSHR and microRNA signaling.
Collapse
Affiliation(s)
- C Löf
- Research Centre for Integrative Physiology and Pharmacology, Institute of Biomedicine, Turku Center for Disease Modeling, University of Turku, Kiinamyllynkatu 10, 20520, Turku, Finland
| | - K Patyra
- Research Centre for Integrative Physiology and Pharmacology, Institute of Biomedicine, Turku Center for Disease Modeling, University of Turku, Kiinamyllynkatu 10, 20520, Turku, Finland
| | - A Kero
- Department of Pediatrics, Turku University Hospital, Kiinamyllynkatu 4-8, 20521, Turku, Finland
| | - J Kero
- Research Centre for Integrative Physiology and Pharmacology, Institute of Biomedicine, Turku Center for Disease Modeling, University of Turku, Kiinamyllynkatu 10, 20520, Turku, Finland; Department of Pediatrics, Turku University Hospital, Kiinamyllynkatu 4-8, 20521, Turku, Finland.
| |
Collapse
|
20
|
Patyra K, Jaeschke H, Löf C, Jännäri M, Ruohonen ST, Undeutsch H, Khalil M, Kero A, Poutanen M, Toppari J, Chen M, Weinstein LS, Paschke R, Kero J. Partial thyrocyte-specific Gα s deficiency leads to rapid-onset hypothyroidism, hyperplasia, and papillary thyroid carcinoma-like lesions in mice. FASEB J 2018; 32:fj201800211R. [PMID: 29799790 DOI: 10.1096/fj.201800211r] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/28/2024]
Abstract
Thyroid function is controlled by thyroid-stimulating hormone (TSH), which binds to its G protein-coupled receptor [thyroid-stimulating hormone receptor (TSHR)] on thyrocytes. TSHR can potentially couple to all G protein families, but it mainly activates the Gs- and Gq/11-mediated signaling cascades. To date, there is a knowledge gap concerning the role of the individual G protein cascades in thyroid pathophysiology. Here, we demonstrate that the thyrocyte-specific deletion of Gs-protein α subunit (Gαs) in adult mice [tamoxifen-inducible Gs protein α subunit deficient (iTGαsKO) mice] rapidly impairs thyrocyte function and leads to hypothyroidism. Consequently, iTGαsKO mice show reduced food intake and activity. However, body weight and the amount of white adipose tissue were decreased only in male iTGαsKO mice. Unexpectedly, hyperplastic follicles and papillary thyroid cancer-like tumor lesions with increased proliferation and slightly increased phospho-ERK1/2 staining were found in iTGαsKO mice at an older age. These tumors developed from nonrecombined thyrocytes still expressing Gαs in the presence of highly elevated serum TSH. In summary, we report that partial thyrocyte-specific Gαs deletion leads to hypothyroidism but also to tumor development in thyrocytes with remaining Gαs expression. Thus, these mice are a novel model to elucidate the pathophysiological consequences of hypothyroidism and TSHR/Gs/cAMP-mediated tumorigenesis.-Patyra, K., Jaeschke, H., Löf, C., Jännäri, M., Ruohonen, S. T., Undeutsch, H., Khalil, M., Kero, A., Poutanen, M., Toppari, J., Chen, M., Weinstein, L. S., Paschke, R., Kero, J. Partial thyrocyte-specific Gαs deficiency leads to rapid-onset hypothyroidism, hyperplasia, and papillary thyroid carcinoma-like lesions in mice.
Collapse
Affiliation(s)
- Konrad Patyra
- Research Centre for Integrative Physiology and Pharmacology, Institute of Biomedicine, University of Turku, Turku, Finland
- Turku Center for Disease Modeling, University of Turku, Turku, Finland
| | - Holger Jaeschke
- Research Centre for Integrative Physiology and Pharmacology, Institute of Biomedicine, University of Turku, Turku, Finland
| | - Christoffer Löf
- Research Centre for Integrative Physiology and Pharmacology, Institute of Biomedicine, University of Turku, Turku, Finland
- Turku Center for Disease Modeling, University of Turku, Turku, Finland
| | - Meeri Jännäri
- Research Centre for Integrative Physiology and Pharmacology, Institute of Biomedicine, University of Turku, Turku, Finland
| | - Suvi T Ruohonen
- Research Centre for Integrative Physiology and Pharmacology, Institute of Biomedicine, University of Turku, Turku, Finland
- Turku Center for Disease Modeling, University of Turku, Turku, Finland
| | - Henriette Undeutsch
- Research Centre for Integrative Physiology and Pharmacology, Institute of Biomedicine, University of Turku, Turku, Finland
| | - Moosa Khalil
- Department of Pathology and Laboratory Medicine, University of Calgary, Calgary,
Alberta, Canada
| | - Andreina Kero
- Department of Pediatrics, Turku University Hospital, Finland
| | - Matti Poutanen
- Research Centre for Integrative Physiology and Pharmacology, Institute of Biomedicine, University of Turku, Turku, Finland
- Turku Center for Disease Modeling, University of Turku, Turku, Finland
| | - Jorma Toppari
- Research Centre for Integrative Physiology and Pharmacology, Institute of Biomedicine, University of Turku, Turku, Finland
- Department of Pediatrics, Turku University Hospital, Finland
| | - Min Chen
- Metabolic Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Lee S Weinstein
- Metabolic Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Ralf Paschke
- Arnie Charbonneau Cancer Research Institute, University of Calgary, Calgary, Alberta, Canada
| | - Jukka Kero
- Research Centre for Integrative Physiology and Pharmacology, Institute of Biomedicine, University of Turku, Turku, Finland
- Turku Center for Disease Modeling, University of Turku, Turku, Finland
- Department of Pediatrics, Turku University Hospital, Finland
| |
Collapse
|
21
|
Abe K, Narumi S, Suwanai AS, Adachi M, Muroya K, Asakura Y, Nagasaki K, Abe T, Hasegawa T. Association between monoallelic TSHR mutations and congenital hypothyroidism: a statistical approach. Eur J Endocrinol 2018; 178:137-144. [PMID: 29092890 DOI: 10.1530/eje-16-1049] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Revised: 09/30/2017] [Accepted: 11/01/2017] [Indexed: 11/08/2022]
Abstract
OBJECTIVE Biallelic TSHR mutations cause congenital hypothyroidism (CH). Serum TSH levels of monoallelic mutation carriers range from normal to mildly elevated, and thus the size of its effect remains unclear. The objectives were to examine the association between monoallelic TSHR mutations and positivity at newborn screening (NBS) for CH, and to test whether the association was modified by another genetic factor. SUBJECTS AND METHODS We enrolled 395 patients that had a positive result in NBS and sequenced TSHR. Monoallelic TSHR mutation carriers were further sequenced for DUOX2. Molecular functions of the mutations were verified in vitro. The frequency of the mutations in the study subjects was compared with a theoretical value in the Japanese general population. Odds ratio (OR) for NBS positivity associated with the mutation was calculated. Using Bayes' theorem, we estimated a posterior probability of NBS positivity given the mutation. RESULTS Twenty-six monoallelic TSHR mutation carriers were found. Four out of the 26 also had a monoallelic DUOX2 mutation (double heterozygotes). The frequencies of monoallelic TSHR mutation carriers (6.6%) and double heterozygotes (1.0%) were significantly higher than those in the general population (0.58% and 0.0087%, respectively). OR for NBS positivity of having a monoallelic TSHR mutation or being a double heterozygote was 12.0 or 117.9, respectively. Posterior probability of NBS positivity was 0.38% in monoallelic TSHR mutation carriers and 3.8% in double heterozygotes. CONCLUSIONS Monoallelic TSHR mutations are significantly associated with NBS positivity, and the association is further strengthened by the coexistence of monoallelic DUOX2 mutations.
Collapse
Affiliation(s)
- Kiyomi Abe
- Department of Pediatrics, Keio University School of Medicine, Tokyo, Japan
| | - Satoshi Narumi
- Department of Pediatrics, Keio University School of Medicine, Tokyo, Japan
- Department of Molecular Endocrinology, National Research Institute for Child Health and Development, Tokyo, Japan
| | - Ayuko S Suwanai
- Department of Pediatrics, Keio University School of Medicine, Tokyo, Japan
| | - Masanori Adachi
- Department of Endocrinology and Metabolism, Kanagawa Children's Medical Center, Yokohama, Japan
| | - Koji Muroya
- Department of Endocrinology and Metabolism, Kanagawa Children's Medical Center, Yokohama, Japan
| | - Yumi Asakura
- Department of Endocrinology and Metabolism, Kanagawa Children's Medical Center, Yokohama, Japan
| | - Keisuke Nagasaki
- Division of Pediatrics, Department of Homeostatic Regulation and Development, Niigata University Graduate School of Medicine and Dental Sciences, Niigata, Japan
| | - Takayuki Abe
- Center for Clinical Research, Keio University School of Medicine, Tokyo, Japan
| | - Tomonobu Hasegawa
- Department of Pediatrics, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
22
|
Abstract
Resistance to thyrotropin (RTSH) is broadly defined as reduced sensitivity of thyroid follicle cells to stimulation by biologically active TSH due to genetic defects. Affected individuals have elevated serum TSH in the absence of goiter, with the severity ranging from nongoitrous isolated hyperthyrotropinemia to severe congenital hypothyroidism with thyroid hypoplasia. Conceptually, defects leading to RTSH impair both aspects of TSH-mediated action, namely thyroid hormone synthesis and gland growth. These include inactivating mutations in the genes encoding the TSH receptor and the PAX8 transcription factor. A common third cause has been genetically mapped to a locus on chromosome 15, but the underlying pathophysiology has not yet been elucidated. This review provides a succinct overview of currently defined causes of nonsyndromic RTSH, their differential diagnoses (autoimmune; partial iodine organification defects; syndromic forms of RTSH) and implications for the clinical approach to patients with RTSH.
Collapse
Affiliation(s)
- Helmut Grasberger
- University of Michigan, 6504 MSRB I, 1150 West Medical Center Drive, Ann Arbor, MI 48109, USA.
| | - Samuel Refetoff
- The University of Chicago, MC3090, 5841 South Maryland Avenue, Chicago, IL 60637, USA.
| |
Collapse
|
23
|
Khajavi N, Mergler S, Biebermann H. 3-Iodothyronamine, a Novel Endogenous Modulator of Transient Receptor Potential Melastatin 8? Front Endocrinol (Lausanne) 2017; 8:198. [PMID: 28861042 PMCID: PMC5561014 DOI: 10.3389/fendo.2017.00198] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Accepted: 07/28/2017] [Indexed: 11/22/2022] Open
Abstract
The decarboxylated and deiodinated thyroid hormone (TH) derivative, 3-iodothyronamine (3-T1AM), is suggested to be involved in energy metabolism and thermoregulation. G protein-coupled receptors (GPCRs) are known as the main targets for 3-T1AM; however, transient receptor potential channels (TRPs) were also recently identified as new targets of 3-T1AM. This article reviews the current knowledge of a putative novel role of 3-T1AM in the modulation of TRPs. Specifically, the TRP melastatin 8 (TRPM8) was identified as a target of 3-T1AM in different cell types including neoplastic cells, whereby 3-T1AM significantly increased cytosolic Ca2+ through TRPM8 activation. Similarly, the β-adrenergic receptor is involved in 3-T1AM-induced Ca2+ influx. Therefore, it has been suggested that 3-T1AM-induced Ca2+ mobilization might be due to β-adrenergic receptor/TRPM8 channel interaction, which adds to the complexity of GPCR regulation by TRPs. It has been revealed that TRPM8 activation leads to a decline in TRPV1 activity, which may be of therapeutic benefit in clinical circumstances such as treatment of TRPV1-mediated inflammatory hyperalgesia, colitis, and dry eye syndrome. This review also summarizes the inverse association between changes in TRPM8 and TRPV1 activity after 3-T1AM stimulation. This finding prompted further detailed investigations of the interplay between 3-T1AM and the GPCR/TRPM8 axis and indicated the probability of additional GPCR/TRP constellations that are modulated by this TH derivative.
Collapse
Affiliation(s)
- Noushafarin Khajavi
- Institute for Experimental Pediatric Endocrinology, Charité University of Medicine Berlin, Berlin, Germany
- *Correspondence: Noushafarin Khajavi,
| | - Stefan Mergler
- Department of Ophthalmology, Charité University of Medicine Berlin, Berlin, Germany
| | - Heike Biebermann
- Institute for Experimental Pediatric Endocrinology, Charité University of Medicine Berlin, Berlin, Germany
| |
Collapse
|
24
|
Abstract
Myo-inositol and phosphatidylinositol(s) play a pivotal function in many metabolic pathways that, if impaired, impact unfavorably on human health. This review analyzes several experimental and clinical investigations regarding the involvement of this class of molecules in physiological and pathological situations, with a major focus on thyroid. Central issues are the relationship between phosphatidylinositol and thyrotropin (TSH) signaling on one hand, and phosphatydylinositol and autoimmunity on the other hand. Other issues are the consequences of malfunction of some receptors, such as those ones for TSH (TSHR), insulin (IR) and insulin-like growth factor-1 (IGF-1R), or the connection between serum TSH concentrations and insulin resistance. Also covered are insulin resistance, metabolic syndrome and their allied disorders (diabetes, polycystic ovary syndrome [PCOS]), autoimmunity and certain malignancies, with their reciprocal links. Myoinositol has promising therapeutic potential. Appreciation of the inositol pathways involved in certain disorders, as mentioned in this review, may stimulate researchers to envisage additional therapeutic applications.
Collapse
Affiliation(s)
- Salvatore Benvenga
- Department of Clinical and Experimental Medicine, University of Messina School of Medicine, via Consolare Valeria, 1, 98125, Messina, Italy.
- Master Program of Childhood, Adolescence and Women's Endocrine Health, University of Messina School of Medicine, via Consolare Valeria, 1, 98125, Messina, Italy.
- Interdepartmental Program of Molecular & Clinical Endocrinology, and Women's Endocrine Health, University hospital, Padiglione H, 4 piano, Policlinico G. Martino, 98125, Messina, Italy.
| | - Alessandro Antonelli
- Department of Clinical and Experimental Medicine, University of Pisa, 56126, Pisa, Italy
| |
Collapse
|
25
|
Bizzarri M, Fuso A, Dinicola S, Cucina A, Bevilacqua A. Pharmacodynamics and pharmacokinetics of inositol(s) in health and disease. Expert Opin Drug Metab Toxicol 2016; 12:1181-96. [PMID: 27351907 DOI: 10.1080/17425255.2016.1206887] [Citation(s) in RCA: 103] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
INTRODUCTION Inositol and its derivatives comprise a huge field of biology. Myo-inositol is not only a prominent component of membrane-incorporated phosphatidylinositol, but participates in its free form, with its isomers or its phosphate derivatives, to a multitude of cellular processes, including ion channel permeability, metabolic homeostasis, mRNA export and translation, cytoskeleton remodeling, stress response. AREAS COVERED Bioavailability, safety, uptake and metabolism of inositol is discussed emphasizing the complexity of interconnected pathways leading to phosphoinositides, inositol phosphates and more complex molecules, like glycosyl-phosphatidylinositols. EXPERT OPINION Besides being a structural element, myo-inositol exerts unexpected functions, mostly unknown. However, several reports indicate that inositol plays a key role during phenotypic transitions and developmental phases. Furthermore, dysfunctions in the regulation of inositol metabolism have been implicated in several chronic diseases. Clinical trials using inositol in pharmacological doses provide amazing results in the management of gynecological diseases, respiratory stress syndrome, Alzheimer's disease, metabolic syndrome, and cancer, for which conventional treatments are disappointing. However, despite the widespread studies carried out to identify inositol-based effects, no comprehensive understanding of inositol-based mechanisms has been achieved. An integrated metabolomics-genomic study to identify the cellular fate of therapeutically administered myo-inositol and its genomic/enzymatic targets is urgently warranted.
Collapse
Affiliation(s)
- Mariano Bizzarri
- a Department of Experimental Medicine , Sapienza University of Rome , Rome , Italy.,b Systems Biology Group Lab , Sapienza University of Rome , Rome , Italy
| | - Andrea Fuso
- b Systems Biology Group Lab , Sapienza University of Rome , Rome , Italy.,c European Center for Brain Research (CERC)/IRCCS Santa Lucia Foundation , Rome , Italy
| | - Simona Dinicola
- d Department of Clinical and Molecular Medicine , Sapienza Universityof Rome , Rome , Italy.,e Department of Surgery 'Pietro Valdoni' , Sapienza University of Rome , Rome , Italy
| | - Alessandra Cucina
- e Department of Surgery 'Pietro Valdoni' , Sapienza University of Rome , Rome , Italy.,f Azienda Policlinico Umberto I , Rome , Italy
| | - Arturo Bevilacqua
- g Department of Psychology, Section of Neuroscience , Sapienza University of Rome , Rome , Italy
| |
Collapse
|
26
|
Montesinos MDM, Nicola JP, Nazar M, Peyret V, Lucero AM, Pellizas CG, Masini-Repiso AM. Nitric oxide-repressed Forkhead factor FoxE1 expression is involved in the inhibition of TSH-induced thyroid peroxidase levels. Mol Cell Endocrinol 2016; 420:105-15. [PMID: 26610751 DOI: 10.1016/j.mce.2015.11.020] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Revised: 11/02/2015] [Accepted: 11/13/2015] [Indexed: 01/07/2023]
Abstract
Thyroid peroxidase (TPO) is essential for thyroid hormone synthesis mediating the covalent incorporation of iodine into tyrosine residues of thyroglobulin process known as organification. Thyroid-stimulating hormone (TSH) via cAMP signaling is the main hormonal regulator of TPO gene expression. In thyroid cells, TSH-stimulated nitric oxide (NO) production inhibits TSH-induced thyroid-specific gene expression, suggesting a potential autocrine role of NO in modulating thyroid function. Indeed, NO donors downregulate TSH-induced iodide accumulation and organification in thyroid cells. Here, using FRTL-5 thyroid cells as model, we obtained insights into the molecular mechanism underlying the inhibitory effects of NO on iodide organification. We demonstrated that NO donors inhibited TSH-stimulated TPO expression by inducing a cyclic guanosine monophosphate-dependent protein kinase-mediated transcriptional repression of the TPO gene. Moreover, we characterized the FoxE1 binding site Z as mediator of the NO-inhibited TPO expression. Mechanistically, we demonstrated that NO decreases TSH-induced FoxE1 expression, thus repressing the transcripcional activation of TPO gene. Taken together, we provide novel evidence reinforcing the inhibitory role of NO on thyroid cell function, an observation of potential pathophysiological relevance associated with human thyroid pathologies that come along with changes in the NO production.
Collapse
Affiliation(s)
- María del Mar Montesinos
- Centro de Investigaciones en Bioquímica Clínica e Inmunología - Consejo Nacional de Investigaciones Científicas y Técnicas (CIBICI-CONICET), Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Juan Pablo Nicola
- Centro de Investigaciones en Bioquímica Clínica e Inmunología - Consejo Nacional de Investigaciones Científicas y Técnicas (CIBICI-CONICET), Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Magalí Nazar
- Centro de Investigaciones en Bioquímica Clínica e Inmunología - Consejo Nacional de Investigaciones Científicas y Técnicas (CIBICI-CONICET), Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Victoria Peyret
- Centro de Investigaciones en Bioquímica Clínica e Inmunología - Consejo Nacional de Investigaciones Científicas y Técnicas (CIBICI-CONICET), Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Ariel Maximiliano Lucero
- Centro de Investigaciones en Bioquímica Clínica e Inmunología - Consejo Nacional de Investigaciones Científicas y Técnicas (CIBICI-CONICET), Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Claudia Gabriela Pellizas
- Centro de Investigaciones en Bioquímica Clínica e Inmunología - Consejo Nacional de Investigaciones Científicas y Técnicas (CIBICI-CONICET), Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Ana María Masini-Repiso
- Centro de Investigaciones en Bioquímica Clínica e Inmunología - Consejo Nacional de Investigaciones Científicas y Técnicas (CIBICI-CONICET), Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina.
| |
Collapse
|
27
|
Kuşcu N, Bizzarri M, Bevilacqua A. Myo-Inositol Safety in Pregnancy: From Preimplantation Development to Newborn Animals. Int J Endocrinol 2016; 2016:2413857. [PMID: 27698667 PMCID: PMC5028874 DOI: 10.1155/2016/2413857] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Accepted: 08/15/2016] [Indexed: 01/11/2023] Open
Abstract
Myo-inositol (myo-Ins) has a physiological role in mammalian gametogenesis and embryonic development and a positive clinical impact on human medically assisted reproduction. We have previously shown that mouse embryo exposure to myo-Ins through preimplantation development in vitro increases proliferation activity and blastocyst production, representing an improvement in culture conditions. We have herein investigated biochemical mechanisms elicited by myo-Ins in preimplantation embryos and evaluated myo-Ins effects on postimplantation/postnatal development. To this end naturally fertilized embryos were cultured in vitro to blastocyst in the presence or absence of myo-Ins and analyzed for activation of the PKB/Akt pathway, known to modulate proliferation/survival cellular processes. In parallel, blastocyst-stage embryos were transferred into pseudopregnant females and allowed to develop to term and until weaning. Results obtained provide evidence that myo-Ins induces cellular pathways involving Akt and show that (a) exposure of preimplantation embryos to myo-Ins increases the number of blastocysts available for uterine transfer and of delivered animals and (b) the developmental patterns of mice obtained from embryos cultured in the presence or absence of myo-Ins, up to three weeks of age, overlap. These data further identify myo-Ins as a possibly important supplement for human preimplantation embryo culture in assisted reproduction technology.
Collapse
Affiliation(s)
- Nilay Kuşcu
- Department of Psychology, Section of Neuroscience, Sapienza University of Rome, Via dei Marsi 78, 00185 Rome, Italy
| | - Mariano Bizzarri
- Department of Experimental Medicine, Sapienza University of Rome, Viale Regina Elena 324, 00161 Rome, Italy
| | - Arturo Bevilacqua
- Department of Psychology, Section of Neuroscience, Sapienza University of Rome, Via dei Marsi 78, 00185 Rome, Italy
- Research Center in Neurobiology Daniel Bovet (CRiN), 00185 Rome, Italy
- *Arturo Bevilacqua:
| |
Collapse
|
28
|
Facchinetti F, Bizzarri M, Benvenga S, D’Anna R, Lanzone A, Soulage C, Di Renzo GC, Hod M, Cavalli P, Chiu TT, Kamenov ZA, Bevilacqua A, Carlomagno G, Gerli S, Oliva MM, Devroey P. Results from the International Consensus Conference on Myo-inositol and d-chiro-inositol in Obstetrics and Gynecology: the link between metabolic syndrome and PCOS. Eur J Obstet Gynecol Reprod Biol 2015; 195:72-76. [DOI: 10.1016/j.ejogrb.2015.09.024] [Citation(s) in RCA: 91] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Revised: 09/11/2015] [Accepted: 09/17/2015] [Indexed: 02/08/2023]
|
29
|
Intranasal and Intramuscular Administration of Lysine-Palmitoylated Peptide 612–627 of Thyroid-Stimulating Hormone Receptor Increases the Level of Thyroid Hormones in Rats. Int J Pept Res Ther 2015. [DOI: 10.1007/s10989-014-9452-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
|
30
|
Medici M, Visser WE, Visser TJ, Peeters RP. Genetic determination of the hypothalamic-pituitary-thyroid axis: where do we stand? Endocr Rev 2015; 36:214-44. [PMID: 25751422 DOI: 10.1210/er.2014-1081] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
For a long time it has been known that both hypo- and hyperthyroidism are associated with an increased risk of morbidity and mortality. In recent years, it has also become clear that minor variations in thyroid function, including subclinical dysfunction and variation in thyroid function within the reference range, can have important effects on clinical endpoints, such as bone mineral density, depression, metabolic syndrome, and cardiovascular mortality. Serum thyroid parameters show substantial interindividual variability, whereas the intraindividual variability lies within a narrow range. This suggests that every individual has a unique hypothalamus-pituitary-thyroid axis setpoint that is mainly determined by genetic factors, and this heritability has been estimated to be 40-60%. Various mutations in thyroid hormone pathway genes have been identified in persons with thyroid dysfunction or altered thyroid function tests. Because these causes are rare, many candidate gene and linkage studies have been performed over the years to identify more common variants (polymorphisms) associated with thyroid (dys)function, but only a limited number of consistent associations have been found. However, in the past 5 years, advances in genetic research have led to the identification of a large number of new candidate genes. In this review, we provide an overview of the current knowledge about the polygenic basis of thyroid (dys)function. This includes new candidate genes identified by genome-wide approaches, what insights these genes provide into the genetic basis of thyroid (dys)function, and which new techniques will help to further decipher the genetic basis of thyroid (dys)function in the near future.
Collapse
Affiliation(s)
- Marco Medici
- Rotterdam Thyroid Center, Department of Internal Medicine, Erasmus Medical Center, 3015 GE Rotterdam, The Netherlands
| | | | | | | |
Collapse
|
31
|
Leach K, Conigrave AD, Sexton PM, Christopoulos A. Towards tissue-specific pharmacology: insights from the calcium-sensing receptor as a paradigm for GPCR (patho)physiological bias. Trends Pharmacol Sci 2015; 36:215-25. [PMID: 25765207 DOI: 10.1016/j.tips.2015.02.004] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2014] [Revised: 02/05/2015] [Accepted: 02/09/2015] [Indexed: 12/25/2022]
Abstract
The calcium-sensing receptor (CaSR) is a widely expressed G protein-coupled receptor (GPCR) that mediates numerous tissue-specific functions. Its multiple ligands and diverse roles attest to the need for exquisite control over the signaling pathways that mediate its effects. 'Biased signaling' is the phenomenon by which distinct ligands stabilize preferred receptor signaling states. The CaSR is subject to biased signaling in response to its endogenous ligands. Interestingly, the 'natural' bias of the CaSR is altered in disease states, and small molecule drugs engender biased allosteric modulation of downstream signaling pathways. Thus, biased signaling from the CaSR also has important implications pathophysiologically and therapeutically. As outlined in this review, this novel paradigm extends to other GPCRs, making the CaSR a model for studies of ligand-biased signaling and for understanding how it may be used to foster selective drug activity in different tissues.
Collapse
Affiliation(s)
- Katie Leach
- Monash Institute of Pharmaceutical Sciences, 381 Royal Parade, Parkville VIC, Australia.
| | - Arthur D Conigrave
- School of Molecular Bioscience, Charles Perkins Centre, University of Sydney, NSW 2006, Australia
| | - Patrick M Sexton
- Monash Institute of Pharmaceutical Sciences, 381 Royal Parade, Parkville VIC, Australia
| | - Arthur Christopoulos
- Monash Institute of Pharmaceutical Sciences, 381 Royal Parade, Parkville VIC, Australia
| |
Collapse
|
32
|
Yang F, Huang H, Tao YX. Biased signaling in naturally occurring mutations in human melanocortin-3 receptor gene. Int J Biol Sci 2015; 11:423-33. [PMID: 25798062 PMCID: PMC4366641 DOI: 10.7150/ijbs.11032] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2014] [Accepted: 01/21/2015] [Indexed: 12/17/2022] Open
Abstract
The melanocortin-3 receptor (MC3R) is primarily expressed in the hypothalamus and plays an important role in the regulation of energy homeostasis. Recently, some studies demonstrated that MC3R also signals through mitogen-activated protein kinases (MAPKs), especially extracellular signal-regulated kinases 1 and 2 (ERK1/2). ERK1/2 signaling is known to alter gene expression, potentially contributing to the prolonged action of melanocortins on energy homeostasis regulation. In the present study, we performed detailed functional studies on 8 novel naturally occurring MC3R mutations recently reported, and the effects of endogenous MC3R agonist, α-melanocyte stimulating hormone (MSH), on ERK1/2 signaling on all 22 naturally occurring MC3R mutations reported to date. We found that mutants D158Y and L299V were potential pathogenic causes to obesity. Four residues, F82, D158, L249 and L299, played critical roles in different aspects of MC3R function. α-MSH exhibited balanced activity in Gs-cAMP and ERK1/2 signaling pathways in 15 of the 22 mutant MC3Rs. The other 7 mutant MC3Rs were biased to either one of the signaling pathways. In summary, we provided novel data about the structure-function relationship of MC3R, identifying residues important for receptor function. We also demonstrated that some mutations exhibited biased signaling, preferentially activating one intracellular signaling pathway, adding a new layer of complexity to MC3R pharmacology.
Collapse
Affiliation(s)
- Fan Yang
- 1. Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL, USA. ; 2. Current address: College of Life Sciences, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia 010018, China
| | - Hui Huang
- 1. Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL, USA
| | - Ya-Xiong Tao
- 1. Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL, USA
| |
Collapse
|
33
|
Abstract
Genetic defects of hormone receptors are the most common form of end-organ hormone resistance. One example of such defects is TSH resistance, which is caused by biallelic inactivating mutations in the TSH receptor gene (TSHR). TSH, a master regulator of thyroid functions, affects virtually all cellular processes involving thyroid hormone production, including thyroidal iodine uptake, thyroglobulin iodination, reuptake of iodinated thyroglobulin and thyroid cell growth. Resistance to TSH results in defective thyroid hormone production from the neonatal period, namely congenital hypothyroidism. Classically, clinical phenotypes of TSH resistance due to inactivating TSHR mutations were thought to vary depending on the residual mutant receptor activity. Nonfunctional mutations in the two alleles produce severe thyroid hypoplasia with overt hypothyroidism (uncompensated TSH resistance), while hypomorphic mutations in at least one allele produce normal-sized thyroid gland with preserved hormone-producing capacity (compensated TSH resistance). More recently, a new subgroup of TSH resistance (nonclassic TSH resistance) that is characterized by paradoxically high thyroidal iodine uptake has been reported. In this article, the pathophysiology and clinical features of TSH resistance due to inactivating TSHR mutations are reviewed, with particular attention to the nonclassic form.
Collapse
Affiliation(s)
- Satoshi Narumi
- Department of Pediatrics, Keio University School of Medicine, Tokyo, Japan
| | | |
Collapse
|
34
|
Brogi S, Tafi A, Désaubry L, Nebigil CG. Discovery of GPCR ligands for probing signal transduction pathways. Front Pharmacol 2014; 5:255. [PMID: 25506327 PMCID: PMC4246677 DOI: 10.3389/fphar.2014.00255] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2014] [Accepted: 11/02/2014] [Indexed: 01/11/2023] Open
Abstract
G protein-coupled receptors (GPCRs) are seven integral transmembrane proteins that are the primary targets of almost 30% of approved drugs and continue to represent a major focus of pharmaceutical research. All of GPCR targeted medicines were discovered by classical medicinal chemistry approaches. After the first GPCR crystal structures were determined, the docking screens using these structures lead to discovery of more novel and potent ligands. There are over 360 pharmaceutically relevant GPCRs in the human genome and to date about only 30 of structures have been determined. For these reasons, computational techniques such as homology modeling and molecular dynamics simulations have proven their usefulness to explore the structure and function of GPCRs. Furthermore, structure-based drug design and in silico screening (High Throughput Docking) are still the most common computational procedures in GPCRs drug discovery. Moreover, ligand-based methods such as three-dimensional quantitative structure–selectivity relationships, are the ideal molecular modeling approaches to rationalize the activity of tested GPCR ligands and identify novel GPCR ligands. In this review, we discuss the most recent advances for the computational approaches to effectively guide selectivity and affinity of ligands. We also describe novel approaches in medicinal chemistry, such as the development of biased agonists, allosteric modulators, and bivalent ligands for class A GPCRs. Furthermore, we highlight some knockout mice models in discovering biased signaling selectivity.
Collapse
Affiliation(s)
- Simone Brogi
- European Research Centre for Drug Discovery and Development (NatSynDrugs), University of Siena Siena, Italy ; Department of Biotechnology, Chemistry and Pharmacy, University of Siena Siena, Italy
| | - Andrea Tafi
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena Siena, Italy
| | - Laurent Désaubry
- Therapeutic Innovation Laboratory, UMR7200, CNRS/University of Strasbourg Illkirch, France
| | - Canan G Nebigil
- Receptor Signaling and Therapeutic Innovations, GPCR and Cardiovascular and Metabolic Regulations, Biotechnology and Cell Signaling Laboratory, UMR 7242, CNRS/University of Strasbourg - LabEx Medalis Illkirch, France
| |
Collapse
|
35
|
He S, Tao YX. Defect in MAPK signaling as a cause for monogenic obesity caused by inactivating mutations in the melanocortin-4 receptor gene. Int J Biol Sci 2014; 10:1128-37. [PMID: 25332687 PMCID: PMC4202029 DOI: 10.7150/ijbs.10359] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2014] [Accepted: 09/15/2014] [Indexed: 12/22/2022] Open
Abstract
The melanocortin-4 receptor (MC4R) is a Family A G protein-coupled receptor that plays an essential role in regulating energy homeostasis, including both energy intake and expenditure. Mutations leading to a reduced MC4R function confer a major gene effect for obesity. More than 170 distinct mutations have been identified in humans. In addition to the conventional Gs-stimulated cAMP pathway, the MC4R also activates MAPKs, especially ERK1/2. We also showed there is biased signaling in the two signaling pathways, with inverse agonists in the Gs-cAMP pathway acting as agonists for the ERK1/2 pathway. In the current study, we sought to determine whether defects in basal or agonist-induced ERK1/2 activation in MC4R mutants might potentially contribute to obesity pathogenesis in patients carrying these mutations. The constitutive and ligand-stimulated ERK1/2 activation were measured in wild type and 73 naturally occurring MC4R mutations. We showed that nineteen mutants had significantly decreased basal pERK1/2 level, and five Class V variants (where no functional defects have been identified previously), C40R, V50M, T112M, A154D and S295P, had impaired ligand-stimulated ERK1/2 activation. Our studies demonstrated for the first time that decreased basal or ligand-stimulated ERK1/2 signaling might contribute to obesity pathogenesis caused by mutations in the MC4R gene. We also observed biased signaling in 25 naturally occurring mutations in the Gs-cAMP and ERK1/2 pathways.
Collapse
Affiliation(s)
- Shan He
- 1. Department of Anatomy, Physiology, and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, Alabama 36849-5519, USA. ; 2. Key Laboratory of Freshwater Animal Breeding, Ministry of Agriculture, College of Fisheries, Huazhong Agricultural University, Hubei Collaborative Innovation Center for Freshwater Aquaculture, Wuhan, Hubei 430070, China
| | - Ya-Xiong Tao
- 1. Department of Anatomy, Physiology, and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, Alabama 36849-5519, USA
| |
Collapse
|
36
|
Sbai O, Monnier C, Dodé C, Pin JP, Hardelin JP, Rondard P. Biased signaling through G-protein-coupled PROKR2 receptors harboring missense mutations. FASEB J 2014; 28:3734-44. [PMID: 24830383 DOI: 10.1096/fj.13-243402] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Various missense mutations in the gene coding for prokineticin receptor 2 (PROKR2), a G-protein-coupled receptor, have been identified in patients with Kallmann syndrome. However, the functional consequences of these mutations on the different signaling pathways of this receptor have not been studied. We first showed that the wild-type PROKR2 can activate different G-protein subtypes (Gq, Gs, and Gi/o) and recruit β-arrestins in transfected HEK-293 cells. We then examined, for each of these signaling pathways, the effects of 9 mutations that did not significantly impair cell surface targeting or ligand binding of the receptor. Four mutant receptors showing defective Gq signaling (R85C, R85H, R164Q, and V331M) could still recruit β-arrestins on ligand activation, which may cause biased signaling in vivo. Conversely, the R80C receptor could activate the 3 types of G proteins but could not recruit β-arrestins. Finally, the R268C receptor could recruit β-arrestins and activate the Gq and Gs signaling pathways but could not activate the Gi/o signaling pathway. Our results validate the concept that mutations in the genes encoding membrane receptors can bias downstream signaling in various ways, possibly leading to pathogenic and, perhaps in some cases, protective (e.g., R268C) effects.
Collapse
Affiliation(s)
- Oualid Sbai
- Centre National de la Recherche Scientifique (CNRS) Unité Mixte de Recherche (UMR) 5203, Institut de Génomique Fonctionnelle, Montpellier, France; Institut National de la Santé et de la Recherche Médicale (INSERM) U661, Montpellier, France; Université Montpellier 1 and 2, Montpellier, France
| | - Carine Monnier
- Centre National de la Recherche Scientifique (CNRS) Unité Mixte de Recherche (UMR) 5203, Institut de Génomique Fonctionnelle, Montpellier, France; Institut National de la Santé et de la Recherche Médicale (INSERM) U661, Montpellier, France; Université Montpellier 1 and 2, Montpellier, France
| | - Catherine Dodé
- EA7331, Faculté des Sciences Pharmaceutiques et Biologiques, Université Paris-Descartes, Paris, France; and
| | - Jean-Philippe Pin
- Centre National de la Recherche Scientifique (CNRS) Unité Mixte de Recherche (UMR) 5203, Institut de Génomique Fonctionnelle, Montpellier, France; Institut National de la Santé et de la Recherche Médicale (INSERM) U661, Montpellier, France; Université Montpellier 1 and 2, Montpellier, France
| | - Jean-Pierre Hardelin
- INSERM Unité Mixte de Recherche en Santé (UMRS) 1120, Département Neuroscience, Institut Pasteur, Paris, France
| | - Philippe Rondard
- Centre National de la Recherche Scientifique (CNRS) Unité Mixte de Recherche (UMR) 5203, Institut de Génomique Fonctionnelle, Montpellier, France; Institut National de la Santé et de la Recherche Médicale (INSERM) U661, Montpellier, France; Université Montpellier 1 and 2, Montpellier, France;
| |
Collapse
|
37
|
Kleinau G, Biebermann H. Constitutive activities in the thyrotropin receptor: regulation and significance. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2014; 70:81-119. [PMID: 24931193 DOI: 10.1016/b978-0-12-417197-8.00003-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
The thyroid-stimulating hormone receptor (TSHR, or thyrotropin receptor) is a family A G protein-coupled receptor. It not only binds thyroid-stimulating hormone (TSH, or thyrotropin) but also interacts with autoantibodies under pathological conditions. The TSHR and TSH are essential for thyroid growth and function and thus for all thyroid hormone-associated physiological superordinated processes, including metabolism and development of the central nervous system. In vitro studies have found that the TSHR permanently stimulates ligand-independent (constitutive) activation of Gs, which ultimately leads to intracellular cAMP accumulation. Furthermore, a vast variety of constitutively activating mutations of TSHR-at more than 50 different amino acid positions-have been reported to enhance basal signaling. These lead in vivo to a "gain-of-function" phenotype of nonautoimmune hyperthyroidism or toxic adenomas. Moreover, many naturally occurring inactivating mutations are known to cause a "loss-of-function" phenotype, resulting in resistance to thyroid hormone or hyperthyrotropinemia. Several of these mutations are also characterized by impaired basal signaling, and these are designated here as "constitutively inactivating mutations" (CIMs). More than 30 amino acid positions with CIMs have been identified so far. Moreover, the permanent TSHR signaling capacity can also be blocked by inverse agonistic antibodies or small drug-like molecules, which both have a potential for clinical usage. In this chapter, information on constitutive activity in the TSHR is described, including up- and downregulation, linked protein conformations, physiological and pathophysiological conditions, and related intracellular signaling.
Collapse
Affiliation(s)
- Gunnar Kleinau
- Institute of Experimental Pediatric Endocrinology, Charité-Universitätsmedizin Berlin, Berlin, Germany.
| | - Heike Biebermann
- Institute of Experimental Pediatric Endocrinology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
38
|
Combined treatment with Myo-inositol and selenium ensures euthyroidism in subclinical hypothyroidism patients with autoimmune thyroiditis. J Thyroid Res 2013; 2013:424163. [PMID: 24224112 PMCID: PMC3809375 DOI: 10.1155/2013/424163] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2013] [Revised: 08/27/2013] [Accepted: 08/27/2013] [Indexed: 12/13/2022] Open
Abstract
Background. Hashimoto's thyroiditis (HT), also known as chronic lymphocytic thyroiditis or chronic autoimmune thyroiditis, is the most common form of thyroiditis affecting more than 10% of females and 2% of males. The present study aims to evaluate the beneficial effect of a combined treatment, Myo-Inositol plus selenomethionine, on subclinical hypothyroidism. Methods. The study was designed as a double-blind randomized controlled trial. Eligible patients were women diagnosed with subclinical hypothyroidism having Tg antibodies (TgAb) titer higher than 350 IU/mL. Outcome measures were Thyroid Stimulating Hormone (TSH) levels, thyroid peroxidase antibodies (TPOAb) and TgAb titer, selenium, and Myo-Inositol plasma concentration. Results. In the present paper, we demonstrated that the beneficial effects obtained by selenomethionine treatment on patients affected by subclinical hypothyroidism, likely due to the presence of autoantibody (TPOAb and TgAb), are further improved by cotreatment with Myo-Inositol. Conclusions. Indeed, due to its action as TSH second messenger, Myo-Inositol treatment reduces TSH levels closer to physiological concentrations.
Collapse
|
39
|
Kleinau G, Neumann S, Grüters A, Krude H, Biebermann H. Novel insights on thyroid-stimulating hormone receptor signal transduction. Endocr Rev 2013; 34:691-724. [PMID: 23645907 PMCID: PMC3785642 DOI: 10.1210/er.2012-1072] [Citation(s) in RCA: 91] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The TSH receptor (TSHR) is a member of the glycoprotein hormone receptors, a subfamily of family A G protein-coupled receptors. The TSHR is of great importance for the growth and function of the thyroid gland. The TSHR and its endogenous ligand TSH are pivotal proteins with respect to a variety of physiological functions and malfunctions. The molecular events of TSHR regulation can be summarized as a process of signal transduction, including signal reception, conversion, and amplification. The steps during signal transduction from the extra- to the intracellular sites of the cell are not yet comprehensively understood. However, essential new insights have been achieved in recent years on the interrelated mechanisms at the extracellular region, the transmembrane domain, and intracellular components. This review contains a critical summary of available knowledge of the molecular mechanisms of signal transduction at the TSHR, for example, the key amino acids involved in hormone binding or in the structural conformational changes that lead to G protein activation or signaling regulation. Aspects of TSHR oligomerization, signaling promiscuity, signaling selectivity, phenotypes of genetic variations, and potential extrathyroidal receptor activity are also considered, because these are relevant to an understanding of the overall function of the TSHR, including physiological, pathophysiological, and pharmacological perspectives. Directions for future research are discussed.
Collapse
Affiliation(s)
- Gunnar Kleinau
- Institute of Experimental Pediatric Endocrinology, Charité-Universitätsmedizin Berlin, Ostring 3, Augustenburger Platz 1, 13353 Berlin, Germany.
| | | | | | | | | |
Collapse
|
40
|
Moia S, Godi M, Walker GE, Roccio M, Agretti P, Tonacchera M, Berardi R, Bellone S, Prodam F, Giordano M, Bona G. The W520X mutation in the TSHR gene brings on subclinical hypothyroidism through an haploinsufficiency mechanism. J Endocrinol Invest 2013; 36:716-21. [PMID: 23563316 DOI: 10.3275/8930] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
BACKGROUND TSHR is a G-protein-coupled seven transmembrane domain receptor that activates the two major signal transduction pathways: the Gαs/adenylate cyclase and the Gαq/11/phospholipase C pathways. Inactivating mutations in the TSHR gene have been demonstrated to be responsible for subclinical hypothyroidism, a disorder characterized by elevated serum TSH concentrations despite normal thyroid hormones levels. AIM We identified in a child a nonsense mutation (W520X) in the third transmembrane domain of the TSHR that causes the lack of the C-terminus portion of the receptor. The functional significance of this variation was assessed in vitro. MATERIAL/SUBJECT AND METHODS The W520X mutation was introduced into the pSVL vector containing the wild-type sequence of TSHR gene. Wild-type and mutated vectors were expressed in Chinese Hamster Ovary (CHO) cells, and cAMP, inositol phosphate (IP), immunofluorescence and FACS analyses were performed. RESULTS Transfection with pSVL-TSHR vector induced basal cAMP and IP production in the absence of TSH stimulation, indicating a constitutive activity for the TSHR. An impairment of receptor function was demonstrated by the observation that cells expressing the mutant TSHR exhibited a lower second messenger production with respect to the wild-type, despite a normal expression of the receptor at the cell surface. CONCLUSIONS The mechanism through which the W520X mutation exerts its effect is more likely haploinsufficiency rather than a dominant-negative effect. This could explain the phenotype of our patient, who has a hormonal pattern in the range of a mild subclinical hypothyroidism, without an overt disease phenotype.
Collapse
Affiliation(s)
- S Moia
- Division of Pediatrics, Department of Health Sciences, University of Piemonte Orientale, Via Solaroli 17, 28100 Novara, Italy.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Nettore IC, Cacace V, De Fusco C, Colao A, Macchia PE. The molecular causes of thyroid dysgenesis: a systematic review. J Endocrinol Invest 2013; 36:654-64. [PMID: 23698639 DOI: 10.3275/8973] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
BACKGROUND Congenital hypothyroidism (CH) is a frequent disease occurring with an incidence of about 1/2500 newborns/year. In 80-85% of the cases CH is caused by alterations in thyroid morphogenesis, generally indicated by the term "thyroid dysgenesis" (TD). TD is generally a sporadic disease, but in about 5% of the cases a genetic origin has been demonstrated. In these cases, mutations in genes playing a role during thyroid morphogenesis (NKX2-1, PAX8, FOXE1, NKX2-5, TSHR) have been reported. AIM This work reviews the main steps of thyroid morphogenesis and all the genetic alterations associated with TD and published in the literature.
Collapse
Affiliation(s)
- I C Nettore
- Department of Clinical Medicine and Surgery, University of Naples "Federico II", Via S. Pansini, 5 - 80131 Naples, Italy
| | | | | | | | | |
Collapse
|
42
|
Duranton F, Lacoste A, Faurous P, Deshayes E, Ribstein J, Avignon A, Mourad G, Argilés À. Exogenous thyrotropin improves renal function in euthyroid patients, while serum creatinine levels are increased in hypothyroidism. Clin Kidney J 2013; 6:478-83. [PMID: 26064512 PMCID: PMC4438406 DOI: 10.1093/ckj/sft092] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2013] [Accepted: 07/09/2013] [Indexed: 11/22/2022] Open
Abstract
Background There is evidence showing that the hypothyroid state results in increased serum creatinine levels. However, whether this is only due to the peripheral thyroid hormones or if thyroid-stimulating hormone (TSH) is also involved is not known. Methods Serum creatinine levels and estimated glomerular filtration rate (eGFR) were assessed in thyroidectomized patients with varying thyroid hormones and TSH levels. Blood samples from Group 1 (21 patients) were obtained 1 month after complete thyroidectomy, while under a hypothyroid state (t1) and a sufficient time after thyroid hormones initiation (euthyroid state, t2). Group 2 (20 euthyroid patients) were sampled after recombinant human thyrotropin injections (rhTSH, t1) and later after rhTSH extinction (t2). Results In Group 1, serum creatinine levels decreased after correction of hypothyroidism (85.3 ± 4.3 versus 78.0 ± 3.9 µmol/L; P = 0.04). In Group 2, serum creatinine levels increased after rhTSH withdrawal (70.6 ± 5.7 µmol/L versus 76.5 ± 5.8 µmol/L; P = 0.007). Between t1 and t2, eGFR varied accordingly [Group 1, 71.7 ± 3.5 versus 81.2 ± 4.5 mL/min/1.73 m² (P = 0.02); Group 2, 97.7 ± 7.4 versus 87.5 ± 5.9 (P = 0.007)]. The changes in TSH and eGFR following supplementation with thyroxine were significantly correlated (r = −0.6, P = 0.0041). Conclusions Iatrogenic hypothyroidism significantly increases serum creatinine and reversibly impairs eGFR, while treatment with rhTSH enhances renal function in euthyroid patients, supporting the existence of an influence of TSH level on renal function. The mechanisms by which peripheral thyroid hormones and TSH influence GFR need to be identified in physiology-orientated studies.
Collapse
Affiliation(s)
| | - Anouchka Lacoste
- Service de Néphrologie, Dialyse et Transplantation, Hôpital Lapeyronie, Université de Montpellier, Montpellier , France
| | | | | | - Jean Ribstein
- Service de Médecine Interne , Hôpital Lapeyronie, Université de Montpellier, Montpellier , France
| | - Antoine Avignon
- Service de Nutrition-Diabète, Hôpital Lapeyronie, Université de Montpellier, Montpellier , France
| | - Georges Mourad
- Service de Néphrologie, Dialyse et Transplantation, Hôpital Lapeyronie, Université de Montpellier, Montpellier , France
| | - Àngel Argilés
- SAS RD - Néphrologie, Montpellier , France ; Service de Néphrologie, Dialyse et Transplantation, Hôpital Lapeyronie, Université de Montpellier, Montpellier , France ; Néphrologie Dialyse St Guilhem, Sète , France
| |
Collapse
|
43
|
Ca(2+)-binding protein expression in primary human thyrocytes. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2013; 1833:2703-2713. [PMID: 23886630 DOI: 10.1016/j.bbamcr.2013.07.014] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/01/2013] [Revised: 06/04/2013] [Accepted: 07/15/2013] [Indexed: 01/06/2023]
Abstract
We recently identified several Ca(2+)-binding proteins (CaBP) from the S100 and annexin family to be regulated by TSH in FRTL-5 cells. Here, we study the regulation of S100A4, S100A6 and ANXA2 in primary human thyrocytes (PHT) derived from surrounding tissues (ST), cold benign thyroid nodules (CTN) and autonomously functioning thyroid nodules (AFTN). We investigated the expression and regulation of CaBP and the effect of their expression on Ca(2+) and TSHR signaling. We used an approach that accounts for the potential of an individual PHT culture to proliferate or to express thyroid differentiation features by assessing the expression of FOS and TPO. We found a strong correlation between the regulation of CaBP and the proliferation-associated transcription factor gene FOS. PKA and MEK1/2 were regulators of ANXA2 expression, while PI3-K and triiodothyronine were additionally involved in S100 regulation. The modulated expression of CaBP was reflected by changes in ATP-elicited Ca(2+) signaling in PHT. S100A4 increased the ratio of subsequent Ca(2+) responses and showed a Ca(2+) buffering effect, while ANXA2 affected the first Ca(2+) response to ATP. Overexpression of S100A4 led to a reduced activation of NFAT by TSH. Using S100A4 E33Q, D63N, F72Q and Y75K mutants we found that the effects of S100A4 expression on Ca(2+) signaling are mediated by protein interaction. We present evidence that TSH has the ability to fine-tune Ca(2+) signals through the regulation of CaBP expression. This represents a novel putative cross-regulating mechanism in thyrocytes that could affect thyrocyte signaling and physiology.
Collapse
|
44
|
Armstrong SP, Seeber RM, Ayoub MA, Feldman BJ, Pfleger KDG. Characterization of three vasopressin receptor 2 variants: an apparent polymorphism (V266A) and two loss-of-function mutations (R181C and M311V). PLoS One 2013; 8:e65885. [PMID: 23762448 PMCID: PMC3675069 DOI: 10.1371/journal.pone.0065885] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2013] [Accepted: 04/30/2013] [Indexed: 02/01/2023] Open
Abstract
Arginine vasopressin (AVP) is released from the posterior pituitary and controls water homeostasis. AVP binding to vasopressin V2 receptors (V2Rs) located on kidney collecting duct epithelial cells triggers activation of Gs proteins, leading to increased cAMP levels, trafficking of aquaporin-2 water channels, and consequent increased water permeability and antidiuresis. Typically, loss-of-function V2R mutations cause nephrogenic diabetes insipidus (NDI), whereas gain-of-function mutations cause nephrogenic syndrome of inappropriate antidiuresis (NSIAD). Here we provide further characterization of two mutant V2Rs, R181C and M311V, reported to cause complete and partial NDI respectively, together with a V266A variant, in a patient diagnosed with NSIAD. Our data in HEK293FT cells revealed that for cAMP accumulation, AVP was about 500- or 30-fold less potent at the R181C and M311V mutants than at the wild-type receptor respectively (and about 4000- and 60-fold in COS7 cells respectively). However, in contrast to wild type V2R, the R181C mutant failed to increase inositol phosphate production, while with the M311V mutant, AVP exhibited only partial agonism in addition to a 37-fold potency decrease. Similar responses were detected in a BRET assay for β-arrestin recruitment, with the R181C receptor unresponsive to AVP, and partial agonism with a 23-fold decrease in potency observed with M311V in both HEK293FT and COS7 cells. Notably, the V266A V2R appeared functionally identical to the wild-type receptor in all assays tested, including cAMP and inositol phosphate accumulation, β-arrestin interaction, and in a BRET assay of receptor ubiquitination. Each receptor was expressed at comparable levels. Hence, the M311V V2R retains greater activity than the R181C mutant, consistent with the milder phenotype of NDI associated with this mutant. Notably, the R181C mutant appears to be a Gs protein-biased receptor incapable of signaling to inositol phosphate or recruiting β-arrestin. The etiology of NSIAD in the patient with V266A V2R remains unknown.
Collapse
MESH Headings
- Animals
- Aquaporin 2/genetics
- Aquaporin 2/metabolism
- Arginine Vasopressin/metabolism
- Arrestins/genetics
- Arrestins/metabolism
- COS Cells
- Chlorocebus aethiops
- Cyclic AMP/metabolism
- Diabetes Insipidus, Nephrogenic/genetics
- Diabetes Insipidus, Nephrogenic/metabolism
- Diabetes Insipidus, Nephrogenic/pathology
- GTP-Binding Protein alpha Subunits, Gs/genetics
- GTP-Binding Protein alpha Subunits, Gs/metabolism
- Gene Expression Regulation
- Genetic Diseases, X-Linked/genetics
- Genetic Diseases, X-Linked/metabolism
- Genetic Diseases, X-Linked/pathology
- HEK293 Cells
- Humans
- Inappropriate ADH Syndrome/genetics
- Inappropriate ADH Syndrome/metabolism
- Inappropriate ADH Syndrome/pathology
- Inositol Phosphates/metabolism
- Mutation
- Polymorphism, Genetic
- Receptors, Vasopressin/genetics
- Receptors, Vasopressin/metabolism
- Signal Transduction
- beta-Arrestins
Collapse
Affiliation(s)
- Stephen P. Armstrong
- Laboratory for Molecular Endocrinology-G Protein-Coupled Receptors, Western Australian Institute for Medical Research and Centre for Medical Research, The University of Western Australia, Nedlands, Perth, Western Australia, Australia
| | - Ruth M. Seeber
- Laboratory for Molecular Endocrinology-G Protein-Coupled Receptors, Western Australian Institute for Medical Research and Centre for Medical Research, The University of Western Australia, Nedlands, Perth, Western Australia, Australia
| | - Mohammed Akli Ayoub
- Laboratory for Molecular Endocrinology-G Protein-Coupled Receptors, Western Australian Institute for Medical Research and Centre for Medical Research, The University of Western Australia, Nedlands, Perth, Western Australia, Australia
- Protein Research Chair - Department of Biochemistry, College of Science, King Saud University, Riyadh, Kingdom of Saudi Arabia
| | - Brian J. Feldman
- Pediatric Endocrinology, Department of Pediatrics, Stanford University, Stanford, California, United States of America
| | - Kevin D. G. Pfleger
- Laboratory for Molecular Endocrinology-G Protein-Coupled Receptors, Western Australian Institute for Medical Research and Centre for Medical Research, The University of Western Australia, Nedlands, Perth, Western Australia, Australia
- * E-mail:
| |
Collapse
|
45
|
Cassio A, Nicoletti A, Rizzello A, Zazzetta E, Bal M, Baldazzi L. Current loss-of-function mutations in the thyrotropin receptor gene: when to investigate, clinical effects, and treatment. J Clin Res Pediatr Endocrinol 2013; 5 Suppl 1:29-39. [PMID: 23154162 PMCID: PMC3608004 DOI: 10.4274/jcrpe.864] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
Abstract
Thyroid-stimulating hormone receptor (TSHR) loss-of-function (LOF) mutations lead to a wide spectrum of phenotypes, ranging from severe congenital hypothyroidism (CH) to mild euthyroid hyperthyrotropinemia. The degree of TSH resistance depends on the severity of the impairment of the receptor function caused by the mutation and on the number of mutated alleles In this review data about genotype-phenotype correlation and criteria for clinical work-up will be presented and discussed. Complete TSH resistance due to biallelic LOF TSHR mutations must be suspected in all patients with severe not syndromic CH and severe thyroid hypoplasia diagnosed at birth by neonatal screening. Partial forms of TSH resistance show a more heterogeneous hormonal and clinical pattern . In these cases TSH serum levels are above the upper limit of normal range for the age but with a very variable pattern, free thyroxine (T4) concentrations are within the normal range and thyroid size can be normal or hypoplastic at ultrasound scan. An early substitutive treatment with L-T4 must be mandatory in all patients with severe CH due to complete uncompensated TSH resistance diagnosed at birth by neonatal screening. The usefulness of substitutive treatment appears much more controversial inpatients with subclinical hypothyroidism due to partial TSH resistance in whom the increased TSH concentration should be able to compensate the mild functional impairment of the mutant receptor. Together with standard criteria we recommend also an accurate clinical work-up to select patients who are candidates for a LOF TSHR mutation.
Collapse
Affiliation(s)
- Alessandra Cassio
- Department of Gynaecologic, Obstetric and Paediatric Sciences, S Orsola-Malpighi Hospital, University of Bologna, Bologna, Italy.
| | - Annalisa Nicoletti
- Department of Gynaecologic, Obstetric and Paediatric Sciences, S.Orsola-Malpighi Hospital, University of Bologna, Bologna, Italy
| | - Angela Rizzello
- Department of Gynaecologic, Obstetric and Paediatric Sciences, S.Orsola-Malpighi Hospital, University of Bologna, Bologna, Italy
| | - Emanuela Zazzetta
- Department of Gynaecologic, Obstetric and Paediatric Sciences, S.Orsola-Malpighi Hospital, University of Bologna, Bologna, Italy
| | - Milva Bal
- Department of Gynaecologic, Obstetric and Paediatric Sciences, S.Orsola-Malpighi Hospital, University of Bologna, Bologna, Italy
| | - Lilia Baldazzi
- Department of Gynaecologic, Obstetric and Paediatric Sciences, S.Orsola-Malpighi Hospital, University of Bologna, Bologna, Italy
| |
Collapse
|
46
|
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: 170] [Impact Index Per Article: 14.2] [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
|
47
|
Bäck CM, Stohr S, Schäfer EAM, Biebermann H, Boekhoff I, Breit A, Gudermann T, Büch TRH. TSH induces metallothionein 1 in thyrocytes via Gq/11- and PKC-dependent signaling. J Mol Endocrinol 2013; 51:79-90. [PMID: 23613280 DOI: 10.1530/jme-12-0200] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Metallothioneins (MTs) are cytoprotective proteins acting as scavengers of toxic metal ions or reactive oxygen species. MTs are upregulated in follicular thyroid carcinoma and are regarded as a marker of thyroid stress in Graves' disease. However, the mechanism of MT regulation in thyrocytes is still elusive. In other cellular systems, cAMP-, calcium-, or protein kinase C (PKC)-dependent signaling cascades have been shown to induce MT expression. Of note, all of these three pathways are activated following the stimulation of the TSH receptor (TSHR). Thus, we hypothesized that TSH represents a key regulator of MT expression in thyrocytes. In fact, TSHR stimulation induced expression of MT isoform 1X (MT1X) in human follicular carcinoma cells. In these cells, Induction of MT1X expression critically relied on intact Gq/11 signaling of the TSHR and was blocked by chelation of intracellular calcium and inhibition of PKC. TSHR-independent stimulation of cAMP formation by treating cells with forskolin also led to an upregulation of MT1X, which was completely dependent on PKA. However, inhibition of PKA did not affect the regulation of MT1X by TSH. As in follicular thyroid carcinoma cells, TSH also induced MT1 protein in primary human thyrocytes, which was PKC dependent as well. In summary, these findings indicate that TSH stimulation induces MT1X expression via Gq/11 and PKC, whereas cAMP-PKA signaling does not play a predominant role. To date, little has been known regarding cAMP-independent effects of TSHR signaling. Our findings extend the knowledge about the PKC-mediated functions of the TSHR.
Collapse
Affiliation(s)
- Christer M Bäck
- Medical Department III, University Hospital Aachen, D-52074 Aachen, Germany
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Sukumaran P, Löf C, Kemppainen K, Kankaanpää P, Pulli I, Näsman J, Viitanen T, Törnquist K. Canonical transient receptor potential channel 2 (TRPC2) as a major regulator of calcium homeostasis in rat thyroid FRTL-5 cells: importance of protein kinase C δ (PKCδ) and stromal interaction molecule 2 (STIM2). J Biol Chem 2012; 287:44345-60. [PMID: 23144458 DOI: 10.1074/jbc.m112.374348] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Mammalian non-selective transient receptor potential cation channels (TRPCs) are important in the regulation of cellular calcium homeostasis. In thyroid cells, including rat thyroid FRTL-5 cells, calcium regulates a multitude of processes. RT-PCR screening of FRTL-5 cells revealed the presence of TRPC2 channels only. Knockdown of TRPC2 using shRNA (shTRPC2) resulted in decreased ATP-evoked calcium peak amplitude and inward current. In calcium-free buffer, there was no difference in the ATP-evoked calcium peak amplitude between control cells and shTRPC2 cells. Store-operated calcium entry was indistinguishable between the two cell lines. Basal calcium entry was enhanced in shTRPC2 cells, whereas the level of PKCβ1 and PKCδ, the activity of sarco/endoplasmic reticulum Ca(2+)-ATPase, and the calcium content in the endoplasmic reticulum were decreased. Stromal interaction molecule (STIM) 2, but not STIM1, was arranged in puncta in resting shTRPC2 cells but not in control cells. Phosphorylation site Orai1 S27A/S30A mutant and non-functional Orai1 R91W attenuated basal calcium entry in shTRPC2 cells. Knockdown of PKCδ with siRNA increased STIM2 punctum formation and enhanced basal calcium entry but decreased sarco/endoplasmic reticulum Ca(2+)-ATPase activity in wild-type cells. Transfection of a truncated, non-conducting mutant of TRPC2 evoked similar results. Thus, TRPC2 functions as a major regulator of calcium homeostasis in rat thyroid cells.
Collapse
Affiliation(s)
- Pramod Sukumaran
- Department of Biosciences, Åbo Akademi University, Tykistökatu 6A, 20520 Turku, Finland
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Löf C, Sukumaran P, Viitanen T, Vainio M, Kemppainen K, Pulli I, Näsman J, Kukkonen JP, Törnquist K. Communication between the calcium and cAMP pathways regulate the expression of the TSH receptor: TRPC2 in the center of action. Mol Endocrinol 2012; 26:2046-57. [PMID: 23015753 DOI: 10.1210/me.2012-1171] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Transient receptor potential (TRP) cation channels are widely expressed and function in many physiologically important processes. Perturbations in the expression or mutations of the channels have implications for diseases. Many thyroid disorders, as excessive growth or disturbed thyroid hormone production, can be a result of dysregulated TSH signaling. In the present study, we found that of TRP canonicals (TRPCs), only TRPC2 was expressed in Fischer rat thyroid low-serum 5% cells (FRTL-5 cells). To investigate the physiological importance of the channel, we developed stable TRPC2 knockdown cells using short hairpin RNA (shTRPC2 cells). In these cells, the ATP-evoked entry of calcium was significantly decreased. This led to increased cAMP production, because inhibitory signals from calcium to adenylate cyclase 5/6 were decreased. Enhanced cAMP signaling projected to Ras-related protein 1-MAPK kinase 1 (MAPK/ERK kinase 1) pathway leading to phosphorylation of ERK1/2. The activated ERK1/2 pathway increased the expression of the TSH receptor. In contrast, secretion of thyroglobulin was decreased in shTRPC2 cells, due to improper folding and glycosylation of the protein. We show here a novel role for TRPC2 in regulating thyroid cell function.
Collapse
Affiliation(s)
- Christoffer Löf
- Department of Biosciences, Åbo Akademi University, Biocity, Tykistökatu 6A, 20520 Turku, Finland
| | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Sriphrapradang C, German A, Dumitrescu AM, Refetoff S. Consecutive mutational events in a TSHR allele of Arab families with resistance to thyroid stimulating hormone. Thyroid 2012; 22:252-7. [PMID: 22313426 PMCID: PMC3286805 DOI: 10.1089/thy.2011.0402] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
Abstract
BACKGROUND Our laboratory identified six distinct inactivating TSHR gene mutations in Arab tribes living in Israel. We recently reported three nucleotide substitutions in exon 3 producing p.[L89L;Q90P] and one in exon 9 of the same allele producing p.P264S in Family A. Family B, reported herein, harbors the identical mutation in exon 3 only. We set to determine whether the mutations have common ancestral origin. METHODS Coding regions of the TSHR were sequenced and flanking microsatellite markers spanning 5.3 cM were used for haplotyping. RESULTS Two siblings of Family B were compound heterozygous for TSHR gene mutations. The paternal allele contained the exon 3 mutation and the maternal allele harbored a mutation in exon 10 (p.L653V). We investigated the possibility of a founder effect with subsequent mutational events for the presence of the same exon 3 mutation in different families. The haplotype of the allele harboring the exon 3 mutation in Family B was identical to that of Family A, also harboring the exon 9 mutation on the same allele, indicating that the latter occurred subsequently. The ancestral wild-type TSHR was present in Family B, suggesting that the mutation in exon 3 was also new in the history of that population. CONCLUSIONS It is more likely that two consecutive mutational events occurred on the ancestral wild-type allele instead of a recombination bringing exon 3 and exon 9 mutations together on the same allele. New mutational events contribute to the high prevalence of TSHR mutations in this population in addition to a founder effect and limited gene pool due to inbreeding.
Collapse
Affiliation(s)
| | - Alina German
- Pediatrics Endocrine Unit, Clalit Health Maintenance Organization, Haifa, Israel
| | | | - Samuel Refetoff
- Department of Medicine, The University of Chicago, Chicago, Illinois
- Department of Pediatrics, The University of Chicago, Chicago, Illinois
- Committee on Genetics, The University of Chicago, Chicago, Illinois
| |
Collapse
|