1
|
Carro GH, Martín M, Savy S, Peyret V, Geysels RC, Montes FA, Bernal Barquero CE, Ricci V, Masnata ME, Masini-Repiso AM, Papendieck P, Tellechea ML, Chiesa AE, Nicola JP. Functional characterization of novel compound heterozygous missense SLC5A5 gene variants causing congenital dyshormonogenic hypothyroidism. Front Endocrinol (Lausanne) 2024; 15:1465176. [PMID: 39749016 PMCID: PMC11693440 DOI: 10.3389/fendo.2024.1465176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Accepted: 11/04/2024] [Indexed: 01/04/2025] Open
Abstract
Introduction The sodium/iodide symporter (NIS) mediates active iodide accumulation in the thyroid follicular cell. Biallelic loss-of-function variants in the NIS-coding SLC5A5 gene cause congenital dyshormonogenic hypothyroidism due to a defect in the accumulation of iodide, which is required for thyroid hormonogenesis. Objective We aimed to identify, and if so to functionally characterize, novel pathogenic SLC5A5 gene variants in a patient diagnosed with severe congenital dyshormonogenic hypothyroidism characterized by undetectable radioiodide accumulation in a eutopic thyroid gland, as well as in the salivary glands. Methods The coding region of the SLC5A5 gene was sequenced using whole-exome sequencing. In silico analysis and in vitro functional characterization of missense SLC5A5 gene variants were performed. Results Proposita's whole-exome sequencing revealed a novel pair of compound heterozygous missense variants in the SLC5A5 gene, c.1,627G>A (p.G543R) and c.1,684T>A (p.L562M). The parents were heterozygous carriers of the variants as determined by Sanger sequencing of the SLC5A5 gene. The p.G543R variant in the homozygous state has previously been associated with congenital hypothyroidism. The novel p.L562M variant was not reported in the Genome Aggregation Consortium dataset. In silico analysis of the pathogenic impact of the p.L562M variant yielded inconclusive results. Functional in vitro studies showed that the p.L562M variant reduces iodide accumulation due to defective expression of the mutant NIS protein at the plasma membrane. Notably, the aliphatic residue Leu at position 562 in the carboxy terminus of the protein, which is highly conserved in NIS orthologues, is required for NIS plasma membrane expression. Conclusions We report novel compound heterozygous missense SLC5A5 gene variants causing defective iodide accumulation, thus leading to congenital dyshormonogenic hypothyroidism.
Collapse
Affiliation(s)
- Gerardo Hernán Carro
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
- Centro de Investigaciones en Bioquímica Clínica e Inmunología, Consejo Nacional de Investigaciones Científicas y Técnicas (CIBICI-CONICET), Córdoba, Argentina
| | - Mariano Martín
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
- Centro de Investigaciones en Bioquímica Clínica e Inmunología, Consejo Nacional de Investigaciones Científicas y Técnicas (CIBICI-CONICET), Córdoba, Argentina
| | - Sofía Savy
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
- Centro de Investigaciones en Bioquímica Clínica e Inmunología, Consejo Nacional de Investigaciones Científicas y Técnicas (CIBICI-CONICET), Córdoba, Argentina
| | - Victoria Peyret
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
- Centro de Investigaciones en Bioquímica Clínica e Inmunología, Consejo Nacional de Investigaciones Científicas y Técnicas (CIBICI-CONICET), Córdoba, Argentina
| | - Romina Celeste Geysels
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
- Centro de Investigaciones en Bioquímica Clínica e Inmunología, Consejo Nacional de Investigaciones Científicas y Técnicas (CIBICI-CONICET), Córdoba, Argentina
| | - Francisco Andrés Montes
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
- Centro de Investigaciones en Bioquímica Clínica e Inmunología, Consejo Nacional de Investigaciones Científicas y Técnicas (CIBICI-CONICET), Córdoba, Argentina
| | - Carlos Eduardo Bernal Barquero
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
- Centro de Investigaciones en Bioquímica Clínica e Inmunología, Consejo Nacional de Investigaciones Científicas y Técnicas (CIBICI-CONICET), Córdoba, Argentina
| | - Valentina Ricci
- División de Endocrinología, Hospital de Niños Dr. Ricardo Gutiérrez, Buenos Aires, Argentina
- Centro de Investigaciones Endocrinológicas Dr. César Bergadá, Consejo Nacional de Investigaciones Científicas y Técnicas (CEDIE-CONICET), Buenos Aires, Argentina
| | - María Eugenia Masnata
- División de Endocrinología, Hospital de Niños Dr. Ricardo Gutiérrez, Buenos Aires, Argentina
- Centro de Investigaciones Endocrinológicas Dr. César Bergadá, Consejo Nacional de Investigaciones Científicas y Técnicas (CEDIE-CONICET), Buenos Aires, Argentina
| | - Ana María Masini-Repiso
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
- Centro de Investigaciones en Bioquímica Clínica e Inmunología, Consejo Nacional de Investigaciones Científicas y Técnicas (CIBICI-CONICET), Córdoba, Argentina
| | - Patricia Papendieck
- División de Endocrinología, Hospital de Niños Dr. Ricardo Gutiérrez, Buenos Aires, Argentina
- Centro de Investigaciones Endocrinológicas Dr. César Bergadá, Consejo Nacional de Investigaciones Científicas y Técnicas (CEDIE-CONICET), Buenos Aires, Argentina
| | - Mariana Lorena Tellechea
- División de Endocrinología, Hospital de Niños Dr. Ricardo Gutiérrez, Buenos Aires, Argentina
- Centro de Investigaciones Endocrinológicas Dr. César Bergadá, Consejo Nacional de Investigaciones Científicas y Técnicas (CEDIE-CONICET), Buenos Aires, Argentina
| | - Ana Elena Chiesa
- División de Endocrinología, Hospital de Niños Dr. Ricardo Gutiérrez, Buenos Aires, Argentina
- Centro de Investigaciones Endocrinológicas Dr. César Bergadá, Consejo Nacional de Investigaciones Científicas y Técnicas (CEDIE-CONICET), Buenos Aires, Argentina
| | - Juan Pablo Nicola
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
- Centro de Investigaciones en Bioquímica Clínica e Inmunología, Consejo Nacional de Investigaciones Científicas y Técnicas (CIBICI-CONICET), Córdoba, Argentina
| |
Collapse
|
2
|
Read ML, Brookes K, Zha L, Manivannan S, Kim J, Kocbiyik M, Fletcher A, Gorvin CM, Firth G, Fruhwirth GO, Nicola JP, Jhiang S, Ringel MD, Campbell MJ, Sunassee K, Blower PJ, Boelaert K, Nieto HR, Smith VE, McCabe CJ. Combined Vorinostat and Chloroquine Inhibit Sodium-Iodide Symporter Endocytosis and Enhance Radionuclide Uptake In Vivo. Clin Cancer Res 2024; 30:1352-1366. [PMID: 37921808 PMCID: PMC7615786 DOI: 10.1158/1078-0432.ccr-23-2043] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 09/12/2023] [Accepted: 11/01/2023] [Indexed: 11/04/2023]
Abstract
PURPOSE Patients with aggressive thyroid cancer are frequently failed by the central therapy of ablative radioiodide (RAI) uptake, due to reduced plasma membrane (PM) localization of the sodium/iodide symporter (NIS). We aimed to understand how NIS is endocytosed away from the PM of human thyroid cancer cells, and whether this was druggable in vivo. EXPERIMENTAL DESIGN Informed by analysis of endocytic gene expression in patients with aggressive thyroid cancer, we used mutagenesis, NanoBiT interaction assays, cell surface biotinylation assays, RAI uptake, and NanoBRET to understand the mechanisms of NIS endocytosis in transformed cell lines and patient-derived human primary thyroid cells. Systemic drug responses were monitored via 99mTc pertechnetate gamma counting and gene expression in BALB/c mice. RESULTS We identified an acidic dipeptide within the NIS C-terminus that mediates binding to the σ2 subunit of the Adaptor Protein 2 (AP2) heterotetramer. We discovered that the FDA-approved drug chloroquine (CQ) modulates NIS accumulation at the PM in a functional manner that is AP2 dependent. In vivo, CQ treatment of BALB/c mice significantly enhanced thyroidal uptake of 99mTc pertechnetate in combination with the histone deacetylase (HDAC) inhibitor vorinostat/SAHA, accompanied by increased thyroidal NIS mRNA. Bioinformatic analyses validated the clinical relevance of AP2 genes with disease-free survival in RAI-treated DTC, enabling construction of an AP2 gene-related risk score classifier for predicting recurrence. CONCLUSIONS NIS internalization is specifically druggable in vivo. Our data, therefore, provide new translatable potential for improving RAI therapy using FDA-approved drugs in patients with aggressive thyroid cancer. See related commentary by Lechner and Brent, p. 1220.
Collapse
Affiliation(s)
- Martin L. Read
- Institute of Metabolism and Systems Research (IMSR), and Centre of Endocrinology, Diabetes and Metabolism (CEDAM), University of Birmingham, Birmingham, UK
| | - Katie Brookes
- Institute of Metabolism and Systems Research (IMSR), and Centre of Endocrinology, Diabetes and Metabolism (CEDAM), University of Birmingham, Birmingham, UK
| | - Ling Zha
- Institute of Metabolism and Systems Research (IMSR), and Centre of Endocrinology, Diabetes and Metabolism (CEDAM), University of Birmingham, Birmingham, UK
| | - Selvambigai Manivannan
- Institute of Metabolism and Systems Research (IMSR), and Centre of Endocrinology, Diabetes and Metabolism (CEDAM), University of Birmingham, Birmingham, UK
| | - Jana Kim
- School of Biomedical Engineering & Imaging Sciences, King’s College London, London, UK
| | - Merve Kocbiyik
- Institute of Metabolism and Systems Research (IMSR), and Centre of Endocrinology, Diabetes and Metabolism (CEDAM), University of Birmingham, Birmingham, UK
| | - Alice Fletcher
- Institute of Metabolism and Systems Research (IMSR), and Centre of Endocrinology, Diabetes and Metabolism (CEDAM), University of Birmingham, Birmingham, UK
| | - Caroline M. Gorvin
- Institute of Metabolism and Systems Research (IMSR), and Centre of Endocrinology, Diabetes and Metabolism (CEDAM), University of Birmingham, Birmingham, UK
| | - George Firth
- School of Biomedical Engineering & Imaging Sciences, King’s College London, London, UK
| | - Gilbert O. Fruhwirth
- Comprehensive Cancer Centre, School of Cancer and Pharmaceutical Sciences, King's College London, Guy's Campus, London, UK
| | - Juan P. Nicola
- Departamento de Bioquímica Clínica (CIBICI-CONICET), Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Sissy Jhiang
- Divison of Endocrinology, Diabetes, and Metabolism and Cancer Biology Program, The Ohio State University College of Medicine and Comprehensive Cancer Center, Columbus, Ohio, USA
| | - Matthew D. Ringel
- Divison of Endocrinology, Diabetes, and Metabolism and Cancer Biology Program, The Ohio State University College of Medicine and Comprehensive Cancer Center, Columbus, Ohio, USA
| | - Moray J. Campbell
- Department of Pharmaceutics and Pharmaceutical Chemistry, College of Pharmacy at The Ohio State University, Columbus, Ohio, USA
| | - Kavitha Sunassee
- School of Biomedical Engineering & Imaging Sciences, King’s College London, London, UK
| | - Philip J. Blower
- School of Biomedical Engineering & Imaging Sciences, King’s College London, London, UK
| | - Kristien Boelaert
- Institute of Applied Health Research, University of Birmingham, Birmingham, UK
| | - Hannah R. Nieto
- Institute of Metabolism and Systems Research (IMSR), and Centre of Endocrinology, Diabetes and Metabolism (CEDAM), University of Birmingham, Birmingham, UK
| | - Vicki E. Smith
- Institute of Metabolism and Systems Research (IMSR), and Centre of Endocrinology, Diabetes and Metabolism (CEDAM), University of Birmingham, Birmingham, UK
| | - Christopher J. McCabe
- Institute of Metabolism and Systems Research (IMSR), and Centre of Endocrinology, Diabetes and Metabolism (CEDAM), University of Birmingham, Birmingham, UK
| |
Collapse
|
3
|
Wang D, Liu X, Li M, Ning J. HIF-1α regulates the cell viability in radioiodine-resistant papillary thyroid carcinoma cells induced by hypoxia through PKM2/NF-κB signaling pathway. Mol Carcinog 2024; 63:238-252. [PMID: 37861358 DOI: 10.1002/mc.23648] [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: 02/19/2023] [Revised: 09/05/2023] [Accepted: 10/01/2023] [Indexed: 10/21/2023]
Abstract
The curative treatment options for papillary thyroid cancer (PTC) encompass surgical intervention, radioactive iodine administration, and chemotherapy. However, the challenges of radioiodine (RAI) resistance, metastasis, and chemotherapy resistance remain inadequately addressed. The objective of this study was to investigate the protective role of hypoxia-inducible factor-1α (HIF-1α) in 131 I-resistant cells and a xenograft model under hypoxic conditions, as well as to explore potential mechanisms. The effects of HIF-1α on 131 I-resistant BCPAP and TPC-1 cells, as well as the xenograft model, were assessed in this study. Cell viability, migration, invasion, and apoptosis rates were measured using Cell Counting Kit-8, wound-healing, Transwell, and flow cytometry assays. Additionally, the expressions of Ki67, matrix metalloproteinase-9 (MMP-9), and pyruvate kinase M2 (PKM2) were examined using immunofluorescence or immunohistochemistry assays. Sodium iodide symporter and PKM2/NF-κBp65 relative protein levels were detected by western blot analysis. The findings of our study indicate that siHIF-1α effectively inhibits cell proliferation, cell migration, and invasion in 131 I-resistant cells under hypoxic conditions. Additionally, the treatment of siHIF-1α leads to alterations in the relative protein levels of Ki67, MMP-9, PKM2, and PKM2/NF-κBp65, both in vivo and in vitro. Notably, the effects of siHIF-1α are modified when DASA-58, an activator of PKM2, is administered. These results collectively demonstrate that siHIF-1α reduces cell viability in PTC cells and rat models, while also mediating the nuclear factor-κB (NF-κB)/PKM2 signaling pathway. Our findings provide a new rationale for further academic and clinical research on RAI-resistant PTC.
Collapse
Affiliation(s)
- Dong Wang
- Thyroid Surgery Ward, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong, China
| | - Xiaoqian Liu
- Department of Hematology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong, China
| | - Meijing Li
- Second Department of Hepatobiliary Surgery, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong, China
| | - Jinyao Ning
- Thyroid Surgery Ward, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong, China
| |
Collapse
|
4
|
Zhang L, Li Z, Zhang M, Zou H, Bai Y, Liu Y, Lv J, Lv L, Liu P, Deng Z, Liu C. Advances in the molecular mechanism and targeted therapy of radioactive-iodine refractory differentiated thyroid cancer. Med Oncol 2023; 40:258. [PMID: 37524925 DOI: 10.1007/s12032-023-02098-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 06/21/2023] [Indexed: 08/02/2023]
Abstract
Most patients with differentiated thyroid cancer have a good prognosis after radioactive iodine-131 treatment, but there are still a small number of patients who are not sensitive to radioiodine treatment and may subsequently show disease progression. Therefore, radioactive-iodine refractory differentiated thyroid cancer treated with radioiodine usually shows reduced radioiodine uptake. Thus, when sodium iodine symporter expression, basolateral membrane localization and recycling degradation are abnormal, radioactive-iodine refractory differentiated thyroid cancer may occur. In recent years, with the deepening of research into the pathogenesis of this disease, an increasing number of molecules have become or are expected to become therapeutic targets. The application of corresponding inhibitors or combined treatment regimens for different molecular targets may be effective for patients with advanced radioactive-iodine refractory differentiated thyroid cancer. Currently, some targeted drugs that can improve the progression-free survival of patients with radioactive-iodine refractory differentiated thyroid cancer, such as sorafenib and lenvatinib, have been approved by the FDA for the treatment of radioactive-iodine refractory differentiated thyroid cancer. However, due to the adverse reactions and drug resistance caused by some targeted drugs, their application is limited. In response to targeted drug resistance and high rates of adverse reactions, research into new treatment combinations is being carried out; in addition to kinase inhibitor therapy, gene therapy and rutin-assisted iodine-131 therapy for radioactive-iodine refractory thyroid cancer have also made some progress. Thus, this article mainly focuses on sodium iodide symporter changes leading to the main molecular mechanisms in radioactive-iodine refractory differentiated thyroid cancer, some targeted drug resistance mechanisms and promising new treatments.
Collapse
Affiliation(s)
- Lu Zhang
- Department of Nuclear Medicine, The Third Affiliated Hospital of Kunming Medical University, Tumor Hospital of Yunnan Province, 519 Kunzhou Road, Xishan District, Kunming, KM, 650118, China
| | - Zhi Li
- Department of Nuclear Medicine, The Third Affiliated Hospital of Kunming Medical University, Tumor Hospital of Yunnan Province, 519 Kunzhou Road, Xishan District, Kunming, KM, 650118, China
| | - Meng Zhang
- Department of Nuclear Medicine, The Third Affiliated Hospital of Kunming Medical University, Tumor Hospital of Yunnan Province, 519 Kunzhou Road, Xishan District, Kunming, KM, 650118, China
| | - Huangren Zou
- Department of Nuclear Medicine, The Third Affiliated Hospital of Kunming Medical University, Tumor Hospital of Yunnan Province, 519 Kunzhou Road, Xishan District, Kunming, KM, 650118, China
| | - Yuke Bai
- Department of Nuclear Medicine, The Third Affiliated Hospital of Kunming Medical University, Tumor Hospital of Yunnan Province, 519 Kunzhou Road, Xishan District, Kunming, KM, 650118, China
| | - Yanlin Liu
- Department of Nuclear Medicine, The Third Affiliated Hospital of Kunming Medical University, Tumor Hospital of Yunnan Province, 519 Kunzhou Road, Xishan District, Kunming, KM, 650118, China
| | - Juan Lv
- Department of Nuclear Medicine, The Third Affiliated Hospital of Kunming Medical University, Tumor Hospital of Yunnan Province, 519 Kunzhou Road, Xishan District, Kunming, KM, 650118, China
| | - Ling Lv
- Department of Nuclear Medicine, The Third Affiliated Hospital of Kunming Medical University, Tumor Hospital of Yunnan Province, 519 Kunzhou Road, Xishan District, Kunming, KM, 650118, China
| | - Pengjie Liu
- Department of Nuclear Medicine, The Third Affiliated Hospital of Kunming Medical University, Tumor Hospital of Yunnan Province, 519 Kunzhou Road, Xishan District, Kunming, KM, 650118, China
| | - Zhiyong Deng
- Department of Nuclear Medicine, The Third Affiliated Hospital of Kunming Medical University, Tumor Hospital of Yunnan Province, 519 Kunzhou Road, Xishan District, Kunming, KM, 650118, China.
| | - Chao Liu
- Department of Nuclear Medicine, The Third Affiliated Hospital of Kunming Medical University, Tumor Hospital of Yunnan Province, 519 Kunzhou Road, Xishan District, Kunming, KM, 650118, China
| |
Collapse
|
5
|
Bernal Barquero CE, Geysels RC, Jacques V, Carro GH, Martín M, Peyret V, Abregú MC, Papendieck P, Masini-Repiso AM, Savagner F, Chiesa AE, Citterio CE, Nicola JP. Targeted Next-Generation Sequencing of Congenital Hypothyroidism-Causative Genes Reveals Unexpected Thyroglobulin Gene Variants in Patients with Iodide Transport Defect. Int J Mol Sci 2022; 23:ijms23169251. [PMID: 36012511 PMCID: PMC9409291 DOI: 10.3390/ijms23169251] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 08/04/2022] [Accepted: 08/08/2022] [Indexed: 01/12/2023] Open
Abstract
Congenital iodide transport defect is an uncommon autosomal recessive disorder caused by loss-of-function variants in the sodium iodide symporter (NIS)-coding SLC5A5 gene and leading to dyshormonogenic congenital hypothyroidism. Here, we conducted a targeted next-generation sequencing assessment of congenital hypothyroidism-causative genes in a cohort of nine unrelated pediatric patients suspected of having a congenital iodide transport defect based on the absence of 99mTc-pertechnetate accumulation in a eutopic thyroid gland. Although, unexpectedly, we could not detect pathogenic SLC5A5 gene variants, we identified two novel compound heterozygous TG gene variants (p.Q29* and c.177-2A>C), three novel heterozygous TG gene variants (p.F1542Vfs*20, p.Y2563C, and p.S523P), and a novel heterozygous DUOX2 gene variant (p.E1496Dfs*51). Splicing minigene reporter-based in vitro assays revealed that the variant c.177-2A>C affected normal TG pre-mRNA splicing, leading to the frameshift variant p.T59Sfs*17. The frameshift TG variants p.T59Sfs*17 and p.F1542Vfs*20, but not the DUOX2 variant p.E1496Dfs*51, were predicted to undergo nonsense-mediated decay. Moreover, functional in vitro expression assays revealed that the variant p.Y2563C reduced the secretion of the TG protein. Our investigation revealed unexpected findings regarding the genetics of congenital iodide transport defects, supporting the existence of yet to be discovered mechanisms involved in thyroid hormonogenesis.
Collapse
Affiliation(s)
- Carlos Eduardo Bernal Barquero
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Cordoba 5000, Argentina
- Centro de Investigaciones en Bioquímica Clínica e Inmunología, Consejo Nacional de Investigaciones Científicas y Técnicas (CIBICI-CONICET), Cordoba 5000, Argentina
| | - Romina Celeste Geysels
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Cordoba 5000, Argentina
- Centro de Investigaciones en Bioquímica Clínica e Inmunología, Consejo Nacional de Investigaciones Científicas y Técnicas (CIBICI-CONICET), Cordoba 5000, Argentina
| | - Virginie Jacques
- Laboratoire de Biochimie, Institut Fédératif de Biologie, Le Centre Hospitalier Universitaire de Toulouse, 31300 Toulouse, France
- Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), Institut National de la Santé et de la Recherche Médicale (INSERM) UMR 1297, 31432 Toulouse, France
| | - Gerardo Hernán Carro
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Cordoba 5000, Argentina
- Centro de Investigaciones en Bioquímica Clínica e Inmunología, Consejo Nacional de Investigaciones Científicas y Técnicas (CIBICI-CONICET), Cordoba 5000, Argentina
| | - Mariano Martín
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Cordoba 5000, Argentina
- Centro de Investigaciones en Bioquímica Clínica e Inmunología, Consejo Nacional de Investigaciones Científicas y Técnicas (CIBICI-CONICET), Cordoba 5000, Argentina
| | - Victoria Peyret
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Cordoba 5000, Argentina
- Centro de Investigaciones en Bioquímica Clínica e Inmunología, Consejo Nacional de Investigaciones Científicas y Técnicas (CIBICI-CONICET), Cordoba 5000, Argentina
| | - María Celeste Abregú
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Cordoba 5000, Argentina
- Centro de Investigaciones en Bioquímica Clínica e Inmunología, Consejo Nacional de Investigaciones Científicas y Técnicas (CIBICI-CONICET), Cordoba 5000, Argentina
| | - Patricia Papendieck
- División de Endocrinología, Hospital de Niños Dr. Ricardo Gutiérrez, Buenos Aires 1006, Argentina
- Centro de Investigaciones Endocrinológicas Dr. César Bergadá, Consejo Nacional de Investigaciones Científicas y Técnicas (CEDIE-CONICET), Buenos Aires 1120, Argentina
| | - Ana María Masini-Repiso
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Cordoba 5000, Argentina
- Centro de Investigaciones en Bioquímica Clínica e Inmunología, Consejo Nacional de Investigaciones Científicas y Técnicas (CIBICI-CONICET), Cordoba 5000, Argentina
| | - Frédérique Savagner
- Laboratoire de Biochimie, Institut Fédératif de Biologie, Le Centre Hospitalier Universitaire de Toulouse, 31300 Toulouse, France
- Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), Institut National de la Santé et de la Recherche Médicale (INSERM) UMR 1297, 31432 Toulouse, France
| | - Ana Elena Chiesa
- División de Endocrinología, Hospital de Niños Dr. Ricardo Gutiérrez, Buenos Aires 1006, Argentina
- Centro de Investigaciones Endocrinológicas Dr. César Bergadá, Consejo Nacional de Investigaciones Científicas y Técnicas (CEDIE-CONICET), Buenos Aires 1120, Argentina
| | - Cintia E. Citterio
- Instituto de Inmunología, Genética y Metabolismo, Consejo Nacional de Investigaciones Científicas y Técnicas (INIGEM-CONIET), Buenos Aires 1120, Argentina
- Division of Metabolism, Endocrinology and Diabetes, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Juan Pablo Nicola
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Cordoba 5000, Argentina
- Centro de Investigaciones en Bioquímica Clínica e Inmunología, Consejo Nacional de Investigaciones Científicas y Técnicas (CIBICI-CONICET), Cordoba 5000, Argentina
- Correspondence: ; Tel.: +54-0351-535-3850 (ext. 55423)
| |
Collapse
|
6
|
Geysels RC, Bernal Barquero CE, Martín M, Peyret V, Nocent M, Sobrero G, Muñoz L, Signorino M, Testa G, Castro RB, Masini-Repiso AM, Miras MB, Nicola JP. Silent but Not Harmless: A Synonymous SLC5A5 Gene Variant Leading to Dyshormonogenic Congenital Hypothyroidism. Front Endocrinol (Lausanne) 2022; 13:868891. [PMID: 35600585 PMCID: PMC9114739 DOI: 10.3389/fendo.2022.868891] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 03/23/2022] [Indexed: 12/25/2022] Open
Abstract
Background Congenital iodide transport defect (ITD) is an uncommon cause of dyshormonogenic congenital hypothyroidism characterized by the absence of active iodide accumulation in the thyroid gland. ITD is an autosomal recessive disorder caused by loss-of-function variants in the sodium/iodide symporter (NIS)-coding SLC5A5 gene. Objective We aimed to identify, and if so to functionally characterize, novel ITD-causing SLC5A5 gene variants in a cohort of five unrelated pediatric patients diagnosed with dyshormonogenic congenital hypothyroidism with minimal to absent 99mTc-pertechnetate accumulation in the thyroid gland. Methods The coding region of the SLC5A5 gene was sequenced using Sanger sequencing. In silico analysis and functional in vitro characterization of a novel synonymous variant were performed. Results Sanger sequencing revealed a novel homozygous synonymous SLC5A5 gene variant (c.1326A>C in exon 11). In silico analysis revealed that the c.1326A>C variant is potentially deleterious for NIS pre-mRNA splicing. The c.1326A>C variant was predicted to lie within a putative exonic splicing enhancer reducing the binding of splicing regulatory trans-acting protein SRSF5. Splicing minigene reporter assay revealed that c.1326A>C causes exon 11 or exon 11 and 12 skipping during NIS pre-mRNA splicing leading to the NIS pathogenic variants p.G415_P443del and p.G415Lfs*32, respectively. Significantly, the frameshift variant p.G415Lfs*32 is predicted to be subjected to degradation by nonsense-mediated decay. Conclusions We identified the first exonic synonymous SLC5A5 gene variant causing aberrant NIS pre-mRNA splicing, thus expanding the mutational landscape of the SLC5A5 gene leading to dyshormonogenic congenital hypothyroidism.
Collapse
Affiliation(s)
- Romina Celeste Geysels
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
- Centro de Investigaciones en Bioquímica Clínica e Inmunología - Consejo Nacional de Investigaciones Científicas y Técnicas (CIBICI-CONICET), Córdoba, Argentina
| | - Carlos Eduardo Bernal Barquero
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
- Centro de Investigaciones en Bioquímica Clínica e Inmunología - Consejo Nacional de Investigaciones Científicas y Técnicas (CIBICI-CONICET), Córdoba, Argentina
| | - Mariano Martín
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
- Centro de Investigaciones en Bioquímica Clínica e Inmunología - Consejo Nacional de Investigaciones Científicas y Técnicas (CIBICI-CONICET), Córdoba, Argentina
| | - Victoria Peyret
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
- Centro de Investigaciones en Bioquímica Clínica e Inmunología - Consejo Nacional de Investigaciones Científicas y Técnicas (CIBICI-CONICET), Córdoba, Argentina
| | - Martina Nocent
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
- Centro de Investigaciones en Bioquímica Clínica e Inmunología - Consejo Nacional de Investigaciones Científicas y Técnicas (CIBICI-CONICET), Córdoba, Argentina
| | - Gabriela Sobrero
- Programa Provincial de Pesquisa Neonatal, Servicio de Endocrinología, Hospital de Niños de la Santísima Trinidad de Córdoba, Córdoba, Argentina
| | - Liliana Muñoz
- Programa Provincial de Pesquisa Neonatal, Servicio de Endocrinología, Hospital de Niños de la Santísima Trinidad de Córdoba, Córdoba, Argentina
| | - Malvina Signorino
- Programa Provincial de Pesquisa Neonatal, Servicio de Endocrinología, Hospital de Niños de la Santísima Trinidad de Córdoba, Córdoba, Argentina
| | - Graciela Testa
- Programa Provincial de Pesquisa Neonatal, Servicio de Endocrinología, Hospital de Niños de la Santísima Trinidad de Córdoba, Córdoba, Argentina
| | | | - Ana María Masini-Repiso
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
- Centro de Investigaciones en Bioquímica Clínica e Inmunología - Consejo Nacional de Investigaciones Científicas y Técnicas (CIBICI-CONICET), Córdoba, Argentina
| | - Mirta Beatriz Miras
- Programa Provincial de Pesquisa Neonatal, Servicio de Endocrinología, Hospital de Niños de la Santísima Trinidad de Córdoba, Córdoba, Argentina
| | - Juan Pablo Nicola
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
- Centro de Investigaciones en Bioquímica Clínica e Inmunología - Consejo Nacional de Investigaciones Científicas y Técnicas (CIBICI-CONICET), Córdoba, Argentina
| |
Collapse
|
7
|
Oh JM, Rajendran RL, Gangadaran P, Hong CM, Jeong JH, Lee J, Ahn BC. Targeting GLI1 Transcription Factor for Restoring Iodine Avidity with Redifferentiation in Radioactive-Iodine Refractory Thyroid Cancers. Cancers (Basel) 2022; 14:cancers14071782. [PMID: 35406554 PMCID: PMC8997411 DOI: 10.3390/cancers14071782] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 03/28/2022] [Accepted: 03/29/2022] [Indexed: 11/25/2022] Open
Abstract
Simple Summary Thyroid cancers have an excellent prognosis by standard therapy of surgery followed by radioactive-iodine therapy. However, metastatic thyroid cancers do not response to radioactive-iodine therapy by losing iodine avidity. Therefore, reversing iodine avidity to metastatic thyroid cancers gives a new chance of applying radioactive-iodine therapy to the cancers. In the current study, we found that GLI1 knockdown can revert iodine non-avid thyroid cancers to iodine avid cancers by increasing expression of thyroid-specific proteins. Restoration of iodine avidity in thyroid cancers makes the cancers sensitive to radioactive-iodine therapy again. Therefore, the GLI1 can be a potential therapeutic target of radioactive-iodine resistant thyroid cancers. Abstract Radioactive-iodine (RAI) therapy is the mainstay for patients with recurrent and metastatic thyroid cancer. However, many patients exhibit dedifferentiation characteristics along with lack of sodium iodide symporter (NIS) functionality, low expression of thyroid-specific proteins, and poor RAI uptake, leading to poor prognosis. Previous studies have demonstrated the effect of GLI family zinc finger 1 (GLI1) inhibition on tumor growth and apoptosis. In this study, we investigated the role of GLI1 in the context of redifferentiation and improvement in the efficacy of RAI therapy for thyroid cancer. We evaluated GLI1 expression in several thyroid cancer cell lines and selected TPC-1 and SW1736 cell lines showing the high expression of GLI. We performed GLI1 knockdown and evaluated the changes of thyroid-specific proteins expression, RAI uptake and I-131-mediated cytotoxicity. The effect of GANT61 (GLI1 inhibitor) on endogenous NIS expression was also assessed. Endogenous NIS expression upregulated by inhibiting GLI1, in addition, increased expression level in plasma membrane. Also, GLI1 knockdown increased expression of thyroid-specific proteins. Restoration of thyroid-specific proteins increased RAI uptake and I-131-mediated cytotoxic effect. Treatment with GANT61 also increased expression of endogenous NIS. Targeting GLI1 can be a potential strategy with redifferentiation for restoring RAI avidity in dedifferentiated thyroid cancers.
Collapse
Affiliation(s)
- Ji Min Oh
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu 41944, Korea; (J.M.O.); (R.L.R.); (P.G.); (C.M.H.); (J.L.)
| | - Ramya Lakshmi Rajendran
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu 41944, Korea; (J.M.O.); (R.L.R.); (P.G.); (C.M.H.); (J.L.)
| | - Prakash Gangadaran
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu 41944, Korea; (J.M.O.); (R.L.R.); (P.G.); (C.M.H.); (J.L.)
- BK21 FOUR KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, Department of Biomedical Science, School of Medicine, Kyungpook National University, Daegu 41944, Korea
| | - Chae Moon Hong
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu 41944, Korea; (J.M.O.); (R.L.R.); (P.G.); (C.M.H.); (J.L.)
- Department of Nuclear Medicine, Kyungpook National University Hospital, Daegu 41944, Korea; zzu--@hanmail.net
| | - Ju Hye Jeong
- Department of Nuclear Medicine, Kyungpook National University Hospital, Daegu 41944, Korea; zzu--@hanmail.net
| | - Jaetae Lee
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu 41944, Korea; (J.M.O.); (R.L.R.); (P.G.); (C.M.H.); (J.L.)
- Department of Nuclear Medicine, Kyungpook National University Hospital, Daegu 41944, Korea; zzu--@hanmail.net
| | - Byeong-Cheol Ahn
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu 41944, Korea; (J.M.O.); (R.L.R.); (P.G.); (C.M.H.); (J.L.)
- BK21 FOUR KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, Department of Biomedical Science, School of Medicine, Kyungpook National University, Daegu 41944, Korea
- Department of Nuclear Medicine, Kyungpook National University Hospital, Daegu 41944, Korea; zzu--@hanmail.net
- Correspondence: ; Tel.: +82-53-420-5583
| |
Collapse
|
8
|
Martín M, Brunello FG, Modenutti CP, Nicola JP, Marti MA. MotSASi: Functional short linear motifs (SLiMs) prediction based on genomic single nucleotide variants and structural data. Biochimie 2022; 197:59-73. [DOI: 10.1016/j.biochi.2022.02.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 01/17/2022] [Accepted: 02/02/2022] [Indexed: 11/28/2022]
|
9
|
Bernal Barquero CE, Martín M, Geysels RC, Peyret V, Papendieck P, Masini-Repiso AM, Chiesa AE, Nicola JP. An Intramolecular Ionic Interaction Linking Defective Sodium/Iodide Symporter Transport to the Plasma Membrane and Dyshormonogenic Congenital Hypothyroidism. Thyroid 2022; 32:19-27. [PMID: 34726525 DOI: 10.1089/thy.2021.0344] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Background: The sodium/iodide symporter (NIS) mediates active iodide accumulation in the thyroid follicular cell. Autosomal recessive iodide transport defect (ITD)-causing loss-of-function NIS variants lead to dyshormonogenic congenital hypothyroidism due to deficient iodide accumulation for thyroid hormonogenesis. Here, we aimed to identify, and if so to functionally characterize, novel ITD-causing NIS pathogenic variants in a patient diagnosed with severe dyshormonogenic congenital hypothyroidism due to a defect in iodide accumulation in the thyroid follicular cell, as suggested by nondetectable radioiodide accumulation in a normally located thyroid gland, as well as in salivary glands. Methods: The proposita NIS-coding SLC5A5 gene was sequenced using Sanger sequencing. In silico analysis and functional in vitro characterization of the novel NIS variants were performed. Results: Sanger sequencing revealed novel compound heterozygous SLC5A5 gene variants (c.970-3C>A and c.1106A>T, p.D369V). In silico analysis suggested that c.970-3C>A disrupts the canonical splice acceptor site located in intron 7. Splicing minigene reporter assay revealed that c.970-3C>A causes exon 8 skipping during NIS pre-mRNA splicing leading to the NIS pathogenic variant p.Y324Hfs*148. Moreover, in silico analysis indicated p.D369V as pathogenic. Functional in vitro studies demonstrated that p.D369V NIS does not mediate iodide accumulation, as p.D369V causes NIS to be retained in the endoplasmic reticulum. Mechanistically, we propose an intramolecular ionic interaction involving the β carboxyl group of D369 and the guanidinium group of R130, located in transmembrane segment 4. Of note, an Asp residue at position 369-which is highly conserved in SLC5A family members-is required for functional NIS expression at the plasma membrane. Conclusions: We uncovered a critical intramolecular interaction between R130 and D369 required for NIS maturation and plasma membrane expression. Moreover, we identified the first intronic variant causing aberrant NIS pre-mRNA splicing, thus expanding the mutational landscape in the SLC5A5 gene leading to dyshormonogenic congenital hypothyroidism.
Collapse
Affiliation(s)
- Carlos Eduardo Bernal Barquero
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
- Centro de Investigaciones en Bioquímica Clínica e Inmunología-Consejo Nacional de Investigaciones Científicas y Técnicas (CIBICI-CONICET), Córdoba, Argentina
| | - Mariano Martín
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
- Centro de Investigaciones en Bioquímica Clínica e Inmunología-Consejo Nacional de Investigaciones Científicas y Técnicas (CIBICI-CONICET), Córdoba, Argentina
| | - Romina Celeste Geysels
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
- Centro de Investigaciones en Bioquímica Clínica e Inmunología-Consejo Nacional de Investigaciones Científicas y Técnicas (CIBICI-CONICET), Córdoba, Argentina
| | - Victoria Peyret
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
- Centro de Investigaciones en Bioquímica Clínica e Inmunología-Consejo Nacional de Investigaciones Científicas y Técnicas (CIBICI-CONICET), Córdoba, Argentina
| | - Patricia Papendieck
- División de Endocrinología, Hospital de Niños Dr. Ricardo Gutiérrez, Buenos Aires, Argentina
- Centro de Investigaciones Endocrinológicas Dr. César Bergadá-Consejo Nacional de Investigaciones Científicas y Técnicas (CEDIE-CONICET), Buenos Aires, Argentina
| | - Ana María Masini-Repiso
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
- Centro de Investigaciones en Bioquímica Clínica e Inmunología-Consejo Nacional de Investigaciones Científicas y Técnicas (CIBICI-CONICET), Córdoba, Argentina
| | - Ana Elena Chiesa
- División de Endocrinología, Hospital de Niños Dr. Ricardo Gutiérrez, Buenos Aires, Argentina
- Centro de Investigaciones Endocrinológicas Dr. César Bergadá-Consejo Nacional de Investigaciones Científicas y Técnicas (CEDIE-CONICET), Buenos Aires, Argentina
| | - Juan Pablo Nicola
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
- Centro de Investigaciones en Bioquímica Clínica e Inmunología-Consejo Nacional de Investigaciones Científicas y Técnicas (CIBICI-CONICET), Córdoba, Argentina
| |
Collapse
|
10
|
Abstract
Background: Iodide transport defect is an uncommon cause of dyshormonogenic congenital hypothyroidism due to homozygous or compound heterozygous pathogenic variants in the SLC5A5 gene, which encodes the sodium/iodide symporter (NIS), causing deficient iodide accumulation in thyroid follicular cells, thus impairing thyroid hormonogenesis. Methods:SLC5A5 gene variants were compiled from public databases and research articles exploring the molecular bases of congenital hypothyroidism. Using a dataset of 198 missense NIS variants classified as either benign or pathogenic, we developed and validated a machine learning-based NIS-specific variant classifier to predict the impact of missense NIS variants. Results: We generated a manually curated dataset containing 7793 unique SLC5A5 variants. As most databases compiled exome sequencing data, variant mapping revealed an increased density of variants in SLC5A5 coding exons. Based on allele frequency (AF) analysis, we established an AF threshold of 1:10,000 above which a variant should be considered benign. Most pathogenic NIS variants were located in the protein-coding region, as most patients were genetically diagnosed by using a candidate gene strategy limited to this region. Significantly, we evidenced that 94.5% of missense NIS variants were classified as of uncertain significance. Therefore, we developed an NIS-specific variant classifier to improve the prediction of pathogenicity of missense variants. Our classifier predicted the clinical outcome of missense variants with high accuracy (90%), outperforming state-of-the-art pathogenicity predictors, such as REVEL, PolyPhen-2, and SIFT. Based on the excellent performance of our classifier, we predicted the mutational landscape of NIS. The analysis of the mutational landscape revealed that most missense variants located in transmembrane segments are frequently pathogenic. Moreover, we predicted that ∼28% of all single-nucleotide variants that could cause missense NIS variants are pathogenic, thus putatively leading to congenital hypothyroidism if present in homozygous or compound heterozygous state. Conclusions: We reported the first NIS-specific variant classifier aiming at improving the interpretation of missense NIS variants in clinical practice. Deciphering the mutational landscape for every protein involved in thyroid hormonogenesis is a relevant task for a deep understanding of the molecular mechanisms causing dyshormonogenic congenital hypothyroidism.
Collapse
Affiliation(s)
- Mariano Martín
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
- Centro de Investigaciones en Bioquímica Clínica e Inmunología Consejo Nacional de Investigaciones Científicas y Técnicas (CIBICI-CONICET), Córdoba, Argentina
| | - Juan Pablo Nicola
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
- Centro de Investigaciones en Bioquímica Clínica e Inmunología Consejo Nacional de Investigaciones Científicas y Técnicas (CIBICI-CONICET), Córdoba, Argentina
| |
Collapse
|