Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
1.
JCI Insight ; 9(4)2024 Jan 09.
Article in English | MEDLINE | ID: mdl-38194289

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.


Subject(s)
Hyperthyroidism , Iodine , Receptors, Thyrotropin , Animals , Humans , Mice , Hyperthyroidism/congenital , Mutation , Phenotype , Receptors, G-Protein-Coupled/genetics , Receptors, Thyrotropin/genetics , Receptors, Thyrotropin/metabolism
2.
Hum Mol Genet ; 31(12): 2063-2077, 2022 06 22.
Article in English | MEDLINE | ID: mdl-35043958

ABSTRACT

Prostate cancer is among the most common cancers in men, with a large fraction of the individual risk attributable to heritable factors. A majority of the diagnosed cases does not lead to a lethal disease, and hence biological markers that can distinguish between indolent and fatal forms of the disease are of great importance for guiding treatment decisions. Although over 300 genetic variants are known to be associated with prostate cancer risk, few have been associated with the risk of an aggressive disease. One such variant is rs77559646 located in ANO7. This variant has a dual function. It constitutes a missense mutation in the short isoform of ANO7 and a splice region mutation in full-length ANO7. In this study, we have analyzed the impact of the variant allele of rs77559646 on ANO7 mRNA splicing using a minigene splicing assay and by performing splicing analysis with the tools IRFinder (intron retention finder), rMATS (replicate multivariate analysis of transcript splicing) and LeafCutter on RNA sequencing data from prostate tissue of six rs77559646 variant allele carriers and 43 non-carriers. The results revealed a severe disruption of ANO7 mRNA splicing in rs77559646 variant allele carriers. Immunohistochemical analysis of prostate samples from patients homozygous for the rs77559646 variant allele demonstrated a loss of apically localized ANO7 protein. Our study is the first to provide a mechanistic explanation for the impact of a prostate cancer risk SNP on ANO7 protein production. Furthermore, the rs77559646 variant is the first known germline loss-of-function mutation described for ANO7. We suggest that loss of ANO7 contributes to prostate cancer progression.


Subject(s)
Anoctamins , Prostatic Neoplasms , RNA Splicing , Anoctamins/genetics , Base Sequence , Humans , Male , Prostatic Neoplasms/genetics , RNA, Messenger/genetics
3.
Thyroid ; 32(4): 459-471, 2022 04.
Article in English | MEDLINE | ID: mdl-35044245

ABSTRACT

Background: The human adrenal cortex undergoes several rapid remodeling steps during its lifetime. In rodents, similar remodeling occurs postnatally in the "X-zone" layer through unknown mechanisms. Furthermore, little is known regarding the impact of thyroid hormone (TH) on adrenal glands in humans. Methods: To investigate the impact of TH on adrenal pathophysiology, we created two genetic murine models mimicking human nonautoimmune hypothyroidism and hyperthyroidism. Moreover, we analyzed serum thyrotropin (TSH) and steroid hormone concentrations in patients diagnosed with congenital hypothyroidism and premature adrenarche (PA). Results: We found that TH receptor beta-mediated hypertrophy of the X-zone significantly elevated the adrenal weights of hyperthyroid women. In the hypothyroid model, the X-zone was poorly developed in both sexes. Moreover, large reciprocal changes in the expression levels of genes that regulate adrenal cortical function were observed with both models. Unexpectedly, up- and downregulation of several genes involved in catecholamine synthesis were detected in the adrenal glands of the hypothyroid and hyperthyroid models, respectively. Furthermore, TSH and adrenal steroid concentrations correlated positively in pediatric patients with congenital hypothyroidism and PA. Conclusions: Our results revealed that congenital hypothyroidism and hyperthyroidism functionally affect adrenal gland development and related steroidogenic activity, as well as the adrenal medulla.


Subject(s)
Congenital Hypothyroidism , Hyperthyroidism , Animals , Child , Congenital Hypothyroidism/genetics , Female , Gene Expression , Humans , Male , Mice , Thyroid Hormones , Thyrotropin
4.
Cell Mol Life Sci ; 78(15): 5827-5846, 2021 Aug.
Article in English | MEDLINE | ID: mdl-34155535

ABSTRACT

Stromal interaction molecule 1 (STIM1) and the ORAI1 calcium channel mediate store-operated calcium entry (SOCE) and regulate a multitude of cellular functions. The identity and function of these proteins in thyroid cancer remain elusive. We show that STIM1 and ORAI1 expression is elevated in thyroid cancer cell lines, compared to primary thyroid cells. Knock-down of STIM1 or ORAI1 attenuated SOCE, reduced invasion, and the expression of promigratory sphingosine 1-phosphate and vascular endothelial growth factor-2 receptors in thyroid cancer ML-1 cells. Cell proliferation was attenuated in these knock-down cells due to increased G1 phase of the cell cycle and enhanced expression of cyclin-dependent kinase inhibitory proteins p21 and p27. STIM1 protein was upregulated in thyroid cancer tissue, compared to normal tissue. Downregulation of STIM1 restored expression of thyroid stimulating hormone receptor, thyroid specific proteins and increased iodine uptake. STIM1 knockdown ML-1 cells were more susceptible to chemotherapeutic drugs, and significantly reduced tumor growth in Zebrafish. Furthermore, STIM1-siRNA-loaded mesoporous polydopamine nanoparticles attenuated invasion and proliferation of ML-1 cells. Taken together, our data suggest that STIM1 is a potential diagnostic and therapeutic target for treatment of thyroid cancer.


Subject(s)
Cell Proliferation/genetics , Neoplasm Proteins/genetics , Stromal Interaction Molecule 1/genetics , Thyroid Epithelial Cells/pathology , Thyroid Gland/pathology , Thyroid Neoplasms/genetics , Thyroid Neoplasms/pathology , Adolescent , Adult , Aged , Aged, 80 and over , Animals , Calcium Channels/genetics , Calcium Signaling/drug effects , Calcium Signaling/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Female , G1 Phase/drug effects , G1 Phase/genetics , Humans , Indoles/administration & dosage , Male , Middle Aged , Nanoparticles/administration & dosage , ORAI1 Protein/genetics , Polymers/administration & dosage , RNA, Small Interfering/administration & dosage , Thyroid Epithelial Cells/drug effects , Thyroid Gland/drug effects , Thyroid Neoplasms/drug therapy , Up-Regulation/drug effects , Up-Regulation/genetics , Young Adult , Zebrafish
5.
JCI Insight ; 3(20)2018 10 18.
Article in English | MEDLINE | ID: mdl-30333321

ABSTRACT

Defects in genes mediating thyroid hormone biosynthesis result in dyshormonogenic congenital hypothyroidism (CH). Here, we report homozygous truncating mutations in SLC26A7 in 6 unrelated families with goitrous CH and show that goitrous hypothyroidism also occurs in Slc26a7-null mice. In both species, the gene is expressed predominantly in the thyroid gland, and loss of function is associated with impaired availability of iodine for thyroid hormone synthesis, partially corrected in mice by iodine supplementation. SLC26A7 is a member of the same transporter family as SLC26A4 (pendrin), an anion exchanger with affinity for iodide and chloride (among others), whose gene mutations cause congenital deafness and dyshormonogenic goiter. However, in contrast to pendrin, SLC26A7 does not mediate cellular iodide efflux and hearing in affected individuals is normal. We delineate a hitherto unrecognized role for SLC26A7 in thyroid hormone biosynthesis, for which the mechanism remains unclear.


Subject(s)
Antiporters/genetics , Congenital Hypothyroidism/genetics , Goiter/genetics , Sulfate Transporters/genetics , Adult , Animals , Child , Child, Preschool , Codon, Nonsense , Congenital Hypothyroidism/diagnosis , DNA Mutational Analysis , Female , Goiter/congenital , Goiter/diagnosis , HEK293 Cells , Homozygote , Humans , Male , Mice , Mice, Knockout , Middle Aged , Pedigree , Thyroid Gland/pathology , Exome Sequencing
6.
Thyroid ; 28(10): 1372-1386, 2018 10.
Article in English | MEDLINE | ID: mdl-30132406

ABSTRACT

BACKGROUND: Constitutively active thyrotropin receptor (TSHR) mutations are the most common etiology of non-autoimmune hyperthyroidism (NAH). Thus far, the functionality of these mutations has been tested in vitro, but the in vivo models are lacking. METHODS: To understand the pathophysiology of NAH, the patient-derived constitutively active TSHR D633H mutation was introduced into the murine Tshr by homologous recombination. RESULTS: In this model, both subclinical and overt hyperthyroidism was observed, depending on the age, sex, and genotype. Homozygous mice presented hyperthyroidism at two months of age, while heterozygous animals showed only suppressed thyrotropin. Interestingly, at six months of age, thyroid hormone concentrations in all mutant mice were analogous to wild-type mice, and they showed colloid goiter with flattened thyrocytes. Strikingly, at one year of age, nearly all homozygous mice presented large papillary thyroid carcinomas. Mechanistically, this papillary thyroid carcinoma phenotype was associated with an overactive thyroid and strongly increased stainings of proliferation-, pERK-, and NKX2-1 markers, but no mutations in the "hot-spot" areas of common oncogenes (Braf, Nras, and Kras) were found. CONCLUSIONS: This is the first study to reveal the dynamic age-, sex-, and genotype-dependent development of NAH. Furthermore, the study shows that a constitutively active TSHR can trigger a malignant transformation of thyrocytes.


Subject(s)
Goiter/genetics , Hyperthyroidism/genetics , Receptors, Thyrotropin/genetics , Thyroid Cancer, Papillary/genetics , Thyroid Neoplasms/genetics , Animals , Goiter/pathology , Hyperthyroidism/pathology , Mice , Mutation , Thyroid Cancer, Papillary/pathology , Thyroid Neoplasms/pathology
7.
FASEB J ; : fj201800211R, 2018 May 25.
Article in English | MEDLINE | ID: mdl-29799790

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.

8.
Thyroid ; 26(9): 1215-24, 2016 09.
Article in English | MEDLINE | ID: mdl-27373559

ABSTRACT

BACKGROUND: Congenital hypothyroidism (CH) is defined as the lack of thyroid hormones at birth. Mutations in at least 15 different genes have been associated with this disease. While up to 20% of CH cases are hereditary, the majority of cases are sporadic with unknown etiology. Apart from a monogenic pattern of inheritance, multigenic mechanisms have been suggested to play a role in CH. The genetics of CH has not been studied in Finland so far. Therefore, multigenic sequencing of CH candidate genes was performed in a Finnish patient cohort with both familial and sporadic CH. METHODS: A targeted next-generation sequencing (NGS) panel, covering all exons of the major CH genes, was applied for 15 patients with sporadic and 11 index cases with familial CH. RESULTS: Among the familial cases, six pathogenic mutations were found in the TPO, PAX8, and TSHR genes. Furthermore, pathogenic NKX2.1 and TG mutations were identified from sporadic cases, together with likely pathogenic variants in the TG, NKX2.5, SLC26A4, and DUOX2 genes. All identified novel pathogenic mutations were confirmed by Sanger-sequencing and characterized in silico and/or in vitro. CONCLUSION: In summary, the CH panel provides an efficient, cost-effective, and multigenic screening tool for both known and novel CH gene mutations. Hence, it may be a useful method to identify accurately the genetic etiology for dyshormogenic, familial, or syndromic forms of CH.


Subject(s)
Autoantigens/genetics , Congenital Hypothyroidism/genetics , Iodide Peroxidase/genetics , Iron-Binding Proteins/genetics , Mutation , PAX8 Transcription Factor/genetics , Receptors, Thyrotropin/genetics , Cohort Studies , Female , Finland , Genetic Testing , Humans , Infant, Newborn , Male , Pedigree
9.
J Biol Chem ; 290(26): 16116-31, 2015 Jun 26.
Article in English | MEDLINE | ID: mdl-25971967

ABSTRACT

The identity of calcium channels in the thyroid is unclear. In human follicular thyroid ML-1 cancer cells, sphingolipid sphingosine 1-phosphate (S1P), through S1P receptors 1 and 3 (S1P1/S1P3), and VEGF receptor 2 (VEGFR2) stimulates migration. We show that human thyroid cells express several forms of transient receptor potential canonical (TRPC) channels, including TRPC1. In TRPC1 knockdown (TRPC1-KD) ML-1 cells, the basal and S1P-evoked invasion and migration was attenuated. Furthermore, the expression of S1P3 and VEGFR2 was significantly down-regulated. Transfecting wild-type ML-1 cells with a nonconducting TRPC1 mutant decreased S1P3 and VEGFR2 expression. In TRPC1-KD cells, receptor-operated calcium entry was decreased. To investigate whether the decreased receptor expression was due to attenuated calcium entry, cells were incubated with the calcium chelator BAPTA-AM (1,2-bis(o-aminophenoxy)ethane-N,N,N',N'-tetraacetic acid). In these cells, and in cells where calmodulin and calmodulin-dependent kinase were blocked pharmacologically, S1P3 and VEGFR2 expression was decreased. In TRPC1-KD cells, both hypoxia-inducible factor 1α expression and the secretion and activity of MMP2 and MMP9 were attenuated, and proliferation was decreased in TRPC1-KD cells. This was due to a prolonged G1 phase of the cell cycle, a significant increase in the expression of the cyclin-dependent kinase inhibitors p21 and p27, and a decrease in the expression of cyclin D2, cyclin D3, and CDK6. Transfecting TRPC1 to TRPC1-KD cells rescued receptor expression, migration, and proliferation. Thus, the expression of S1P3 and VEGFR2 is mediated by a calcium-dependent mechanism. TRPC1 has a crucial role in this process. This regulation is important for the invasion, migration, and proliferation of thyroid cancer cells.


Subject(s)
Cell Proliferation , Receptors, Lysosphingolipid/genetics , Receptors, Vascular Endothelial Growth Factor/genetics , TRPC Cation Channels/metabolism , Thyroid Neoplasms/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism , Cell Movement , Cyclin D2/genetics , Cyclin D2/metabolism , Cyclin-Dependent Kinases/genetics , Cyclin-Dependent Kinases/metabolism , Humans , Receptors, Lysosphingolipid/metabolism , Receptors, Vascular Endothelial Growth Factor/metabolism , Sphingolipids/metabolism , TRPC Cation Channels/genetics , Thyroid Neoplasms/genetics , Thyroid Neoplasms/physiopathology , Vascular Endothelial Growth Factor Receptor-2/genetics
10.
Biochim Biophys Acta ; 1853(9): 2173-82, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25892494

ABSTRACT

Caveolae are plasma membrane invaginations enriched in sterols and sphingolipids. Sphingosine kinase 1 (SK1) is an oncogenic protein that converts sphingosine to sphingosine 1-phosphate (S1P), which is a messenger molecule involved in calcium signaling. Caveolae contain calcium responsive proteins, but the effects of SK1 or S1P on caveolar calcium signaling have not been investigated. We generated a Caveolin-1-Aequorin fusion protein (Cav1-Aeq) that can be employed for monitoring the local calcium concentration at the caveolae ([Ca²âº]cav). In HeLa cells, Cav1-Aeq reported different [Ca²âº] as compared to the plasma membrane [Ca²âº] in general (reported by SNAP25-Aeq) or as compared to the cytosolic [Ca²âº] (reported by cyt-Aeq). The Ca²âº signals detected by Cav1-Aeq were significantly attenuated when the caveolar structures were disrupted by methyl-ß-cyclodextrin, suggesting that the caveolae are specific targets for Ca²âº signaling. HeLa cells overexpressing SK1 showed increased [Ca²âº]cav during histamine-induced Ca²âº mobilization in the absence of extracellular Ca²âº as well as during receptor-operated Ca²âº entry (ROCE). The SK1-induced increase in [Ca²âº]cav during ROCE was reverted by S1P receptor antagonists. In accordance, pharmacologic inhibition of SK1 reduced the [Ca²âº]cav during ROCE. S1P treatment stimulated the [Ca²âº]cav upon ROCE. The Ca²âº responses at the plasma membrane in general were not affected by SK1 expression. In summary, our results show that SK1/S1P-signaling regulates Ca²âº signals at the caveolae. This article is part of a Special Issue entitled: 13th European Symposium on Calcium.


Subject(s)
Aequorin/biosynthesis , Calcium Signaling/physiology , Caveolae/metabolism , Caveolin 1/biosynthesis , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Recombinant Fusion Proteins/biosynthesis , Aequorin/genetics , Calcium/metabolism , Calcium Signaling/drug effects , Caveolin 1/genetics , HeLa Cells , Humans , Lysophospholipids/pharmacology , Phosphotransferases (Alcohol Group Acceptor)/genetics , Recombinant Fusion Proteins/genetics , Sphingosine/analogs & derivatives , Sphingosine/pharmacology
11.
Pflugers Arch ; 466(11): 2025-34, 2014 Nov.
Article in English | MEDLINE | ID: mdl-24722829

ABSTRACT

In addition to the TSH-cyclic AMP signalling pathway, calcium signalling is of crucial importance in thyroid cells. Although the importance of calcium signalling has been thoroughly investigated for several decades, the nature of the calcium channels involved in signalling is unknown. In a recent series of investigations using the well-studied rat thyroid FRTL-5 cell line, we showed that these cells exclusively express the transient receptor potential canonical 2 (TRPC2) channel. Our results suggested that the TRPC2 channel is of significant importance in regulating thyroid cell function. These investigations were the first to show that thyroid cells express a member of the TRPC family of ion channels. In this review, we will describe the importance of the TRPC2 channel in regulating TSH receptor expression, thyroglobulin maturation, intracellular calcium and iodide homeostasis and that the channel also regulates thyroid cell proliferation.


Subject(s)
Cell Physiological Phenomena/physiology , TRPC Cation Channels/metabolism , Thyroid Gland/metabolism , Thyroid Gland/physiology , Animals , Calcium/metabolism , Humans , Rats , Receptors, Thyrotropin/metabolism , Thyroglobulin/metabolism
12.
Genesis ; 52(4): 333-40, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24395757

ABSTRACT

We have created a mouse model expressing tamoxifen-inducible Cre recombinase (CreER(T2) ) under the control of the thyroglobulin (Tg) gene promoter to be able to study the role of defined genetic modifications in the regulation of thyroid function. We chose the thyroglobulin promoter, as it is expressed specifically in the thyroid. In order to obtain reliable expression under the control of the Tg promoter, we used a P1 artificial chromosome (PAC) containing a large piece of the Tg promoter. A tamoxifen inducible CreER(T2) construct was selected to avoid the possible consequences of the gene deletion for the development of the thyroid gland, and to study the role of gene deletion in the adult thyroid. Transgenic lines (TgCreER(T2) ) carrying this construct were generated and analyzed by crossing the TgCreER(T2) mice with the ROSA26LacZ reporter strain. The activity and specificity of the Cre recombinase was tested by staining for ß-galactosidase activity and by immunohistochemistry using an anti-Cre-antibody. In the TgCreER(T2) xROSA26LacZ reporter line, Cre-mediated recombination occurred specifically in the thyrocytes only after tamoxifen administration, and no significant staining was observed in controls. The recombination efficiency was nearly complete, since almost all thyrocytes showed X-gal staining. We could also induce the recombination in utero by giving tamoxifen to the pregnant female. In addition, mice expressing TgCreER(T2) had no obvious histological changes, hormonal alterations, or different response to growth stimuli as compared to controls. These results demonstrate that the TgCreER(T2) mouse line is a powerful tool to study temporally controlled deletion of floxed genes in the thyroid.


Subject(s)
Integrases/genetics , Tamoxifen/pharmacology , Thyroid Gland/metabolism , Transcriptional Activation/drug effects , Animals , Female , Integrases/biosynthesis , Male , Mice, Inbred C57BL , Mice, Transgenic , Organ Specificity , Promoter Regions, Genetic , Recombination, Genetic , Thyroglobulin/genetics , Thyroid Gland/cytology
13.
Mol Cell Endocrinol ; 374(1-2): 10-21, 2013 Jul 15.
Article in English | MEDLINE | ID: mdl-23578584

ABSTRACT

Mammalian transient receptor potential (TRP) channels are involved in many physiologically important processes. Here, we have studied the significance of the TRPC2 channel in the regulation of rat thyroid FRTL-5 cell proliferation, migration, adhesion and invasion, using stable TRPC2 (shTRPC2) knock-down cells. In the shTRPC2 cells, proliferation was decreased due to a prolonged G1/S cell cycle phase. The tumor suppressor p53 and the cyclin-dependant kinase inhibitors p27 and p21 were upregulated. Cell invasion, adhesion and migration were also attenuated in shTRPC2 cells, probably due to decreased activity of both Rac and calpain, and a decreased secretion and activity of matrix metalloproteinase 2. The attenuated proliferation, migration, invasion and ATP-evoked calcium entry was mimicked by overexpressing a non-conducting, truncated TRPC2 (TRPC2-DN) in wild type cells, and was reversed by overexpression of TRPC2-GFP in shTRPC2 cells. In conclusion, TRPC2 is an important regulator of rat thyroid cell function.


Subject(s)
Gene Expression Regulation , TRPC Cation Channels/genetics , Thyroid Gland/metabolism , Adenosine Triphosphate/metabolism , Animals , Calcium/metabolism , Calpain/genetics , Calpain/metabolism , Cell Adhesion , Cell Line , Cell Movement , Cell Proliferation , Cyclin-Dependent Kinase Inhibitor p27/genetics , Cyclin-Dependent Kinase Inhibitor p27/metabolism , G1 Phase Cell Cycle Checkpoints , Gene Knockdown Techniques , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 2/metabolism , Protein Structure, Tertiary , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Rats , Signal Transduction , TRPC Cation Channels/metabolism , Thyroid Gland/cytology , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , p21-Activated Kinases/genetics , p21-Activated Kinases/metabolism
14.
J Cell Physiol ; 228(4): 814-23, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23018590

ABSTRACT

The initial step in a synthesis of thyroid hormones is the uptake of iodide from the circulation. Iodide (I(-)) is transported into thyroid cells via a Na(+)/I(-) symporter (NIS), which is electrogenic and thus sensitive to alterations in membrane potential (V(m)). I(-) is then released to the lumen of thyroid follicles where the hormones are synthesised and stored. The mechanisms of I(-) release to follicle lumen are poorly characterised. Our whole-cell voltage clamp recordings revealed the presence of a Ca(2+) activated Cl(-) current (CaCC) in Fisher rat thyroid cell line 5 (FRTL-5). Transcripts of anoctamin 1 (ANO1) and anoctamin 10 (ANO10), putative molecular constituents of CaCC, were detected. The anion channels underlying CaCC are highly permeable to I(-). Both niflumic acid (NFA) and 2-aminoethyl diphenylborinate (2-APB), antagonists of CaCC and transient receptor potential channels, respectively, inhibited CaCC. Canonical transient receptor potential channel 2 (TRPC2) is the only TRPC member present in FRTL-5 cells. The activation rate of CaCC was markedly slower in shTRPC2 knock-down cells, indicating that Ca(2+) entry via TRPC2 contributes to CaCC activation. The uptake of iodide was enhanced and the resting V(m) was more depolarised in TRPC2 knock-down cells. We suggest that the interplay between TRPC2 and ANO1 may have dual effects on iodide transport, modulating I(-) release via ANO channels and I(-) uptake via the V(m) sensitive NIS.


Subject(s)
Chloride Channels/metabolism , Homeostasis/physiology , Iodides/metabolism , TRPC Cation Channels/metabolism , Thyroid Gland/metabolism , Voltage-Dependent Anion Channels/metabolism , Animals , Anions/metabolism , Anoctamin-1 , Boron Compounds/pharmacology , Calcium/metabolism , Cell Line , Chlorides/metabolism , Homeostasis/drug effects , Membrane Potentials/drug effects , Membrane Potentials/physiology , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/drug effects , Membrane Proteins/metabolism , Niflumic Acid/pharmacology , Rats , TRPC Cation Channels/antagonists & inhibitors , Thyroid Gland/drug effects
15.
J Biol Chem ; 287(53): 44345-60, 2012 Dec 28.
Article in English | MEDLINE | ID: mdl-23144458

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 PKCß1 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.


Subject(s)
Calcium-Binding Proteins/metabolism , Calcium/metabolism , Membrane Proteins/metabolism , Protein Kinase C-delta/metabolism , TRPC Cation Channels/metabolism , Thyroid Gland/metabolism , Animals , Calcium-Binding Proteins/genetics , Cell Line, Tumor , Gene Expression Regulation , Homeostasis , Membrane Proteins/genetics , Protein Kinase C-delta/genetics , Rats , Sarcoplasmic Reticulum Calcium-Transporting ATPases/genetics , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Stromal Interaction Molecule 2 , TRPC Cation Channels/genetics , Thyroid Gland/enzymology
16.
Mol Endocrinol ; 26(12): 2046-57, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23015753

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.


Subject(s)
Calcium/metabolism , Cyclic AMP/metabolism , Receptors, Thyrotropin/metabolism , TRPC Cation Channels/metabolism , Thyroglobulin/metabolism , Thyroid Gland/metabolism , Animals , Cell Line , Extracellular Signal-Regulated MAP Kinases/metabolism , MAP Kinase Signaling System , Protein Processing, Post-Translational , RNA Interference , RNA, Small Interfering , Rats , TRPC Cation Channels/genetics , Thyroid Gland/cytology
17.
Adv Exp Med Biol ; 704: 125-34, 2011.
Article in English | MEDLINE | ID: mdl-21290292

ABSTRACT

Relatively little is known in regard to the physiological significance of TRPC2 and its regulation or interaction with other calcium regulating signalling molecules. In rodents, however, the importance of TRPC2 is indisputable. In mice, transcripts for TRPC2 have been found in testis, sperm, in neurons in the vomeronasal organ, and both in the dorsal root ganglion and in the brain. In rats, TRPC2 is thought to be expressed exclusively in the vomeronasal organ. In mice, TRPC2 is of importance in regulating both sexual and social behaviour. In sperm, TRPC2 is of importance in the acrosome reaction. This review will summarize the known physiological effects of TRPC2 channels, and the regulation of the function of the channel. In addition, some new preliminary data on the role of TRPC2 in rat thyroid cells will be presented.


Subject(s)
TRPC Cation Channels/physiology , Animals , Calcium/metabolism , Humans , Ion Transport , Male , Mice , Rats , Signal Transduction , Spermatozoa/metabolism , TRPC Cation Channels/metabolism
18.
BMC Cell Biol ; 11: 45, 2010 Jun 24.
Article in English | MEDLINE | ID: mdl-20573281

ABSTRACT

BACKGROUND: Sphingosine-1-phosphate (S1P) is a bioactive lipid that regulates a multitude of cellular functions, including cell proliferation, survival, migration and angiogenesis. S1P mediates its effects either by signaling through G protein-coupled receptors (GPCRs) or through an intracellular mode of action. In this study, we have investigated the mechanism behind S1P-induced survival signalling. RESULTS: We found that S1P protected cells from FasL-induced cell death in an NF-kappaB dependent manner. NF-kappaB was activated by extracellular S1P via S1P2 receptors and Gi protein signaling. Our study also demonstrates that extracellular S1P stimulates cells to rapidly produce and secrete additional S1P, which can further amplify the NF-kappaB activation. CONCLUSIONS: We propose a self-amplifying loop of autocrine S1P with capacity to enhance cell survival. The mechanism provides increased understanding of the multifaceted roles of S1P in regulating cell fate during normal development and carcinogenesis.


Subject(s)
Lysophospholipids/biosynthesis , NF-kappa B/metabolism , Receptors, Lysosphingolipid/metabolism , Sphingosine/analogs & derivatives , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Autocrine Communication , Calcium-Calmodulin-Dependent Protein Kinases/antagonists & inhibitors , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/genetics , Enzyme Activation/drug effects , Enzyme Activation/genetics , Fas Ligand Protein/metabolism , Feedback, Physiological , Flavonoids/pharmacology , Humans , Lysophospholipids/antagonists & inhibitors , Lysophospholipids/genetics , Pyrazoles/pharmacology , Pyridines/pharmacology , RNA, Small Interfering/genetics , Receptors, Lysosphingolipid/genetics , Signal Transduction/drug effects , Signal Transduction/genetics , Sphingosine/antagonists & inhibitors , Sphingosine/biosynthesis , Sphingosine/genetics , Transgenes/genetics
19.
Endocrinology ; 151(7): 2994-3005, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20501673

ABSTRACT

Sphingosine 1-phosphate (S1P) and vascular endothelial growth factor receptor 2 (VEGFR-2) signaling have been shown to integrate in many biological processes. The follicular thyroid carcinoma cell line ML-1 expresses VEGFR-2 and secretes substantial amounts of both vascular endothelial growth factor (VEGF)-A and VEGF-C. ML-1 cells also express S1P-receptors (S1P(1-3,5)). S1P is able to phosphorylate VEGFR-2, and inhibiting VEGFR-2 attenuates S1P-induced migration and down-regulates S1P(1) expression in ML-1 cells. In the present study, we focused on the interactions between S1P(1) and VEGFR-2. We show that S1P receptors form complexes with VEGFR-2 and that the S1P(1)/VEGFR-2 complex associates with protein kinase C (PKC)-alpha and ERK1/2. Furthermore, the complex evokes bidirectional signaling since the S1P-induced ERK1/2 phosphorylation is sensitive to VEGFR-2 kinase inhibition and VEGF-A-induced ERK1/2 phosphorylation is sensitive to pertussis toxin treatment as well as S1P(1) small interfering RNA (siRNA) treatment. Both S1P- and VEGF-A-induced haptotaxis is sensitive to pertussis toxin treatment and S1P(1) siRNA treatment. Phosphorylation of ERK1/2 evoked by both VEGF-A and the S1P(1) agonist SEW-2871 is inhibited by PKC-alpha and PKC-betaI siRNA. We hypothesize that VEGFR-2 forms a signaling complex with S1P(1), evoking bidirectional signaling regulating both ERK1/2 phosphorylation and haptotaxis of ML-1 cells.


Subject(s)
Cell Movement/drug effects , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Protein Kinase C-alpha/metabolism , Receptors, Lysosphingolipid/metabolism , Thyroid Neoplasms/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism , Blotting, Western , Cell Line, Tumor , Cell Movement/genetics , Humans , Immunohistochemistry , Immunoprecipitation , Lysophospholipids/pharmacology , Phosphorylation/drug effects , Protein Binding/drug effects , Protein Binding/genetics , Receptors, Lysosphingolipid/agonists , Receptors, Lysosphingolipid/antagonists & inhibitors , Receptors, Lysosphingolipid/genetics , Signal Transduction/drug effects , Signal Transduction/genetics , Sphingosine/analogs & derivatives , Sphingosine/pharmacology , Thyroid Neoplasms/genetics , Vascular Endothelial Growth Factor A/pharmacology , Vascular Endothelial Growth Factor Receptor-2/agonists , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors
20.
Endocrinology ; 150(11): 5125-34, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19797403

ABSTRACT

Calcium entry is one of the main regulators of intracellular signaling. Here, we have described the importance of sphingosine, sphingosine kinase 1 (SK1), and sphingosine 1-phosphate (S1P) in regulating calcium entry in thyroid FRTL-5 cells. In cells incubated with the phosphatase inhibitor calyculin A, which evokes calcium entry without mobilizing sequestered intracellular calcium, sphingosine inhibited calcium entry in a concentration-dependent manner. Furthermore, inhibiting SK1 or the ATP-binding cassette ABCC1 multidrug transporter attenuated calcium entry. The addition of exogenous S1P restored calcium entry. Neither sphingosine nor inhibition of SK1 attenuated thapsigargin-evoked calcium entry. Blocking S1P receptor 2 or phospholipase C attenuated calcium entry, whereas blocking S1P receptor 3 did not. Overexpression of wild-type SK1, but not SK2, enhanced calyculin-evoked calcium entry compared with mock-transfected cells, whereas calcium entry was decreased in cells transfected with the dominant-negative G82D SK1 mutant. Exogenous S1P restored calcium entry in G82D cells. Our results suggest that the calcium entry pathway is blocked by sphingosine and that activation of SK1 and the production of S1P, through an autocrine mechanism, facilitate calcium entry through activation of S1P receptor 2. This is a novel mechanism by which the sphingosine-S1P rheostat regulates cellular calcium homeostasis.


Subject(s)
Autocrine Communication , Calcium/metabolism , Lysophospholipids/metabolism , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Signal Transduction , Sphingosine/analogs & derivatives , Thyroid Gland/metabolism , Animals , Cell Line , Rats , Receptors, Lysosphingolipid/metabolism , Sphingosine/metabolism , Thyroid Gland/cytology , Thyroid Gland/enzymology
SELECTION OF CITATIONS
SEARCH DETAIL
...