Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
2.
Physiol Int ; 110(3): 251-266, 2023 Sep 05.
Article in English | MEDLINE | ID: mdl-37540593

ABSTRACT

Background: Hormone level changes after heart surgeries are a widely observed phenomenon due to neurohormonal feedback mechanisms that may affect postoperative morbidity and mortality. The current study aimed to analyze the changes in thyroid and sex hormones in the first 24 postoperative hours after heart surgery. Methods: This prospective, observational study (registered on ClinicalTrials.gov: NCT03736499; 09/11/2018) included 49 patients who underwent elective cardiac surgical procedures at a tertiary heart center between March 2019 and December 2019. Thyroid hormones, including thyroid-stimulating hormone (TSH), triiodothyronine (T3), and thyroxine (T4), and sex hormones, including prolactin (PRL) and total testosterone, were measured preoperatively and at 24 h postoperatively. Results: Significant decreases in serum TSH (P < 0.001), T3 (P < 0.001) and total testosterone (P < 0.001) levels were noted, whereas T4 (P = 0.554) and PRL (P = 0.616) did not significantly change. Intensive care unit (ICU) hours (P < 0.001), mechanical ventilation (P < 0.001) and Vasoactive-Inotropic Score (VIS) (P = 0.006) were associated with postoperative T3 level. ICU hours were associated with postoperative T4 level (P = 0.028). Postoperative and delta testosterone levels were in connection with lengths of stay in ICU (P = 0.032, P = 0.010 respectively). Model for End-Stage Liver Disease (MELD) scores were associated with thyroid hormone levels and serum testosterone. Conclusions: T3 may represent a marker of nonthyroidal illness syndrome and testosterone may reflect hepatic dysfunction. In addition, PRL may act as a stress hormone in female patients.


Subject(s)
Cardiac Surgical Procedures , End Stage Liver Disease , Humans , Female , Prospective Studies , Severity of Illness Index , Thyroid Hormones , Thyrotropin , Cardiac Surgical Procedures/adverse effects , Testosterone
3.
Arch Endocrinol Metab ; 67(2): 256-261, 2023 Mar 10.
Article in English | MEDLINE | ID: mdl-36913678

ABSTRACT

Objective: The aim of this study was to investigate how polarized sodium iodide symporter (NIS) expression may regulate iodide metabolism in vivo. Materials and methods: Polarized NIS expression was analyzed in tissues that accumulate iodide by the use of immunohistochemistry and polyclonal antibody against the C-terminal end of human NIS (hNIS). Results: Iodide absorption in the human intestine occurs via NIS expressed in the apical membrane. Iodide is secreted into the lumen of the stomach and salivary glands via NIS expressed in the basolateral membrane and then circulates back from the small intestine to the bloodstream via NIS expressed in the apical membrane. Conclusion: Polarized NIS expression in the human body regulates intestinal-bloodstream recirculation of iodide, perhaps prolonging the availability of iodide in the bloodstream. This leads to more efficient iodide trapping by the thyroid gland. Understanding the regulation and manipulating gastrointestinal iodide recirculation could increase radioiodine availability during theranostic NIS applications.


Subject(s)
Iodine , Symporters , Humans , Iodides/metabolism , Iodine Radioisotopes , Human Body , Symporters/metabolism
4.
Arch. endocrinol. metab. (Online) ; 67(2): 256-261, 2023. graf
Article in English | LILACS-Express | LILACS | ID: biblio-1429726

ABSTRACT

ABSTRACT Objective: The aim of this study was to investigate how polarized sodium iodide symporter (NIS) expression may regulate iodide metabolism in vivo. Materials and methods: Polarized NIS expression was analyzed in tissues that accumulate iodide by the use of immunohistochemistry and polyclonal antibody against the C-terminal end of human NIS (hNIS). Results: Iodide absorption in the human intestine occurs via NIS expressed in the apical membrane. Iodide is secreted into the lumen of the stomach and salivary glands via NIS expressed in the basolateral membrane and then circulates back from the small intestine to the bloodstream via NIS expressed in the apical membrane. Conclusion: Polarized NIS expression in the human body regulates intestinal-bloodstream recirculation of iodide, perhaps prolonging the availability of iodide in the bloodstream. This leads to more efficient iodide trapping by the thyroid gland. Understanding the regulation and manipulating gastrointestinal iodide recirculation could increase radioiodine availability during theranostic NIS applications.

5.
J Cardiothorac Vasc Anesth ; 33(6): 1629-1635, 2019 Jun.
Article in English | MEDLINE | ID: mdl-30467031

ABSTRACT

OBJECTIVE: The effect of thyroid dysfunction on adverse outcomes has been studied in many different patient populations. The objective of this study was to investigate the effect of thyroid hormone supplementation of donors and recipients on postoperative outcomes after orthotopic heart transplantation. DESIGN: Retrospective. SETTING: Single center, university hospital. PARTICIPANTS: Two-hundred and sixty-six consecutive patients undergoing heart transplantation. INTERVENTIONS: No interventions. MEASUREMENTS AND MAIN RESULTS: Demographic, hemodynamic, and clinical characteristics; donor and recipient United Network for Organ Sharing scores; and information on thyroid hormone support of donors and recipients were recorded. During the median follow-up of 4.59 years (interquartile range 4.26-4.92 y), 70 patients (26.3%) died. After adjustments were made for the United Network for Organ Sharing score, recipients who were treated preoperatively with l-thyroxine had a lower risk for all-cause mortality (adjusted hazard ratio [HR] 0.24, 95% confidence interval [CI] 0.06-0.98; p = 0.047) compared with recipients who were not treated with l-thyroxine. In addition, l-thyroxine treatment of donors was associated with a better recipient survival (HR 0.31, 95% CI 0.11-0.87; p = 0.025). CONCLUSIONS: Pretransplantation thyroid hormone supplementation of donors and recipients was associated with improved long-term survival after heart transplantation.


Subject(s)
Heart Transplantation/adverse effects , Postoperative Complications/prevention & control , Preoperative Care/methods , Thyroid Diseases/prevention & control , Thyroxine/therapeutic use , Tissue Donors , Transplant Recipients , Adult , Aged , Female , Follow-Up Studies , Humans , Hungary/epidemiology , Incidence , Male , Middle Aged , Postoperative Complications/epidemiology , Postoperative Complications/etiology , Prognosis , Registries , Retrospective Studies , Survival Rate/trends , Thyroid Diseases/epidemiology , Thyroid Diseases/etiology , Time Factors , Treatment Outcome
6.
Proc Natl Acad Sci U S A ; 108(44): 17933-8, 2011 Nov 01.
Article in English | MEDLINE | ID: mdl-22011571

ABSTRACT

I(-) uptake in the thyroid, the first step in thyroid hormone biosynthesis, is mediated by the Na(+)/I(-) symporter (NIS) with an electrogenic 2Na(+):1I(-) stoichiometry. We have obtained mechanistic information on NIS by characterizing the congenital I(-) transport defect-causing NIS mutant G93R. This mutant is targeted to the plasma membrane but is inactive. Substitutions at position 93 show that the longer the side chain of the neutral residue at this position, the higher the K(m) for the anion substrates. Unlike WT NIS, which mediates symport of Na(+) and the environmental pollutant perchlorate electroneutrally, G93T/N/Q/E/D NIS, strikingly, do it electrogenically with a 21 stoichiometry. Furthermore, G93E/Q NIS discriminate between anion substrates, a discovery with potential clinical relevance. A 3D homology model of NIS based on the structure of the bacterial Na(+)/galactose transporter identifies G93 as a critical player in the mechanism of the transporter: the changes from an outwardly to an inwardly open conformation during the transport cycle use G93 as a pivot.


Subject(s)
Iodides/metabolism , Sodium/metabolism , Symporters/metabolism , Animals , Anions , COS Cells , Cell Line , Chlorocebus aethiops , Humans , Models, Molecular , Symporters/chemistry
7.
Proc Natl Acad Sci U S A ; 104(51): 20250-5, 2007 Dec 18.
Article in English | MEDLINE | ID: mdl-18077370

ABSTRACT

The Na(+)/I(-) symporter (NIS) is a key plasma membrane protein that mediates active I(-) uptake in the thyroid, lactating breast, and other tissues with an electrogenic stoichiometry of 2 Na(+) per I(-). In the thyroid, NIS-mediated I(-) uptake is the first step in the biosynthesis of the iodine-containing thyroid hormones, which are essential early in life for proper CNS development. In the lactating breast, NIS mediates the translocation of I(-) to the milk, thus supplying this essential anion to the nursing newborn. Perchlorate (ClO(4)(-)) is a well known competitive inhibitor of NIS. Exposure to food and water contaminated with ClO(4)(-) is common in the U.S. population, and the public health impact of such exposure is currently being debated. To date, it is still uncertain whether ClO(4)(-) is a NIS blocker or a transported substrate of NIS. Here we show in vitro and in vivo that NIS actively transports ClO(4)(-), including ClO(4)(-) translocation to the milk. A simple mathematical fluxes model accurately predicts the effect of ClO(4)(-) transport on the rate and extent of I(-) accumulation. Strikingly, the Na(+)/ ClO(4)(-) transport stoichiometry is electroneutral, uncovering that NIS translocates different substrates with different stoichiometries. That NIS actively concentrates ClO(4)(-) in maternal milk suggests that exposure of newborns to high levels of ClO(4)(-) may pose a greater health risk than previously acknowledged because ClO(4)(-) would thus directly inhibit the newborns' thyroidal I(-) uptake.


Subject(s)
Environmental Pollutants/metabolism , Iodine/metabolism , Perchlorates/metabolism , Symporters/metabolism , Animals , Biological Transport , Cell Line , Dogs , Iodine/analysis , Kinetics , Milk/chemistry , Models, Theoretical , Perchlorates/analysis , Rats , Transfection
8.
BMC Cancer ; 7: 5, 2007 Jan 10.
Article in English | MEDLINE | ID: mdl-17214887

ABSTRACT

BACKGROUND: The sodium/iodide symporter (NIS) is a plasma membrane glycoprotein that mediates iodide (I-) transport in the thyroid, lactating breast, salivary glands, and stomach. Whereas NIS expression and regulation have been extensively investigated in healthy and neoplastic thyroid and breast tissues, little is known about NIS expression and function along the healthy and diseased gastrointestinal tract. METHODS: Thus, we investigated NIS expression by immunohistochemical analysis in 155 gastrointestinal tissue samples and by immunoblot analysis in 17 gastric tumors from 83 patients. RESULTS: Regarding the healthy Gl tract, we observed NIS expression exclusively in the basolateral region of the gastric mucin-producing epithelial cells. In gastritis, positive NIS staining was observed in these cells both in the presence and absence of Helicobacter pylori. Significantly, NIS expression was absent in gastric cancer, independently of its histological type. Only focal faint NIS expression was detected in the direct vicinity of gastric tumors, i.e., in the histologically intact mucosa, the expression becoming gradually stronger and linear farther away from the tumor. Barrett mucosa with junctional and fundic-type columnar metaplasia displayed positive NIS staining, whereas Barrett mucosa with intestinal metaplasia was negative. NIS staining was also absent in intestinalized gastric polyps. CONCLUSION: That NIS expression is markedly decreased or absent in case of intestinalization or malignant transformation of the gastric mucosa suggests that NIS may prove to be a significant tumor marker in the diagnosis and prognosis of gastric malignancies and also precancerous lesions such as Barrett mucosa, thus extending the medical significance of NIS beyond thyroid disease.


Subject(s)
Barrett Esophagus/metabolism , Biomarkers, Tumor/antagonists & inhibitors , Biomarkers, Tumor/genetics , Gene Expression Regulation, Neoplastic/genetics , Intestinal Mucosa/metabolism , Stomach Neoplasms/metabolism , Symporters/antagonists & inhibitors , Symporters/genetics , Barrett Esophagus/genetics , Barrett Esophagus/pathology , Biomarkers, Tumor/biosynthesis , Down-Regulation/genetics , Female , Humans , Intestinal Mucosa/pathology , Iodine/metabolism , Male , Metaplasia/metabolism , Metaplasia/pathology , Middle Aged , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Symporters/biosynthesis
9.
Mol Endocrinol ; 20(5): 1121-37, 2006 May.
Article in English | MEDLINE | ID: mdl-16439463

ABSTRACT

The sodium/iodide symporter (NIS) mediates a remarkably effective targeted radioiodide therapy in thyroid cancer; this approach is an emerging candidate for treating other cancers that express NIS, whether endogenously or by exogenous gene transfer. Thus far, the only extrathyroidal malignancy known to express functional NIS endogenously is breast cancer. Therapeutic efficacy in thyroid cancer requires that radioiodide uptake be maximized in tumor cells by manipulating well-known regulatory factors of NIS expression in thyroid cells, such as TSH, which stimulates NIS expression via cAMP. Similarly, therapeutic efficacy in breast cancer will likely depend on manipulating NIS regulation in mammary cells, which differs from that in the thyroid. Human breast adenocarcinoma MCF-7 cells modestly express endogenous NIS when treated with all-trans-retinoic acid (tRa). We report here that hydrocortisone and ATP each markedly stimulates tRa-induced NIS protein expression and plasma membrane targeting in MCF-7 cells, leading to at least a 100% increase in iodide uptake. Surprisingly, the adenyl cyclase activator forskolin, which promotes NIS expression in thyroid cells, markedly decreases tRa-induced NIS protein expression in MCF-7 cells. Isobutylmethylxanthine increases tRa-induced NIS expression in MCF-7 cells, probably through a purinergic signaling system independent of isobutylmethylxanthine's action as a phosphodiesterase inhibitor. We also observed that neither iodide, which at high concentrations down-regulates NIS in the thyroid, nor cAMP has a significant effect on NIS expression in MCF-7 cells. Our findings may open new strategies for breast-selective pharmacological modulation of functional NIS expression, thus improving the feasibility of using radioiodide to effectively treat breast cancer.


Subject(s)
Adenosine Triphosphate/pharmacology , Breast Neoplasms/metabolism , Hydrocortisone/pharmacology , Iodides/metabolism , Symporters/agonists , 1-Methyl-3-isobutylxanthine/pharmacology , Adenylyl Cyclase Inhibitors , Adenylyl Cyclases/pharmacology , Biological Transport/drug effects , Cell Membrane/metabolism , Cells, Cultured , Colforsin/pharmacology , Female , Humans , Phosphodiesterase Inhibitors/pharmacology , Signal Transduction , Symporters/metabolism , Thyroid Gland/metabolism , Tretinoin/pharmacology
10.
Clin Cancer Res ; 10(13): 4294-302, 2004 Jul 01.
Article in English | MEDLINE | ID: mdl-15240514

ABSTRACT

PURPOSE: The Na(+)/I(-) symporter (NIS) is a key plasma membrane protein that mediates active iodide (I(-)) transport in the thyroid, lactating breast, and other tissues. Functional NIS expression in thyroid cancer accounts for the longstanding success of radioactive iodide ((131)I) ablation of metastases after thyroidectomy. Breast cancer is the only other cancer demonstrating endogenous functional NIS expression. Until now, NIS activity in breast cancer metastases (BCM) was unproven. EXPERIMENTAL DESIGN: Twenty-seven women were scanned with (99m)TcO(4)(-) or (123)I(-) to assess NIS activity in their metastases. An (131)I dosimetry study was offered to patients with I(-)-accumulating tumors. Selective down-regulation of thyroid NIS was tested in 13 patients with T(3) and in one case with T(3) + methimazole (MMI; blocks I(-) organification). NIS expression was evaluated in index and/or metastatic tumor samples by immunohistochemistry. RESULTS: I(-) uptake was noted in 25% of NIS-expressing tumors (two of eight). The remaining cases did not show NIS expression or activity. Thyroid I(-) uptakes were decreased to

Subject(s)
Breast Neoplasms/pathology , Down-Regulation , Iodides/pharmacokinetics , Neoplasm Metastasis , Symporters/physiology , Breast Neoplasms/metabolism , Breast Neoplasms/radiotherapy , Female , Humans , Immunohistochemistry , Neoplasms , Radiometry , Radionuclide Imaging , Symporters/metabolism , Thyroid Gland/pathology , Thyroid Gland/radiation effects , Thyroid Neoplasms/metabolism , Time Factors , Triiodothyronine/metabolism
11.
J Nucl Med ; 45(3): 500-7, 2004 Mar.
Article in English | MEDLINE | ID: mdl-15001694

ABSTRACT

UNLABELLED: Pertechnetate (as (99m)TcO(4)(-)), (123)I(-), and (131)I(-) have a long and successful history of use in the diagnosis and therapy of thyroid cancer, with uptake into thyroid tissue mediated by the sodium-iodide symporter (NIS). NIS has also emerged as a potential target for radiotherapy of nonthyroid malignancies that express the endogenous or transfected symporter. Perrhenates (as (188)ReO(4)(-) and (186)ReO(4)(-)) are promising therapeutic substrates of NIS, although less is known about their behavior in vivo. In this study, we endeavored to characterize the biologic behavior of perrhenate, especially in relation to iodide and pertechnetate, to better explore its possible therapeutic role. METHODS: We describe the simultaneous biodistribution and uptake in vivo of iodide, pertechnetate, and perrhenate in groups of healthy CD1 mice, either with or without coadministration of perchlorate (ClO(4)(-)), a potent NIS inhibitor. Animals administered single radiopharmaceuticals were imaged as a means of illustrating these findings. Kinetic properties of perrhenate were compared with those of iodide in a stably transfected NIS-bearing Madin-Darby canine kidney (MDCK) cell line. RESULTS: Biodistributions of iodide, pertechnetate, and perrhenate in live mice were remarkably similar. Activity in salivary gland and stomach was severalfold greater than in blood, remained elevated over the initial 2 h, and subsequently washed out. A similar pattern characterized pertechnetate and perrhenate uptake by the thyroid, in which the 2-h concentration was slightly more elevated than at the 20-min time point. However, uptake subsequently decreased by 19 h. In contrast, iodide continued to increase through the 19-h time point, presumably as a result of organification. The addition of perchlorate sharply decreased uptake of all 3 radiopharmaceuticals by the stomach, salivary glands, and thyroid and resulted in their rapid clearance, paralleling blood-pool clearance. In tissues that do not express NIS (liver, muscle, spleen), uptake of all 3 radiopharmaceuticals was low and rapidly decreased over time, paralleling blood-pool clearance. Similar findings were seen in kidney, where only minimal amounts of NIS are expressed in tubular cells. In stably transfected MDCK cells, steady-state accumulation of iodide was approximately 4-fold higher than that of perrhenate at 30 min. No active transport was demonstrated in nontransfected MDCK cell lines or after perchlorate administration. Uptake values measured at different concentrations of substrate demonstrated saturation kinetics. Apparent maximal velocity values for perrhenate and iodide were 25.6 +/- 1.4 and 106 +/- 3.2 pmol/ micro g, respectively, and corresponding affinity constant values were 4.06 +/- 0.87 and 24.6 +/- 1.81 micro mol/L. CONCLUSION: Perrhenate is avidly taken up by NIS in a manner similar to iodide and pertechnetate in vivo, with the exception of organification of iodide by the thyroid. By more fully appreciating the behavior of perrhenate, especially in relation to iodide and pertechnetate, we can better realize its potential role in the diagnosis and therapy of NIS-bearing tissues.


Subject(s)
Iodine Radioisotopes/pharmacokinetics , Kidney/metabolism , Rhenium/pharmacokinetics , Sodium Pertechnetate Tc 99m/pharmacokinetics , Symporters/metabolism , Technetium/pharmacokinetics , Animals , Cell Line , Dogs , Kidney/diagnostic imaging , Metabolic Clearance Rate , Mice , Organ Specificity , Radionuclide Imaging , Tissue Distribution , Whole-Body Counting
13.
J Clin Endocrinol Metab ; 88(4): 1880-8, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12679487

ABSTRACT

Extrathyroidal cancers could potentially be targeted with (131)I, if the Na(+)/I(-) symporter (NIS) were functional. Using immunohistochemical methods we probed 1278 human samples with anti-NIS antibody, including 253 thyroid and 169 breast conventional whole tissue sections (CWTS). Four high density tissue microarrays containing a wide variety of breast lesions, normal tissues, and carcinoma cores were tested. The results of the normal microarray were corroborated in 50 CWTS. Nineteen of 34 normal tissues, including bladder, colon, endometrium, kidney, prostate, and pancreas, expressed NIS. Nineteen of 25 carcinomas demonstrated NIS immunopositivity; 55.7% of 479 carcinoma microarray cores expressed NIS, including prostate (74%), ovary (73%), lung (65%), colon (62.6%), and endometrium (56%). NIS protein was present in 75% benign thyroid lesions, 73% thyroid cancers, 30% normal-appearing, peritumoral breasts, 88% ductal carcinomas in situ, and 76% invasive breast carcinoma CWTS. Comparatively, breast microarray cores had lower immunoreactivity. Plasma membrane immunopositivity was confirmed in thyrocytes, salivary ductal, gastric mucosa, and lactating mammary cells. In other tissues, immunoreactivity was predominantly intracellular, particularly in malignant lesions. Thus, NIS is present in many normal epithelial tissues and is predominantly expressed intracellularly in many carcinomas. Elucidating the regulatory mechanisms that render NIS functional in extrathyroidal carcinomas may make (131)I therapy feasible.


Subject(s)
Breast Neoplasms/chemistry , Immunohistochemistry , Neoplasms/chemistry , Symporters/analysis , Thyroid Neoplasms/chemistry , Colonic Neoplasms/chemistry , Endometrial Neoplasms/chemistry , Female , Humans , Iodine Radioisotopes/therapeutic use , Lung Neoplasms/chemistry , Male , Organ Specificity , Ovarian Neoplasms/chemistry , Prostatic Neoplasms/chemistry , Thyroid Neoplasms/radiotherapy
14.
Endocr Rev ; 24(1): 48-77, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12588808

ABSTRACT

The Na(+)/I(-) symporter (NIS) is an integral plasma membrane glycoprotein that mediates active I(-) transport into the thyroid follicular cells, the first step in thyroid hormone biosynthesis. NIS-mediated thyroidal I(-) transport from the bloodstream to the colloid is a vectorial process made possible by the selective targeting of NIS to the basolateral membrane. NIS also mediates active I(-) transport in other tissues, including salivary glands, gastric mucosa, and lactating mammary gland, in which it translocates I(-) into the milk for thyroid hormone biosynthesis by the nursing newborn. NIS provides the basis for the effective diagnostic and therapeutic management of thyroid cancer and its metastases with radioiodide. NIS research has proceeded at an astounding pace after the 1996 isolation of the rat NIS cDNA, comprising the elucidation of NIS secondary structure and topology, biogenesis and posttranslational modifications, transcriptional and posttranscriptional regulation, electrophysiological analysis, isolation of the human NIS cDNA, and determination of the human NIS genomic organization. Clinically related topics include the analysis of congenital I(-) transport defect-causing NIS mutations and the role of NIS in thyroid cancer. NIS has been transduced into various kinds of cancer cells to render them susceptible to destruction with radioiodide. Most dramatically, the discovery of endogenous NIS expression in more than 80% of human breast cancer samples has raised the possibility that radioiodide may be a valuable novel tool in breast cancer diagnosis and treatment.


Subject(s)
Symporters/genetics , Symporters/physiology , Autoimmune Diseases , Biological Transport/genetics , Breast Neoplasms , Gene Expression Regulation , Humans , Iodides/metabolism , Mutation , Organ Specificity , Signal Transduction , Thyroid Diseases , Thyroid Neoplasms , Transcription, Genetic , Transfection
15.
Mol Cell Endocrinol ; 213(1): 59-70, 2003 Dec 31.
Article in English | MEDLINE | ID: mdl-15062574

ABSTRACT

The Na(+)/I(-) symporter (NIS) is a plasma membrane glycoprotein that mediates active iodide uptake in the thyroid-the essential first step in thyroid hormone biosynthesis-and in other tissues, such as salivary and lactating mammary glands. Thyroidal radioiodide uptake has been used for over 60 years in the diagnosis and effective treatment of thyroid cancer and other diseases. However, the NIS cDNA was only isolated in 1996 by expression cloning in Xenopus laevis oocytes, marking the beginning of the molecular characterization of NIS and the study of its regulation, both in the thyroid and other tissues. One of the most exciting current areas of NIS research-radioiodide treatment of extrathyroidal cancers-was launched by the discovery of functional expression of endogenous NIS in breast cancer and by the ectopic transfer of the NIS gene into otherwise non NIS-expressing cancers. This review summarizes the main findings in NIS research, emphasizing the most recent developments.


Subject(s)
Symporters/physiology , Thyroid Gland/metabolism , Animals , Breast Neoplasms/etiology , Breast Neoplasms/therapy , Genetic Therapy , Humans , Symporters/chemistry , Symporters/genetics , Symporters/metabolism , Thyroid Gland/chemistry , Thyroid Neoplasms/etiology , Thyroid Neoplasms/therapy
16.
Mol Endocrinol ; 16(8): 1893-902, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12145342

ABSTRACT

Active iodide uptake in the thyroid is mediated by the Na(+)/I(-) symporter (NIS), a key plasma membrane glycoprotein. Several NIS mutations have been shown to cause I(-) transport defect, a condition that, if untreated, can lead to congenital hypothyroidism and, ultimately, cretinism. The study of I(-) transport defect-causing NIS mutations provides valuable insights into the structure-function and mechanistic properties of NIS. Here we report the thorough analysis of the G395R NIS mutation. We observed no I(-) uptake activity at saturating or even supersaturating external I(-) concentrations in COS-7 cells transiently transfected with G395R NIS cDNA, even though we demonstrated normal expression of G395R NIS and proper targeting to the plasma membrane. Several amino acid substitutions at position 395 showed that the presence of an uncharged amino acid residue with a small side chain at position 395 is required for NIS function, suggesting that glycine 395 is located in a tightly packed region of NIS. Substitutions of large amino acid residues at position 395 resulted in lower V(max) without affecting K(m) values for I(-) and Na(+), suggesting that these residues hamper the Na(+)/I(-) coupling reaction.


Subject(s)
Symporters/chemistry , Symporters/metabolism , Amino Acid Substitution , Amino Acids, Neutral/chemistry , Animals , Base Sequence , COS Cells , DNA/genetics , Humans , Iodides/metabolism , Kinetics , Models, Molecular , Mutagenesis, Site-Directed , Protein Structure, Secondary , Rats , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sodium/metabolism , Symporters/genetics , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...