Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
Más filtros

Bases de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Hum Mutat ; 43(3): 420-433, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34979047

RESUMEN

Transporter-dependent steroid hormone uptake into target cells was demonstrated in genetically engineered mice and fruit flies. We hypothesized that mutations in such transporters may cause differences in sex development (DSD) in humans. Exome sequencing was performed in 16 genetically unsolved cases of 46,XY DSD selected from an anonymized collection of 708 lines of genital fibroblasts (GF) that were taken from individuals with incomplete virilization. Selection criteria were based on available biochemical characterization of GF compatible with reduced androgen uptake. Two unrelated individuals were identified with mutations in LDL receptor-related protein 2 (LRP2), a gene previously associated with partial sex steroid insensitivity in mice. Like Lrp2-/- mice, affected individuals had non-descended testes. Western blots on GF confirmed reduced LRP2 expression, and endocytosis of sex hormone-binding globulin was reduced. In three unrelated individuals, two with undescended testes, mutations in another endocytic receptor gene, limb development membrane protein 1 like (LMBR1L), were detected. Two of these individuals had mutations affecting the same codon. In a transfected cell model, mutated LMBR1L showed reduced cell surface expression. Our findings suggest that endocytic androgen uptake in complex with sex hormone-binding globulin is relevant in human. LMBR1L may play a similar role in androgen uptake.


Asunto(s)
Síndrome de Resistencia Androgénica , Síndrome de Resistencia Androgénica/genética , Andrógenos , Animales , Femenino , Genómica , Humanos , Masculino , Ratones , Mutación , Receptores Androgénicos/genética , Receptores Androgénicos/metabolismo , Receptores de Superficie Celular/genética , Globulina de Unión a Hormona Sexual/genética , Desarrollo Sexual/genética
2.
Int J Mol Sci ; 23(10)2022 May 11.
Artículo en Inglés | MEDLINE | ID: mdl-35628173

RESUMEN

The three isoenzymes of iodothyronine deiodinases (DIO1-3) are membrane-anchored homo-dimeric selenoproteins which share the thioredoxin-fold structure. Several questions regarding their catalytic mechanisms still remain open. Here, we addressed the roles of several cysteines which are conserved among deiodinase isoenzymes and asked whether they may contribute to dimerization and reduction of the oxidized enzyme with physiological reductants. We also asked whether amino acids previously identified in DIO3 play the same role in DIO1. Human DIO1 and 2 were recombinantly expressed in insect cells with selenocysteine replaced with cysteine (DIO1U126C) or in COS7 cells as selenoprotein. Enzyme activities were studied by radioactive deiodination assays with physiological reducing agents and recombinant proteins were characterized by mass spectrometry. Mutation of Cys124 in DIO1 prevented reduction by glutathione, while 20 mM dithiothreitol still regenerated the enzyme. Protein thiol reductants, thioredoxin and glutaredoxin, did not reduce DIO1U126C. Mass spectrometry demonstrated the formation of an intracellular disulfide between the side-chains of Cys124 and Cys(Sec)126. We conclude that the proximal Cys124 forms a selenenyl-sulfide with the catalytic Sec126 during catalysis, which is the substrate of the physiological reductant glutathione. Mutagenesis studies support the idea of a proton-relay pathway from solvent to substrate that is shared between DIO1 and DIO3.


Asunto(s)
Yoduro Peroxidasa , Animales , Células COS , Chlorocebus aethiops , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Humanos , Yoduro Peroxidasa/genética , Yoduro Peroxidasa/metabolismo , Isoenzimas , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/genética , Yodotironina Deyodinasa Tipo II
3.
J Biol Chem ; 294(39): 14185-14200, 2019 09 27.
Artículo en Inglés | MEDLINE | ID: mdl-31350336

RESUMEN

Recoding of UGA codons as selenocysteine (Sec) codons in selenoproteins depends on a selenocysteine insertion sequence (SECIS) in the 3'-UTR of mRNAs of eukaryotic selenoproteins. SECIS-binding protein 2 (SECISBP2) increases the efficiency of this process. Pathogenic mutations in SECISBP2 reduce selenoprotein expression and lead to phenotypes associated with the reduction of deiodinase activities and selenoprotein N expression in humans. Two functions have been ascribed to SECISBP2: binding of SECIS elements in selenoprotein mRNAs and facilitation of co-translational Sec insertion. To separately probe both functions, we established here two mouse models carrying two pathogenic missense mutations in Secisbp2 previously identified in patients. We found that the C696R substitution in the RNA-binding domain abrogates SECIS binding and does not support selenoprotein translation above the level of a complete Secisbp2 null mutation. The R543Q missense substitution located in the selenocysteine insertion domain resulted in residual activity and caused reduced selenoprotein translation, as demonstrated by ribosomal profiling to determine the impact on UGA recoding in individual selenoproteins. We found, however, that the R543Q variant is thermally unstable in vitro and completely degraded in the mouse liver in vivo, while being partially functional in the brain. The moderate impairment of selenoprotein expression in neurons led to astrogliosis and transcriptional induction of genes associated with immune responses. We conclude that differential SECISBP2 protein stability in individual cell types may dictate clinical phenotypes to a much greater extent than molecular interactions involving a mutated amino acid in SECISBP2.


Asunto(s)
Errores Innatos del Metabolismo/genética , Mutación Missense , Proteínas de Unión al ARN/metabolismo , Selenoproteínas/biosíntesis , Animales , Sitios de Unión , Encéfalo/metabolismo , Hígado/metabolismo , Ratones , Ratones Endogámicos C57BL , Fenotipo , Unión Proteica , Estabilidad Proteica , Proteolisis , Proteínas de Unión al ARN/química , Proteínas de Unión al ARN/genética , Ribosomas/metabolismo , Selenocisteína/metabolismo
4.
Cell Mol Neurobiol ; 40(5): 695-710, 2020 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-31808010

RESUMEN

Cathepsin K deficiency in male mice (Ctsk-/-) results in decreased numbers of hippocampal astrocytes and altered neuronal patterning as well as learning and memory deficits. Additionally, cathepsin K carries essential roles in the thyroid gland where it contributes to the liberation of thyroid hormones (TH). Because TH are essential for brain development, in particular for the cerebellum, we investigated whether cathepsin K's function in the thyroid is directly linked to the brain phenotype of Ctsk-/- mice. Serum levels of thyroid stimulating hormone, brain concentrations of free TH, and deiodinase 2 (Dio2) activity in brain parenchyma as well as cerebellar development were comparable in Ctsk-/- and WT animals, suggesting regular thyroid states and TH metabolism. Despite unaltered transcript levels, protein expression of two TH transporters was enhanced in specific brain regions in Ctsk-/- mice, suggesting altered TH supply to these regions. Thyrotropin releasing hormone (Trh) mRNA levels were enhanced threefold in the hippocampus of Ctsk-/- mice. In the striatum of Ctsk-/- mice the mRNA for Dio2 and hairless were approximately 1.3-fold enhanced, while mRNA levels for monocarboxylate transporter 8 and Trh were reduced to 60% and 40%, respectively, pointing to altered striatal physiology. We conclude that the role of cathepsin K in the thyroid gland is not directly associated with its function in the central nervous system (CNS) of mice. Future studies will show whether the brain region-specific alterations in Trh mRNA may eventually result in altered neuroprotection that could explain the neurobehavioral defects of Ctsk-/- mice.


Asunto(s)
Catepsina K/fisiología , Sistema Nervioso Central/enzimología , Glándula Tiroides/enzimología , Animales , Catepsina K/genética , Cerebelo/enzimología , Cerebelo/crecimiento & desarrollo , Masculino , Ratones , Ratones Noqueados , ARN Mensajero/análisis , Tirotropina/sangre , Tiroxina/sangre , Triyodotironina/sangre
5.
Cell Mol Life Sci ; 74(12): 2299-2318, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28132097

RESUMEN

Monocarboxylate transporter 8 (MCT8) mediates thyroid hormone (TH) transport across the plasma membrane in many cell types. In order to better understand its mechanism, we have generated three new MCT8 homology models based on sugar transporters XylE in the intracellular opened (PDB ID: 4aj4) and the extracellular partly occluded (PDB ID: 4gby) conformations as well as FucP (PDB ID: 3o7q) and GLUT3 (PDB ID: 4zwc) in the fully extracellular opened conformation. T3-docking studies from both sides revealed interactions with His192, His415, Arg445 and Asp498 as previously identified. Selected mutations revealed further transport-sensitive positions mainly at the discontinuous transmembrane helices TMH7 and 10. Lys418 is potentially involved in neutralising the charge of the TH substrate because it can be replaced by charged, but not by uncharged, amino acids. The side chain of Thr503 was hypothesised to stabilise a helix break at TMH10 that undergoes a prominent local shift during the transport cycle. A T503V mutation accordingly affected transport. The aromatic Tyr419, the polar Ser313 and Ser314 as well as the charged Glu422 and Glu423 lining the transport channel have been studied. Based on related sugar transporters, we suggest an alternating access mechanism for MCT8 involving a series of amino acid positions previously and newly identified as critical for transport.


Asunto(s)
Membrana Celular/metabolismo , Proteínas de Transporte de Membrana/metabolismo , Hormonas Tiroideas/metabolismo , Sustitución de Aminoácidos , Aminoácidos/metabolismo , Animales , Transporte Biológico , Cristalografía por Rayos X , Perros , Células de Riñón Canino Madin Darby , Proteínas de Transporte de Membrana/química , Simulación del Acoplamiento Molecular , Dominios Proteicos , Estabilidad Proteica , Transporte de Proteínas , Especificidad por Sustrato , Xenopus
6.
Proc Natl Acad Sci U S A ; 111(29): 10526-31, 2014 Jul 22.
Artículo en Inglés | MEDLINE | ID: mdl-25002520

RESUMEN

Local levels of active thyroid hormone (3,3',5-triiodothyronine) are controlled by the action of activating and inactivating iodothyronine deiodinase enzymes. Deiodinases are selenocysteine-dependent membrane proteins catalyzing the reductive elimination of iodide from iodothyronines through a poorly understood mechanism. We solved the crystal structure of the catalytic domain of mouse deiodinase 3 (Dio3), which reveals a close structural similarity to atypical 2-Cys peroxiredoxin(s) (Prx). The structure suggests a route for proton transfer to the substrate during deiodination and a Prx-related mechanism for subsequent recycling of the transiently oxidized enzyme. The proposed mechanism is supported by biochemical experiments and is consistent with the effects of mutations of conserved amino acids on Dio3 activity. Thioredoxin and glutaredoxin reduce the oxidized Dio3 at physiological concentrations, and dimerization appears to activate the enzyme by displacing an autoinhibitory loop from the iodothyronine binding site. Deiodinases apparently evolved from the ubiquitous Prx scaffold, and their structure and catalytic mechanism reconcile a plethora of partly conflicting data reported for these enzymes.


Asunto(s)
Biocatálisis , Yoduro Peroxidasa/química , Yoduro Peroxidasa/metabolismo , Peroxirredoxinas/metabolismo , Selenocisteína/metabolismo , Animales , Cristalografía por Rayos X , Enlace de Hidrógeno , Isomerismo , Ratones , Oxidación-Reducción , Multimerización de Proteína , Protones
7.
Artículo en Inglés | MEDLINE | ID: mdl-38469646

RESUMEN

CONTEXT: Monocarboxylate transporter 8 (MCT8) deficiency is a rare genetic disease that leads to severe global developmental delay. MCT8 facilitates thyroid hormone (TH) transport across the cell membrane, and the serum TH profile is characterized by high T3 and low T4 levels. Recent studies have shown that the chemical chaperone sodium phenylbutyrate (NaPB) restored mutant MCT8 function and increased TH content in patient-derived induced pluripotent stem cells, making it a potential treatment for MCT8 deficiency. OBJECTIVE: We aimed to assess the efficacy and safety of glycerol phenylbutyrate (GPB) in MCT8 deficiency. METHODS: We treated two monozygotic twins aged 14.5 years with MCT8 deficiency due to P321L mutation with escalating doses of GPB over 13 months. We recorded TH, Vital signs, anthropometric measurements and neurocognitive functions. Resting metabolic rate (RMR) was measured by indirect calorimetry. Serum metabolites of GPB were monitored as a safety measure. In-vitro effects of NaPB were evaluated in MDCK1 cells stably expressing the MCT8P321L mutation. The effects of GPB were compared to the effects of DITPA and TRIAC, thyromimetic medications that the patients received in the past. RESULTS: NaPB restored mutant MCT8 expression in MDCK1 cells and increased T3 transport into cells carrying the P321L mutation. GPB treatment reduced high T3 and increased low T4 levels. The patients showed a significant weight gain simultaneously with a reduction in RMR. Only minor neuro-cognitive improvement was observed, in hyperreflexia score and in cognitive functions. Serum metabolites did not exceed the toxic range but elevated liver transaminases were observed. CONCLUSIONS: In the first report of GPB treatment in MCT8 deficiency we found an improvement in TH profile and body-mass index, with minor neuro-developmental changes.

8.
Artículo en Inglés | MEDLINE | ID: mdl-38781537

RESUMEN

CONTEXT: Monocarboxylate transporter 8 (MCT8) deficiency is a rare X-chromosomal inherited disease leading to severe cognitive impairment, muscular hypotonia and symptoms of peripheral thyrotoxicosis. Experimental approaches aiming to functionally rescue mutant MCT8 activity by the chemical chaperone phenylbutyrate (PB) demonstrated promising effects in vitro for several MCT8 missense mutations. OBJECTIVE: The objective was to evaluate biochemical and clinical effects of PB in doses equivalent to those approved for the treatment of urea cycle disorders in a boy with MCT8 deficiency due to a novel MCT8 missense mutation c.703G > T (p.V235L). RESULTS: During a treatment period of 13 months, PB led to a significant decrease of elevated TSH and T3 serum concentrations, while fT4 increased. Weight z-score of the toddler remained remarkably stable during the treatment period. Neurodevelopmental assessments (BSID-III) revealed a slight increase of gross motor skills from developmental age 4 to 6 months. However, increasing liver enzyme serum activities and accumulation of phenylacetate (PAA) in urine led to treatment interruptions and dose alterations. In vitro analyses in MDCK1 cells confirmed the pathogenicity of MCT8 p.V235L. However, while PB increased expression of the mutant protein, it did not rescue T3 transport, suggesting a PB effect on thyroid function tests independent of restoring MCT8 activity. CONCLUSION: In a clinical attempt of PB treatment in MCT8 deficiency we observed a significant improvement of thyroid hormone function tests, tendencies towards body weight stabilization and slight neurodevelopmental improvement. Hepatotoxicity of PB may be a limiting factor in MCT8 deficiency and requires further investigation.

9.
Thyroid ; 2024 May 27.
Artículo en Inglés | MEDLINE | ID: mdl-38801167

RESUMEN

BACKGROUND: TRIAC (3,5,3'-triiodothyroacetic acid) is a T3-receptor agonist pharmacologically used in patients to mitigate T3 resistance. It is additionally explored to treat some symptoms of patients with inactivating mutations in the thyroid hormone (TH) transporter MCT8 (SLC16A2). MCT8 is expressed along the blood-brain-barrier, on neurons, astrocytes, and oligodendrocytes. Hence, pathogenic variants in MCT8 limit the access of TH into and their functions within the brain. TRIAC was shown to enter the brain independently of MCT8 and to modulate expression of TH-dependent genes. The aim of the study was to identify transporters that facilitate TRIAC uptake into cells. METHODS: We performed a whole-genome RNAi screen in HepG2 cells stably expressing a T3-receptor-dependent luciferase reporter gene. Validation of hits from the primary and confirmatory secondary screen involved a counter screen with siRNAs and compared the cellular response to TRIAC to the effect of T3, in order to exclude siRNAs targeting the gene expression machinery. MDCK1 cells were stably transfected with cDNA encoding C-terminally myc-tagged versions of the identified TRIAC-preferring transporters. Several individual clones were selected after immunocytochemical characterization for biochemical characterization of their 125I-TRIAC transport activities. RESULTS: We identified SLC22A9 and SLC29A2 as transporters mediating cellular uptake of TRIAC. SLC22A9 encodes the organic anion transporting polypeptide 7 (OAT7), a sodium-independent organic anion transporter expressed in the plasma membrane in brain, pituitary, liver and other organs. Competition with the SLC22A9/OAT7 substrate estrone-3-sulfate reduced 125I-TRIAC uptake. SLC29A2 encodes the equilibrative nucleoside transporter 2 (ENT2), which is ubiquitously expressed including pituitary and brain. Co-incubation with the SLC29A2/ENT2 inhibitor nitrobenzyl-6-thioinosine reduced 125I-TRIAC uptake. Moreover, ABCD1, an ATP-dependent peroxisomal pump, was identified as a 125I-TRIAC exporter in transfected MDCK1 cells. CONCLUSIONS: Knowledge of TRIAC transporter expression patterns, also during brain development, may thus in the future help to interpret observations on TRIAC effects as well as understand why TRIAC may not show a desirable effect on cells or organs not expressing appropriate transporters. The identification of ABCD1 highlights the sensitivity of our established screening assay, but it may not hold significant relevance for patients undergoing TRIAC treatment.

10.
J Mol Endocrinol ; 70(1)2023 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-36129170

RESUMEN

Proteolytic cleavage of thyroglobulin (Tg) for thyroid hormone (TH) liberation is followed by TH release from thyroid follicles into the circulation, enabled by TH transporters. The existence of a functional link between Tg-processing cathepsin proteases and TH transporters has been shown to be independent of the hypothalamus-pituitary-thyroid axis. Thus, lack of cathepsin K, combined with genetic defects in the TH transporters Mct8 and Mct10, that is the Ctsk-/-/Mct8-/y/Mct10-/- genotype, results in persistent Tg proteolysis due to autophagy induction. Because amino acid transport by L-type amino acid transporter 2 (Lat2) has been described to regulate autophagy, we asked whether Lat2 availability is affected in Ctsk-/-/Mct8-/y/Mct10-/- thyroid glands. Our data revealed that while mRNA amounts and subcellular localization of Lat2 remained unaltered in thyroid tissue of Ctsk-/-/Mct8-/y/Mct10-/- mice in comparison to WT controls, the Lat2 protein amounts were significantly reduced. These data suggest a direct link between Lat2 function and autophagy induction in Ctsk-/-/Mct8-/y/Mct10-/- mice. Indeed, thyroid tissue of Lat2-/- mice showed enhanced endo-lysosomal cathepsin activities, increased autophagosome formation, and enhanced autophagic flux. Collectively, these results suggest a mechanistic link between insufficient Lat2 protein function and autophagy induction in the thyroid gland of male mice.


Asunto(s)
Sistemas de Transporte de Aminoácidos , Autofagia , Glándula Tiroides , Animales , Masculino , Ratones , Autofagia/genética , Catepsinas , Genotipo
11.
Biochem J ; 439(2): 249-55, 2011 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-21726201

RESUMEN

LAT2 (system L amino acid transporter 2) is composed of the subunits Slc7a8/Lat2 and Slc3a2/4F2hc. This transporter is highly expressed along the basolateral membranes of absorptive epithelia in kidney and small intestine, but is also abundant in the brain. Lat2 is an energy-independent exchanger of neutral amino acids, and was shown to transport thyroid hormones. We report in the present paper that targeted inactivation of Slc7a8 leads to increased urinary loss of small neutral amino acids. Development and growth of Slc7a8(-/-) mice appears normal, suggesting functional compensation of neutral amino acid transport by alternative transporters in kidney, intestine and placenta. Movement co-ordination is slightly impaired in mutant mice, although cerebellar development and structure remained inconspicuous. Circulating thyroid hormones, thyrotropin and thyroid hormone-responsive genes remained unchanged in Slc7a8(-/-) mice, possibly because of functional compensation by the thyroid hormone transporter Mct8 (monocarboxylate transporter 8), which is co-expressed in many cell types. The reason for the mild neurological phenotype remains unresolved.


Asunto(s)
Errores Innatos del Metabolismo de los Aminoácidos/sangre , Sistema de Transporte de Aminoácidos y+/metabolismo , Cadenas Ligeras de la Proteína-1 Reguladora de Fusión/metabolismo , Transducción de Señal , Hormonas Tiroideas/sangre , Sistema de Transporte de Aminoácidos y+/genética , Animales , Secuencia de Bases , Western Blotting , Encéfalo/crecimiento & desarrollo , Cartilla de ADN , Cadenas Ligeras de la Proteína-1 Reguladora de Fusión/genética , Inmunohistoquímica , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Reacción en Cadena de la Polimerasa
12.
Thyroid ; 32(7): 860-870, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35357974

RESUMEN

Background: Monocarboxylate transporter 8 (MCT8) deficiency is a rare genetic disease leading to a severe developmental delay due to a lack of thyroid hormones (THs) during critical stages of human brain development. Some MCT8-deficient patients are not as severely affected as others. Previously, we hypothesized that these patients' mutations do not affect the functionality but destabilize the MCT8 protein, leading to a diminished number of functional MCT8 molecules at the cell surface. Methods: We have already demonstrated that the chemical chaperone sodium phenylbutyrate (NaPB) rescues the function of these mutants by stabilizing their protein expression in an overexpressing cell system. Here, we expanded our previous work and used iPSC (induced pluripotent stem cell)-derived brain microvascular endothelial-like cells (iBMECs) as a physiologically relevant cell model of human origin to test for NaPB responsiveness. The effects on mutant MCT8 expression and function were tested by Western blotting and radioactive uptake assays. Results: We found that NaPB rescues decreased mutant MCT8 expression and restores transport function in iBMECs carrying patient's mutation MCT8-P321L. Further, we identified MCT10 as an alternative TH transporter in iBMECs that contributes to triiodothyronine uptake, the biological active TH. Our results indicate an upregulation of MCT10 after NaPB treatment. In addition, we detected an increase in thyroxine (T4) uptake after NaPB treatment that was not mediated by rescued MCT8 but an unidentified T4 transporter. Conclusions: We demonstrate that NaPB is suitable to stabilize a pathogenic missense mutation in a human-derived cell model. Further, it activates TH transport independent of MCT8. Both options fuel future studies to investigate repurposing the Food and Drug Administration-approved drug NaPB in selected cases of MCT8 deficiency.


Asunto(s)
Transportadores de Ácidos Monocarboxílicos , Simportadores , Transporte Biológico , Encéfalo/metabolismo , Humanos , Discapacidad Intelectual Ligada al Cromosoma X , Transportadores de Ácidos Monocarboxílicos/genética , Transportadores de Ácidos Monocarboxílicos/metabolismo , Hipotonía Muscular , Atrofia Muscular , Fenilbutiratos , Simportadores/genética , Simportadores/metabolismo , Hormonas Tiroideas/metabolismo , Triyodotironina/metabolismo , Triyodotironina/farmacología
13.
Glia ; 59(3): 463-71, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21264952

RESUMEN

Cellular thyroid hormone uptake and efflux are mediated by transmembrane transport proteins. One of these, monocarboxylate transporter 8 (MCT8) is mutated in Allan-Herndon-Dudley syndrome, a severe mental retardation associated with abnormal thyroid hormone constellations. Since mice deficient in Mct8 exhibit a milder neurological phenotype than patients, we hypothesized that alternative thyroid hormone transporters may compensate in murine brain cells for the lack of Mct8. Using qPCR, Western Blot, and immunocytochemistry, we investigated the expression of three different thyroid hormone transporters, i.e., Mct8 and L-type amino acid transporters Lat1 and Lat2, in mouse brain. All three thyroid hormone transporters are expressed from corticogenesis and peak around birth. Primary cultures of neurons and astrocytes express Mct8, Lat1, and Lat2. Microglia specifically expresses Mct10 and Slco4a1 in addition to high levels of Lat2 mRNA and protein. As in vivo, a brain microvascular endothelial cell line expressed Mct8 and Lat1. 158N, an oligodendroglial cell line expressed Mct8 protein, consistent with delayed myelination in MCT8-deficient patients. Functional T(3)- and T(4)-transport assays into primary astrocytes showed K(M) values of 4.2 and 3.7 µM for T(3) and T(4). Pharmacological inhibition of L-type amino acid transporters by BCH and genetic inactivation of Lat2 reduced astrocytic T(3) uptake to the same extent. BSP, a broad spectrum inhibitor, including Mct8, reduced T(3) uptake further suggesting the cooperative activity of several T(3) transporters in astrocytes.


Asunto(s)
Encéfalo/crecimiento & desarrollo , Encéfalo/metabolismo , Regulación del Desarrollo de la Expresión Génica/fisiología , Proteínas de Transporte de Membrana/genética , Neuronas/metabolismo , Tiroxina/metabolismo , Triyodotironina/metabolismo , Sistema de Transporte de Aminoácidos y+/biosíntesis , Sistema de Transporte de Aminoácidos y+/metabolismo , Sistema de Transporte de Aminoácidos y+L , Animales , Animales Recién Nacidos , Astrocitos/citología , Astrocitos/metabolismo , Encéfalo/citología , Línea Celular , Células Cultivadas , Técnicas de Cocultivo , Cadenas Ligeras de la Proteína-1 Reguladora de Fusión/biosíntesis , Cadenas Ligeras de la Proteína-1 Reguladora de Fusión/metabolismo , Células HEK293 , Humanos , Proteínas de Transporte de Membrana/deficiencia , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Transportadores de Ácidos Monocarboxílicos , Neuronas/citología , Transporte de Proteínas/fisiología , Simportadores
14.
Rev Neurosci ; 21(3): 173-86, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20879691

RESUMEN

Thyroid hormones are essential for brain development. The active thyroid hormone, T3, binds to several products of two genes, the nuclear thyroid hormone receptors alpha and beta, and thus regulates gene expression. Mutations in a thyroid hormone transmembrane transport protein, monocarboxylate transporter 8 (MCT8), underlie one of the first described X-linked mental retardation syndromes, the Allan-Herndon-Dudley syndrome. This discovery sparked great interest in the process of thyroid hormone transmembrane transport. Iodothyronines are charged amino acid derivatives and require protein facilitators to cross cellular membranes. Thyroid hormones are translocated across lipid bilayers by several members of the major facilitator superfamily, including monocarboxylate transporters, amino acid transporters, and organic anion transporting polypeptides. Although until recently few researchers considered thyroid hormone transporters an important object of study, there is now a large number of candidate transporters to be reckoned with in the brain. Moreover, to finally cross the neuronal plasma membrane, any iodothyronine molecule on its way toward a neuronal nucleus has to cross consecutively the lumenal and ablumenal membranes of the capillary endothelium, enter astrocytic foot processes, and leave the astrocyte through the plasma membrane. Moreover, microglia, oligodendrocytes, and precursor and stem cells are thyroid hormone responsive and likely express thyroid hormone transporters. Hence, the many roles played by thyroid hormones in the development, function, and regeneration of the nervous system are dependent on the spatiotemporal expression of several transmembrane transport proteins.


Asunto(s)
Encéfalo/metabolismo , Transportadores de Ácidos Monocarboxílicos/metabolismo , Hormonas Tiroideas/metabolismo , Animales , Transporte Biológico/fisiología , Encéfalo/patología , Humanos , Discapacidad Intelectual Ligada al Cromosoma X/genética , Transportadores de Ácidos Monocarboxílicos/química , Transportadores de Ácidos Monocarboxílicos/genética , Mutación/genética , Simportadores , Hormonas Tiroideas/genética
15.
Eur Thyroid J ; 9(5): 269-280, 2020 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-33088796

RESUMEN

INTRODUCTION: The monocarboxylate transporter 8 (MCT8; SLC16A2) is a specific transporter for thyroid hormones. MCT8 deficiency, formerly known as the Allan-Herndon-Dudley syndrome, is a rare genetic disease that leads to neurological impairments and muscle weakness. Current experimental treatment options rely on thyromimetic agonists that do not depend on MCT8 for cellular uptake. Another approach comes from studies with the chemical chaperone sodium phenylbutyrate (NaPB), which was able to stabilize MCT8 mutants having protein folding defects in vitro. In addition, NaPB is known as a compound that assists with plasma membrane translocation. OBJECTIVE: The pathogenic MCT8L291R leads to the same severe neurological impairments found for other MCT8-deficient patients but, unexpectedly, lacks alterations in plasma 3,3',5-triiodothyronine (T3) levels. Here we tried to unravel the underlying mechanism of MCT8 deficiency and tested whether the pathogenic MCT8L291R mutant responds to NaPB treatment. Therefore, we overexpressed the mutant in Madin-Darby canine kidney cells in the human choriocarcinoma cell line JEG1 and in COS7 cells of African green monkey origin. RESULTS: In our recent study we describe that the MCT8L291R mutation most likely leads to a translocation defect. The pathogenic mutant is not located at the plasma membrane, but shows overlapping expression with a marker protein of the lysosome. Mutation of the corresponding amino acid in murine Mct8 (Mct8L223R) displays a similar effect on cell surface expression and transport function as seen before for MCT8L291R. NaPB was able to correct the translocation defect of MCT8L291R/Mct8L223R and restored protein function by increasing T3 transport activity. Furthermore, we detected enhanced mRNA levels of wild-type and mutant MCT8/Mct8 after NaPB treatment. The increase in mRNA levels could be an explanation for the positive effect on protein expression and function detected for wild-type MCT8. CONCLUSION: NaPB is not only suitable for the treatment of mutations leading to misfolding and protein degradation, but also for a mutant wrongly sorted inside a cell which is otherwise functional.

16.
Front Cell Dev Biol ; 8: 567, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32850778

RESUMEN

Previous studies in developing Xenopus and zebrafish reported that the phosphate transporter slc20a1a is expressed in pronephric kidneys. The recent identification of SLC20A1 as a monoallelic candidate gene for cloacal exstrophy further suggests its involvement in the urinary tract and urorectal development. However, little is known of the functional role of SLC20A1 in urinary tract development. Here, we investigated this using morpholino oligonucleotide knockdown of the zebrafish ortholog slc20a1a. This caused kidney cysts and malformations of the cloaca. Moreover, in morphants we demonstrated dysfunctional voiding and hindgut opening defects mimicking imperforate anus in human cloacal exstrophy. Furthermore, we performed immunohistochemistry of an unaffected 6-week-old human embryo and detected SLC20A1 in the urinary tract and the abdominal midline, structures implicated in the pathogenesis of cloacal exstrophy. Additionally, we resequenced SLC20A1 in 690 individuals with bladder exstrophy-epispadias complex (BEEC) including 84 individuals with cloacal exstrophy. We identified two additional monoallelic de novo variants. One was identified in a case-parent trio with classic bladder exstrophy, and one additional novel de novo variant was detected in an affected mother who transmitted this variant to her affected son. To study the potential cellular impact of SLC20A1 variants, we expressed them in HEK293 cells. Here, phosphate transport was not compromised, suggesting that it is not a disease mechanism. However, there was a tendency for lower levels of cleaved caspase-3, perhaps implicating apoptosis pathways in the disease. Our results suggest SLC20A1 is involved in urinary tract and urorectal development and implicate SLC20A1 as a disease-gene for BEEC.

17.
Endocrinology ; 160(6): 1536-1546, 2019 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-31127274

RESUMEN

Allan-Herndon-Dudley syndrome (AHDS) is a severe genetic disease caused by mutations in the monocarboxylate transporter 8 (MCT8) gene. MCT8 mediates transport of thyroid hormones in and out of cells, which is thought to play a pivotal role for embryonic and postnatal development of the human brain. Disconcertingly, MCT8R271H leads to a severe form of AHDS but shows residual transport activity when expressed in several types of cultured cells. Here we try to determine the mechanism behind the transport function of MCT8R271H found in overexpressing cell systems. Mutations of Arg271 were introduced into human MCT8 and stably transfected into Madin-Darby canine kidney cells and the human-derived cell line JEG1. Radioactive thyroid hormone-uptake experiments were performed to analyze the pH-dependent effect of the mutation on transport activity. Arg271His transports thyroid hormones in and out of cells in a pH-dependent manner. Its transport activity increases below pH 7.3 and is clearly diminished at physiological pH. The Michaelis constant of the mutant is unaltered, whereas the maximum velocity is reduced. The expression of Arg271His in JEG1 cells leads to an almost nonfunctional transporter at physiological pH replicating the human phenotype for this mutant in vitro and demonstrates, again, that mutant MCT8 activity depends on cellular background. The protonation of His271 at acidic pH restores activity of the mutant protein, which is not active in its deprotonated form at physiological pH. Thus, experimental parameters must be controlled carefully when modeling MCT8 deficiency in cells.


Asunto(s)
Transportadores de Ácidos Monocarboxílicos/genética , Mutación , Fenotipo , Hormonas Tiroideas/metabolismo , Animales , Línea Celular , Perros , Humanos , Células de Riñón Canino Madin Darby , Discapacidad Intelectual Ligada al Cromosoma X/genética , Discapacidad Intelectual Ligada al Cromosoma X/metabolismo , Transportadores de Ácidos Monocarboxílicos/metabolismo , Hipotonía Muscular/genética , Hipotonía Muscular/metabolismo , Atrofia Muscular/genética , Atrofia Muscular/metabolismo , Transporte de Proteínas , Simportadores
18.
Vitam Horm ; 106: 19-44, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29407435

RESUMEN

Thyroid hormones orchestrate developmental processes and are among the most important regulators of energy metabolism. Thyroid hormone actions are mostly, but not exclusively, mediated by nuclear hormone receptors. As amino acid derivatives, thyroid hormones need plasma membrane transporters in order to reach their nuclear receptors. Several transporters from different gene families mediate thyroid hormone uptake into cells. Monocarboxylate transporter 8 is a specific thyroid hormone transporter found mutated in patients with severe psychomotor retardation and strangely abnormal thyroid hormone constellations. These patients display a syndrome in which some organs are exposed to increased thyroid hormone signaling, while other organs are lacking thyroid hormone signaling due to complete lack of thyroid hormone uptake. Investigations in many organ systems using mouse models of thyroid hormone transmembrane transporter deficiency have helped complete our picture of thyroid hormone metabolism and action in the body during development and under different physiological conditions. Incorporating the concept of thyroid hormone transmembrane transport has helped understand previously enigmatic drug interactions and may explain how the hormonal set points in the hypothalamus-pituitary-thyroid axis are established.


Asunto(s)
Transporte de Proteínas/fisiología , Hormonas Tiroideas/metabolismo , Animales , Regulación de la Expresión Génica , Humanos , Proteínas de Transporte de Membrana/fisiología , Unión Proteica
19.
Endocrinology ; 158(3): 678-691, 2017 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-27977298

RESUMEN

Mutations in the thyroid hormone transporter monocarboxylate transporter 8 (MCT8) prevent appropriate entry of thyroid hormones into brain cells during development and cause severe mental retardation in affected patients. The current treatment options are thyromimetic compounds that enter the brain independently of MCT8. Some MCT8-deficient patients (e.g., those carrying MCT8delF501) will not be as severely affected as most others. We have shown that the MCT8delF501 protein has decreased protein stability but important residual function once it reaches the plasma membrane. We were able to rescue protein expression and the function of MCT8delF501 in a Madin-Darby canine kidney cell model by application of the chemical chaperone sodium phenylbutyrate (NaPB), a drug that has been used to treat patients with cystic fibrosis and urea cycle defects for extended periods of time. In the present study, we have extended our previous study and report on the NaPB-dependent rescue of a series of other pathogenic MCT8 mutants associated with milder patient phenotypes. We show that NaPB can functionally rescue the expression and activities of Ser194Phe, Ser290Phe, Leu434Trp, Arg445Cys, Leu492Pro, and Leu568Pro mutations in MCT8 in a dose-dependent manner. The soy isoflavone genistein, a dietary supplement, which was effective in MCT8delF501, was also effective in increasing the expression and transport of these MCT8 mutants; however, the effect size differed among mutants. Kinetic analyses revealed that the Michaelis constants of the mutants toward the primary substrate 3,3',5-triiodothyronine were not much different from the wild-type value, suggesting that these mutants are not impaired in their interaction with substrate but rather destabilized by the mutation and degraded.


Asunto(s)
Antineoplásicos/uso terapéutico , Discapacidad Intelectual Ligada al Cromosoma X/genética , Transportadores de Ácidos Monocarboxílicos/genética , Hipotonía Muscular/genética , Atrofia Muscular/genética , Fenilbutiratos/uso terapéutico , Animales , Chlorocebus aethiops , Perros , Evaluación Preclínica de Medicamentos , Genisteína , Humanos , Células de Riñón Canino Madin Darby , Discapacidad Intelectual Ligada al Cromosoma X/tratamiento farmacológico , Chaperonas Moleculares/uso terapéutico , Transportadores de Ácidos Monocarboxílicos/metabolismo , Hipotonía Muscular/tratamiento farmacológico , Atrofia Muscular/tratamiento farmacológico , Mutación , Fenotipo , Simportadores
20.
Mol Endocrinol ; 30(7): 796-808, 2016 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-27244477

RESUMEN

Monocarboxylate transporters (MCTs) belong to the SLC16 family within the major facilitator superfamily of transmembrane transporters. MCT8 is a thyroid hormone transporter mutated in the Allan-Herndon-Dudley syndrome, a severe psychomotor retardation syndrome. MCT10 is closely related to MCT8 and is known as T-type amino acid transporter. Both transporters mediate T3 transport, but although MCT8 also transports rT3 and T4, these compounds are not efficiently transported by MCT10, which, in contrast, transports aromatic amino acids. Based on the 58% amino acid identity within the transmembrane regions among MCT8 and MCT10, we reasoned that substrate specificity may be primarily determined by a small number of amino acid differences between MCT8 and MCT10 along the substrate translocation channel. Inspecting the homology model of MCT8 and a structure-guided alignment between both proteins, we selected 8 amino acid positions and prepared chimeric MCT10 proteins with selected amino acids changed to the corresponding amino acids in MCT8. The MCT10 mutant harboring 8 amino acid substitutions was stably expressed in Madin-Darby canine kidney 1 cells and found to exhibit T4 transport activity. We then successively reduced the number of amino acid substitutions and eventually identified a minimal set of 2-3 amino acid exchanges which were sufficient to allow T4 transport. The resulting MCT10 chimeras exhibited KM values for T4 similar to MCT8 but transported T4 at a slower rate. The acquisition of T4 transport by MCT10 was associated with complete loss of the capacity to transport Phe, when Tyr184 was mutated to Phe.


Asunto(s)
Sistemas de Transporte de Aminoácidos Neutros/química , Sistemas de Transporte de Aminoácidos Neutros/metabolismo , Aminoácidos/metabolismo , Transportadores de Ácidos Monocarboxílicos/química , Transportadores de Ácidos Monocarboxílicos/metabolismo , Sistemas de Transporte de Aminoácidos Neutros/genética , Aminoácidos/química , Aminoácidos/genética , Animales , Biotinilación , Western Blotting , Línea Celular , Cromatografía Liquida , Perros , Transportadores de Ácidos Monocarboxílicos/genética , Mutagénesis Sitio-Dirigida , Especificidad por Sustrato , Espectrometría de Masas en Tándem , Hormonas Tiroideas/metabolismo , Triyodotironina/metabolismo , Xenopus
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA