Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Development ; 150(10)2023 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-37191061

RESUMEN

Thyroid tissue, the site of de novo thyroid hormone biosynthesis, is derived from ventral pharyngeal endoderm and defects in morphogenesis are a predominant cause of congenital thyroid diseases. The first molecularly recognizable step of thyroid development is the specification of thyroid precursors in anterior foregut endoderm. Recent studies have identified crucial roles of FGF and BMP signaling in thyroid specification, but the interplay between signaling cues and thyroid transcription factors remained elusive. By analyzing Pax2a and Nkx2.4b expression dynamics in relation to endodermal FGF and BMP signaling activities in zebrafish embryos, we identified a Pax2a-expressing thyroid progenitor population that shows enhanced FGF signaling but lacks Nkx2.4b expression and BMP signaling. Concurrent with upregulated BMP signaling, a subpopulation of these progenitors subsequently differentiates into lineage-committed thyroid precursors co-expressing Pax2a and Nkx2.4b. Timed manipulation of FGF/BMP activities suggests a model in which FGF signaling primarily regulates Pax2a expression, whereas BMP signaling regulates both Pax2a and Nkx2.4b expression. Our observation of similar expression dynamics of Pax8 and Nkx2-1 in mouse embryos suggests that this refined model of thyroid cell specification is evolutionarily conserved in mammals.


Asunto(s)
Factores de Crecimiento de Fibroblastos , Pez Cebra , Animales , Ratones , Pez Cebra/genética , Pez Cebra/metabolismo , Factores de Crecimiento de Fibroblastos/metabolismo , Glándula Tiroides , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo , Transducción de Señal/genética , Regulación del Desarrollo de la Expresión Génica , Endodermo/metabolismo , Mamíferos/metabolismo
2.
Hum Mol Genet ; 31(23): 3967-3974, 2022 11 28.
Artículo en Inglés | MEDLINE | ID: mdl-35535691

RESUMEN

Congenital hypothyroidism due to thyroid dysgenesis (TD), presented as thyroid aplasia, hypoplasia or ectopia, is one of the most prevalent rare diseases with an isolated organ malformation. The pathogenesis of TD is largely unknown, although a genetic predisposition has been suggested. We performed a genome-wide association study (GWAS) with 142 Japanese TD cases and 8380 controls and found a significant locus at 2q33.3 (top single nucleotide polymorphism, rs9789446: P = 4.4 × 10-12), which was replicated in a German patient cohort (P = 0.0056). A subgroup analysis showed that rs9789446 confers a risk for thyroid aplasia (per allele odds ratio = 3.17) and ectopia (3.12) but not for hypoplasia. Comprehensive epigenomic characterization of the 72-kb disease-associated region revealed that it was enriched for active enhancer signatures in human thyroid. Analysis of chromosome conformation capture data showed long-range chromatin interactions of this region with promoters of two genes, FZD5 and CCNYL1, mediating Wnt signaling. Moreover, rs9789446 was found to be a thyroid-specific quantitative trait locus, adding further evidence for a cis-regulatory function of this region in thyroid tissue. Specifically, because the risk rs9789446 allele is associated with increased thyroidal expression of FDZ5 and CCNYL1 and given the recent demonstration of perturbed early thyroid development following overactivation of Wnt signaling in zebrafish embryos, an enhanced Wnt signaling in risk allele carriers provides a biologically plausible TD mechanism. In conclusion, our work found the first risk locus for TD, exemplifying that in rare diseases with relatively low biological complexity, GWAS may provide mechanistic insights even with a small sample size.


Asunto(s)
Estudio de Asociación del Genoma Completo , Disgenesias Tiroideas , Animales , Humanos , Pez Cebra/genética , Vía de Señalización Wnt/genética , Enfermedades Raras , Disgenesias Tiroideas/genética , Predisposición Genética a la Enfermedad
3.
Proc Natl Acad Sci U S A ; 117(47): 29684-29690, 2020 11 24.
Artículo en Inglés | MEDLINE | ID: mdl-33184177

RESUMEN

Battling metastasis through inhibition of cell motility is considered a promising approach to support cancer therapies. In this context, Ena/VASP-depending signaling pathways, in particular interactions with their EVH1 domains, are promising targets for pharmaceutical intervention. However, protein-protein interactions involving proline-rich segments are notoriously difficult to address by small molecules. Hence, structure-based design efforts in combination with the chemical synthesis of additional molecular entities are required. Building on a previously developed nonpeptidic micromolar inhibitor, we determined 22 crystal structures of ENAH EVH1 in complex with inhibitors and rationally extended our library of conformationally defined proline-derived modules (ProMs) to succeed in developing a nanomolar inhibitor ([Formula: see text] Da). In contrast to the previous inhibitor, the optimized compounds reduced extravasation of invasive breast cancer cells in a zebrafish model. This study represents an example of successful, structure-guided development of low molecular weight inhibitors specifically and selectively addressing a proline-rich sequence-recognizing domain that is characterized by a shallow epitope lacking defined binding pockets. The evolved high-affinity inhibitor may now serve as a tool in validating the basic therapeutic concept, i.e., the suppression of cancer metastasis by inhibiting a crucial protein-protein interaction involved in actin filament processing and cell migration.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Moléculas de Adhesión Celular/metabolismo , Proteínas de Unión al ADN/metabolismo , Proteínas de Microfilamentos/metabolismo , Fosfoproteínas/metabolismo , Dominios y Motivos de Interacción de Proteínas/efectos de los fármacos , Bibliotecas de Moléculas Pequeñas/farmacología , Animales , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Femenino , Humanos , Células Jurkat , Prolina/metabolismo , Unión Proteica/efectos de los fármacos , Pez Cebra
4.
Nature ; 491(7422): 66-71, 2012 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-23051751

RESUMEN

The primary function of the thyroid gland is to metabolize iodide by synthesizing thyroid hormones, which are critical regulators of growth, development and metabolism in almost all tissues. So far, research on thyroid morphogenesis has been missing an efficient stem-cell model system that allows for the in vitro recapitulation of the molecular and morphogenic events regulating thyroid follicular-cell differentiation and subsequent assembly into functional thyroid follicles. Here we report that a transient overexpression of the transcription factors NKX2-1 and PAX8 is sufficient to direct mouse embryonic stem-cell differentiation into thyroid follicular cells that organize into three-dimensional follicular structures when treated with thyrotropin. These in vitro-derived follicles showed appreciable iodide organification activity. Importantly, when grafted in vivo into athyroid mice, these follicles rescued thyroid hormone plasma levels and promoted subsequent symptomatic recovery. Thus, mouse embryonic stem cells can be induced to differentiate into thyroid follicular cells in vitro and generate functional thyroid tissue.


Asunto(s)
Diferenciación Celular , Células Madre Embrionarias/citología , Glándula Tiroides/citología , Glándula Tiroides/fisiología , Animales , Modelos Animales de Enfermedad , Células Madre Embrionarias/metabolismo , Femenino , Humanos , Hipotiroidismo/patología , Hipotiroidismo/cirugía , Hipotiroidismo/terapia , Ratones , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Factor de Transcripción PAX8 , Factores de Transcripción Paired Box/genética , Factores de Transcripción Paired Box/metabolismo , Glándula Tiroides/anatomía & histología , Glándula Tiroides/efectos de los fármacos , Glándula Tiroides/trasplante , Factor Nuclear Tiroideo 1 , Tirotropina/sangre , Tirotropina/farmacología , Tiroxina/sangre , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
5.
Proc Natl Acad Sci U S A ; 112(16): 5011-6, 2015 Apr 21.
Artículo en Inglés | MEDLINE | ID: mdl-25848013

RESUMEN

Small-molecule competitors of protein-protein interactions are urgently needed for functional analysis of large-scale genomics and proteomics data. Particularly abundant, yet so far undruggable, targets include domains specialized in recognizing proline-rich segments, including Src-homology 3 (SH3), WW, GYF, and Drosophila enabled (Ena)/vasodilator-stimulated phosphoprotein (VASP) homology 1 (EVH1) domains. Here, we present a modular strategy to obtain an extendable toolkit of chemical fragments (ProMs) designed to replace pairs of conserved prolines in recognition motifs. As proof-of-principle, we developed a small, selective, peptidomimetic inhibitor of Ena/VASP EVH1 domain interactions. Highly invasive MDA MB 231 breast-cancer cells treated with this ligand showed displacement of VASP from focal adhesions, as well as from the front of lamellipodia, and strongly reduced cell invasion. General applicability of our strategy is illustrated by the design of an ErbB4-derived ligand containing two ProM-1 fragments, targeting the yes-associated protein 1 (YAP1)-WW domain with a fivefold higher affinity.


Asunto(s)
Dominios Proteicos Ricos en Prolina , Mapeo de Interacción de Proteínas , Animales , Moléculas de Adhesión Celular/química , Línea Celular Tumoral , Permeabilidad de la Membrana Celular , Cristalografía por Rayos X , Drosophila melanogaster/metabolismo , Esterificación , Técnica del Anticuerpo Fluorescente , Humanos , Cinética , Ligandos , Proteínas de Microfilamentos/química , Modelos Moleculares , Peso Molecular , Péptidos/química , Fosfoproteínas/química , Unión Proteica , Estructura Terciaria de Proteína , Seudópodos , Fibras de Estrés/metabolismo , Zixina/química
6.
Chemistry ; 21(23): 8464-70, 2015 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-25906737

RESUMEN

With the aim of developing polyproline type II helix (PPII) secondary-structure mimetics for the modulation of prolin-rich-mediated protein-protein interactions, the novel diproline mimetic ProM-2 was designed by bridging the two pyrrolidine rings of a diproline (Pro-Pro) unit through a Z-vinylidene moiety. This scaffold, which closely resembles a section of a PPII helix, was then stereoselectively synthesized by exploiting a ruthenium-catalyzed ring-closing metathesis (RCM) as a late key step. The required vinylproline building blocks, that is, (R)-N-Boc-2-vinylproline (Boc=tert-butyloxycarbonyl) and (S,S)-5-vinylproline-tert-butyl ester, were prepared on a gram scale as pure stereoisomers. The difficult peptide coupling of the sterically demanding building blocks was achieved in good yield and without epimerization by using 2-(1H-7-azabenzotriazol-1-yl)-1,1,3,3-tetramethyluronium hexafluorophosphate (HATU)/N,N-diisopropylethylamine (DIPEA). The RCM proceeded smoothly in the presence of the Grubbs II catalyst. Stereostructural assignments for several intermediates were secured by X-ray crystallography. As a proof of concept, it was shown that certain peptides containing ProM-2 exhibited improved (canonical) binding towards the Ena/VASP homology 1 (EVH1) domain as a relevant protein interaction target.


Asunto(s)
Péptidos/química , Proteínas/química , Dipéptidos/química , Peptidomiméticos , Conformación Proteica , Estereoisomerismo
7.
Sci Rep ; 14(1): 9355, 2024 04 23.
Artículo en Inglés | MEDLINE | ID: mdl-38654093

RESUMEN

Thyroid hormones (TH) play critical roles during nervous system development and patients carrying coding variants of MCT8 (monocarboxylate transporter 8) or THRA (thyroid hormone receptor alpha) present a spectrum of neurological phenotypes resulting from perturbed local TH action during early brain development. Recently, human cerebral organoids (hCOs) emerged as powerful in vitro tools for disease modelling recapitulating key aspects of early human cortex development. To begin exploring prospects of this model for thyroid research, we performed a detailed characterization of the spatiotemporal expression of MCT8 and THRA in developing hCOs. Immunostaining showed MCT8 membrane expression in neuronal progenitor cell types including early neuroepithelial cells, radial glia cells (RGCs), intermediate progenitors and outer RGCs. In addition, we detected robust MCT8 protein expression in deep layer and upper layer neurons. Spatiotemporal SLC16A2 mRNA expression, detected by fluorescent in situ hybridization (FISH), was highly concordant with MCT8 protein expression across cortical cell layers. FISH detected THRA mRNA expression already in neuroepithelium before the onset of neurogenesis. THRA mRNA expression remained low in the ventricular zone, increased in the subventricular zone whereas strong THRA expression was observed in excitatory neurons. In combination with a robust up-regulation of known T3 response genes following T3 treatment, these observations show that hCOs provide a promising and experimentally tractable model to probe local TH action during human cortical neurogenesis and eventually to model the consequences of impaired TH function for early cortex development.


Asunto(s)
Corteza Cerebral , Transportadores de Ácidos Monocarboxílicos , Neurogénesis , Organoides , ARN Mensajero , Simportadores , Receptores alfa de Hormona Tiroidea , Femenino , Humanos , Embarazo , Corteza Cerebral/embriología , Corteza Cerebral/metabolismo , Regulación del Desarrollo de la Expresión Génica , Transportadores de Ácidos Monocarboxílicos/genética , Transportadores de Ácidos Monocarboxílicos/metabolismo , Células-Madre Neurales/metabolismo , Células-Madre Neurales/citología , Neurogénesis/genética , Neuronas/metabolismo , Organoides/metabolismo , Primer Trimestre del Embarazo/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Simportadores/genética , Simportadores/metabolismo , Receptores alfa de Hormona Tiroidea/genética , Receptores alfa de Hormona Tiroidea/metabolismo , Hormonas Tiroideas/metabolismo , Hormonas Tiroideas/genética
8.
Thyroid ; 34(6): 796-805, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38526409

RESUMEN

Background: Thyroid hormones regulate cardiac functions mainly through direct actions in the heart and by binding to the thyroid hormone receptor (TR) isoforms α1 and ß. While the role of the most abundantly expressed isoform, TRα1, is widely studied and well characterized, the role of TRß in regulating heart functions is still poorly understood, primarily due to the accompanying elevation of circulating thyroid hormone in TRß knockout mice (TRß-KO). However, their hyperthyroidism is ameliorated at thermoneutrality, which allows studying the role of TRß without this confounding factor. Methods: Here, we noninvasively monitored heart rate in TRß-KO mice over several days using radiotelemetry at different housing temperatures (22°C and 30°C) and upon 3,3',5-triiodothyronine (T3) administration in comparison to wild-type animals. Results: TRß-KO mice displayed normal average heart rate at both 22°C and 30°C with only minor changes in heart rate frequency distribution, which was confirmed by independent electrocardiogram recordings in freely-moving conscious mice. Parasympathetic nerve activity was, however, impaired in TRß-KO mice at 22°C, and only partly rescued at 30°C. As expected, oral treatment with pharmacological doses of T3 at 30°C led to tachycardia in wild-types, accompanied by broader heart rate frequency distribution and increased heart weight. The TRß-KO mice, in contrast, showed blunted tachycardia, as well as resistance to changes in heart rate frequency distribution and heart weight. At the molecular level, these observations were paralleled by a blunted cardiac mRNA induction of several important genes, including the pacemaker channels Hcn2 and Hcn4, as well as Kcna7. Conclusions: The phenotyping of TRß-KO mice conducted at thermoneutrality allows novel insights on the role of TRß in cardiac functions in the absence of the usual confounding hyperthyroidism. Even though TRß is expressed at lower levels than TRα1 in the heart, our findings demonstrate an important role for this isoform in the cardiac response to thyroid hormones.


Asunto(s)
Cardiomegalia , Frecuencia Cardíaca , Ratones Noqueados , Taquicardia , Receptores beta de Hormona Tiroidea , Triyodotironina , Animales , Receptores beta de Hormona Tiroidea/genética , Receptores beta de Hormona Tiroidea/metabolismo , Taquicardia/fisiopatología , Taquicardia/metabolismo , Ratones , Cardiomegalia/metabolismo , Cardiomegalia/fisiopatología , Cardiomegalia/genética , Triyodotironina/sangre , Masculino , Hormonas Tiroideas/metabolismo , Sistema Nervioso Parasimpático/fisiopatología , Temperatura , Electrocardiografía
9.
Dev Biol ; 372(2): 203-16, 2012 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-23022354

RESUMEN

Among the various organs derived from foregut endoderm, the thyroid gland is unique in that major morphogenic events such as budding from foregut endoderm, descent into subpharyngeal mesenchyme and growth expansion occur in close proximity to cardiovascular tissues. To date, research on thyroid organogenesis was missing one vital tool-a transgenic model that allows to track the dynamic changes in thyroid size, shape and location relative to adjacent cardiovascular tissues in live embryos. In this study, we generated a novel transgenic zebrafish line, tg(tg:mCherry), in which robust and thyroid-specific expression of a membrane version of mCherry enables live imaging of thyroid development in embryos from budding stage throughout formation of functional thyroid follicles. By using various double transgenic models in which EGFP expression additionally labels cardiovascular structures, a high coordination was revealed between thyroid organogenesis and cardiovascular development. Early thyroid development was found to proceed in intimate contact with the distal ventricular myocardium and live imaging confirmed that thyroid budding from the pharyngeal floor is tightly coordinated with the descent of the heart. Four-dimensional imaging of live embryos by selective plane illumination microscopy and 3D-reconstruction of confocal images of stained embryos yielded novel insights into the role of specific pharyngeal vessels, such as the hypobranchial artery (HA), in guiding late thyroid expansion along the pharyngeal midline. An important role of the HA was corroborated by the detailed examination of thyroid development in various zebrafish models showing defective cardiovascular development. In combination, our results from live imaging as well es from 3D-reconstruction of thyroid development in tg(tg:mCherry) embryos provided a first dynamic view of late thyroid organogenesis in zebrafish-a critical resource for the design of future studies addressing the molecular mechanisms of these thyroid-vasculature interactions.


Asunto(s)
Sistema Cardiovascular/embriología , Glándula Tiroides/embriología , Pez Cebra/embriología , Animales , Animales Modificados Genéticamente , Sistema Cardiovascular/enzimología , Embrión no Mamífero/enzimología , Receptores de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Pez Cebra/metabolismo
10.
Stem Cell Res ; 73: 103256, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-38006677

RESUMEN

The X-linked Allan-Herndon-Dudley syndrome (AHDS) is characterized by severely impaired psychomotor development and is caused by mutations in the SLC16A2 gene encoding the thyroid hormone transporter MCT8 (monocarboxylate transporter 8). By targeting exon 3 of SLC16A2 using CRISPR/Cas9 with single-stranded oligodeoxynucleotides as homology-directed repair templates, we introduced the AHDS patient missense variant G401R and a novel knock-out deletion variant (F400Sfs*17) into the male healthy donor hiPSC line BIHi001-B. We successfully generated cerebral organoids from these genome-edited lines, demonstrating the utility of the novel lines for modelling the effects of MCT8-deficency on human neurodevelopment.


Asunto(s)
Células Madre Pluripotentes Inducidas , Discapacidad Intelectual Ligada al Cromosoma X , Simportadores , Humanos , Masculino , Hormonas Tiroideas , Mutación , Transportadores de Ácidos Monocarboxílicos/genética , Discapacidad Intelectual Ligada al Cromosoma X/genética , Simportadores/genética
11.
Sci Transl Med ; 15(705): eadg1659, 2023 07 19.
Artículo en Inglés | MEDLINE | ID: mdl-37467315

RESUMEN

Increasing evidence points toward epigenetic variants as a risk factor for developing obesity. We analyzed DNA methylation of the POMC (pro-opiomelanocortin) gene, which is pivotal for satiety regulation. We identified sex-specific and nongenetically determined POMC hypermethylation associated with a 1.4-fold (confidence interval, 1.03 to 2.04) increased individual risk of developing obesity. To investigate the early embryonic establishment of POMC methylation states, we established a human embryonic stem cell (hESC) model. Here, hESCs (WA01) were transferred into a naïve state, which was associated with a reduction of DNA methylation. Naïve hESCs were differentiated via a formative state into POMC-expressing hypothalamic neurons, which was accompanied by re-establishment of DNA methylation patterning. We observed that reduced POMC gene expression was associated with increased POMC methylation in POMC-expressing neurons. On the basis of these findings, we treated POMC-hypermethylated obese individuals (n = 5) with an MC4R agonist and observed a body weight reduction of 4.66 ± 2.16% (means ± SD) over a mean treatment duration of 38.4 ± 26.0 weeks. In summary, we identified an epigenetic obesity risk variant at the POMC gene fulfilling the criteria for a metastable epiallele established in early embryonic development that may be addressable by MC4R agonist treatment to reduce body weight.


Asunto(s)
Obesidad , Proopiomelanocortina , Masculino , Embarazo , Femenino , Humanos , Proopiomelanocortina/genética , Proopiomelanocortina/metabolismo , Obesidad/genética , Obesidad/metabolismo , Peso Corporal/fisiología , Metilación de ADN/genética , Factores de Riesgo , Receptor de Melanocortina Tipo 4/genética , Receptor de Melanocortina Tipo 4/metabolismo
12.
Exp Clin Endocrinol Diabetes ; 130(2): 134-140, 2022 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-34352913

RESUMEN

The monocarboxylate transporter 8 (MCT8) is a specific thyroid hormone transporter and plays an essential role in fetal development. Inactivating mutations in the MCT8 encoding gene SLC16A2 (solute carrier family 16, member 2) lead to the Allan-Herndon-Dudley syndrome, a condition presenting with severe endocrinological and neurological phenotypes. However, the cellular distribution pattern and dynamic expression profile are still not well known for early human neural development. OBJECTIVE: Development and characterization of fluorescent MCT8 reporters that would permit live-cell monitoring of MCT8 protein expression in vitro in human induced pluripotent stem cell (hiPSC)-derived cell culture models. METHODS: A tetracysteine (TC) motif was introduced into the human MCT8 sequence at four different positions as binding sites for fluorescent biarsenical dyes. Human Embryonic Kidney 293 cells were transfected and stained with fluorescein-arsenical hairpin-binder (FlAsH). Counterstaining with specific MCT8 antibody was performed. Triiodothyronine (T3) uptake was indirectly measured with a T3 responsive luciferase-based reporter gene assay in Madin-Darby Canine Kidney 1 cells for functional characterization. RESULTS: FlAsH staining and antibody counterstaining of all four constructs showed cell membrane expression of all MCT8 constructs. The construct with the tag after the first start codon demonstrated comparable T3 uptake to the MCT8 wildtype. CONCLUSION: Our data indicate that introduction of a TC-tag directly after the first start codon generates a MCT8 reporter with suitable characteristics for live-cell monitoring of MCT8 expression. One promising future application will be generation of stable hiPSC MCT8 reporter lines to characterize MCT8 expression patterns during in vitro neuronal development.


Asunto(s)
Expresión Génica , Transportadores de Ácidos Monocarboxílicos , Simportadores , Fluoresceína , Colorantes Fluorescentes , Células HEK293 , Humanos , Células Madre Pluripotentes Inducidas , Coloración y Etiquetado
13.
Med Genet ; 33(2): 121-131, 2021 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38836035

RESUMEN

Whole exome sequencing discovers causative mutations in less than 50 % of rare disease patients, suggesting the presence of additional mutations in the non-coding genome. So far, non-coding mutations have been identified in less than 0.2 % of individuals with genetic diseases listed in the ClinVar database and exhibit highly diverse molecular mechanisms. In contrast to our capability to sequence the whole genome, our ability to discover and functionally confirm such non-coding mutations is lagging behind severely. We discuss the problems and present examples of confirmed mutations in deep intronic sequences, non-coding triplet repeats, enhancers, and larger structural variants and highlight their proposed disease mechanisms. Finally, we discuss the type of data that would be required to establish non-coding mutation detection in routine diagnostics.

14.
Thyroid ; 31(3): 420-438, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-32777984

RESUMEN

Background: Congenital hypothyroidism due to thyroid dysgenesis is a frequent congenital endocrine disorder for which the molecular mechanisms remain unresolved in the majority of cases. This situation reflects, in part, our still limited knowledge about the mechanisms involved in the early steps of thyroid specification from the endoderm, in particular the extrinsic signaling cues that regulate foregut endoderm patterning. In this study, we used small molecules and genetic zebrafish models to characterize the role of various signaling pathways in thyroid specification. Methods: We treated zebrafish embryos during different developmental periods with small-molecule compounds known to manipulate the activity of Wnt signaling pathway and observed effects in thyroid, endoderm, and cardiovascular development using whole-mount in situ hybridization and transgenic fluorescent reporter models. We used the antisense morpholino (MO) technique to create a zebrafish acardiac model. For thyroid rescue experiments, bone morphogenetic protein (BMP) pathway induction in zebrafish embryos was obtained by manipulation of heat-shock inducible transgenic lines. Results: Combined analyses of thyroid and cardiovascular development revealed that overactivation of Wnt signaling during early development leads to impaired thyroid specification concurrent with severe defects in the cardiac specification. When using a model of MO-induced blockage of cardiomyocyte differentiation, a similar correlation was observed, suggesting that defective signaling between cardiac mesoderm and endodermal thyroid precursors contributes to thyroid specification impairment. Rescue experiments through transient overactivation of BMP signaling could partially restore thyroid specification in models with defective cardiac development. Conclusion: Collectively, our results indicate that BMP signaling is critically required for thyroid cell specification and identify cardiac mesoderm as a likely source of BMP signals.


Asunto(s)
Proteína Morfogenética Ósea 2/metabolismo , Proteína Morfogenética Ósea 4/metabolismo , Hipotiroidismo Congénito/metabolismo , Proteínas del Citoesqueleto/metabolismo , Cardiopatías Congénitas/metabolismo , Miocitos Cardíacos/metabolismo , Disgenesias Tiroideas/metabolismo , Glándula Tiroides/metabolismo , Proteínas Wnt/metabolismo , Vía de Señalización Wnt , Proteínas de Pez Cebra/metabolismo , Animales , Animales Modificados Genéticamente , Proteína Morfogenética Ósea 2/genética , Proteína Morfogenética Ósea 4/genética , Hipotiroidismo Congénito/genética , Hipotiroidismo Congénito/patología , Proteínas del Citoesqueleto/genética , Modelos Animales de Enfermedad , Desarrollo Embrionario , Endodermo/anomalías , Endodermo/metabolismo , Regulación del Desarrollo de la Expresión Génica , Cardiopatías Congénitas/genética , Cardiopatías Congénitas/patología , Mesodermo/anomalías , Mesodermo/metabolismo , Morfolinos/genética , Morfolinos/metabolismo , Miocitos Cardíacos/patología , Oligonucleótidos Antisentido/genética , Oligonucleótidos Antisentido/metabolismo , Disgenesias Tiroideas/genética , Disgenesias Tiroideas/patología , Glándula Tiroides/anomalías , Proteínas Wnt/genética , Pez Cebra/embriología , Pez Cebra/genética , Pez Cebra/metabolismo , Proteínas de Pez Cebra/genética
15.
J Clin Endocrinol Metab ; 106(9): 2606-2616, 2021 08 18.
Artículo en Inglés | MEDLINE | ID: mdl-34036349

RESUMEN

CONTEXT: Pro-opiomelanocortin (POMC) and the melanocortin-4 receptor (MC4R) play a pivotal role in the leptin-melanocortin pathway. Mutations in these genes lead to monogenic types of obesity due to severe hyperphagia. In addition to dietary-induced obesity, a cardiac phenotype without hypertrophy has been identified in MC4R knockout mice. OBJECTIVE: We aimed to characterize cardiac morphology and function as well as tissue Na+ content in humans with mutations in POMC and MC4R genes. METHODS: A cohort of 42 patients (5 patients with bi-allelic POMC mutations, 6 heterozygous MC4R mutation carriers, 19 obese controls without known monogenic cause, and 12 normal weight controls) underwent cardiac magnetic resonance (CMR) imaging and 23Na-MRI. RESULTS: Monogenic obese patients with POMC or MC4R mutation respectively had a significantly lower left ventricular mass/body surface area (BSA) than nonmonogenic obese patients. Left ventricular end-diastolic volume/BSA was significantly lower in POMC- and MC4R-deficient patients than in nonmonogenic obese patients. Subcutaneous fat and skin Na+ content was significantly higher in POMC- and MC4R-deficient patients than in nonmonogenic obese patients. In these compartments, the water content was significantly higher in patients with POMC and MC4R mutation than in control groups. CONCLUSION: Patients with POMC or MC4R mutations carriers had a lack of transition to hypertrophy, significantly lower cardiac muscle mass/BSA, and stored more Na+ within the subcutaneous fat tissue than nonmonogenic obese patients. The results point towards the role of the melanocortin pathway for cardiac function and tissue Na+ storage and the importance of including cardiologic assessments into the diagnostic work-up of these patients.


Asunto(s)
Hipertrofia Ventricular Izquierda/etiología , Mutación , Proopiomelanocortina/genética , Receptor de Melanocortina Tipo 4/genética , Sodio/metabolismo , Función Ventricular Izquierda/fisiología , Adolescente , Agua Corporal/metabolismo , Femenino , Humanos , Hipertrofia Ventricular Izquierda/genética , Imagen por Resonancia Magnética , Masculino , Obesidad/complicaciones , Fenotipo , Proopiomelanocortina/fisiología , Receptor de Melanocortina Tipo 4/fisiología
16.
Mol Cell Endocrinol ; 500: 110635, 2020 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-31678421

RESUMEN

Thyroid hormone (TH) synthesis requires extracellular hydrogen peroxide generated by the NADPH oxidases, DUOX1 and DUOX2, with maturation factors, DUOXA1 and DUOXA2. In zebrafish, only one duox and one duoxa gene are present. Using a thyroid-specific reporter line, we investigated the role of Duox and Duoxa for TH biosynthesis in zebrafish larvae. Analysis of several zebrafish duox and duoxa mutant models consistently recovered hypothyroid phenotypes with hyperplastic goiter caused by impaired TH synthesis. Mutant larvae developed enlarged thyroids and showed increased expression of the EGFP reporter and thyroid functional markers including wild-type and mutated duox and duoxa transcripts. Treatment of zebrafish larvae with the NADPH oxidase inhibitor VAS2870 phenocopied the thyroid effects observed in duox or duoxa mutants. Additional functional in vitro assays corroborated the pharmacological inhibition of Duox activity by VAS2870. These data support the utility of this new experimental model to characterize endocrine disruptors of the thyroid function.


Asunto(s)
Benzoxazoles/farmacología , Oxidasas Duales/genética , Bocio/genética , Peróxido de Hidrógeno/metabolismo , NADPH Oxidasas/genética , Hormonas Tiroideas/biosíntesis , Triazoles/farmacología , Proteínas de Pez Cebra/genética , Animales , Modelos Animales de Enfermedad , Oxidasas Duales/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Redes Reguladoras de Genes/efectos de los fármacos , Bocio/metabolismo , Mutación , NADPH Oxidasas/metabolismo , Pez Cebra/embriología , Pez Cebra/genética , Pez Cebra/metabolismo , Proteínas de Pez Cebra/metabolismo
17.
Thyroid ; 30(9): 1366-1383, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32143555

RESUMEN

Background: Mutations of monocarboxylate transporter 8 (MCT8), a thyroid hormone (TH)-specific transmembrane transporter, cause a severe neurodevelopmental disorder, the Allan-Herndon-Dudley syndrome. In MCT8 deficiency, TH is not able to reach those areas of the brain where TH uptake depends on MCT8. Currently, therapeutic options for MCT8-deficient patients are missing, as TH treatment is not successful in improving neurological deficits. Available data on MCT8 protein and transcript levels indicate complex expression patterns in neural tissue depending on species, brain region, sex, and age. However, information on human MCT8 expression is still scattered and additional efforts are needed to map sites of MCT8 expression in neurovascular units and neural tissue. This is of importance because new therapeutic strategies for this disease are urgently needed. Methods: To identify regions and time windows of MCT8 expression, we used highly specific antibodies against MCT8 to perform immunofluorescence labeling of postnatal murine brains, adult human brain tissue, and human cerebral organoids. Results: Qualitative and quantitative analyses of murine brain samples revealed stable levels of MCT8 protein expression in endothelial cells of the blood-brain barrier (BBB), choroid plexus epithelial cells, and tanycytes during postnatal development. Conversely, the neuronal MCT8 protein expression that was robustly detectable in specific brain regions of young mice strongly declined with age. Similarly, MCT8 immunoreactivity in adult human brain tissue was largely confined to endothelial cells of the BBB. Recently, cerebral organoids emerged as promising models of human neural development and our first analyses of forebrain-like organoids revealed MCT8 expression in early neuronal progenitor cell populations. Conclusions: With respect to MCT8-deficient conditions, our analyses not only strongly support the contention that the BBB presents a lifelong barrier to TH uptake but also highlight the need to decipher the TH transport role of MCT8 in early neuronal cell populations in more detail. Improving the understanding of the spatiotemporal expression in latter barriers will be critical for therapeutic strategies addressing MCT8 deficiency in the future.


Asunto(s)
Regulación de la Expresión Génica , Transportadores de Ácidos Monocarboxílicos/biosíntesis , Mutación , Simportadores/biosíntesis , Anciano , Anciano de 80 o más Años , Animales , Encéfalo/metabolismo , Línea Celular , Perros , Células Endoteliales/metabolismo , Perfilación de la Expresión Génica , Humanos , Discapacidad Intelectual Ligada al Cromosoma X/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Hipotonía Muscular/metabolismo , Atrofia Muscular/metabolismo , Neurogénesis , Neuronas/metabolismo , Prosencéfalo/metabolismo , Triyodotironina/metabolismo
18.
Thyroid ; 29(11): 1683-1703, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31507237

RESUMEN

Background: Defects in embryonic development of the thyroid gland are a major cause for congenital hypothyroidism in human newborns, but the underlying molecular mechanisms are still poorly understood. Organ development relies on a tightly regulated interplay between extrinsic signaling cues and cell intrinsic factors. At present, however, there is limited knowledge about the specific extrinsic signaling cues that regulate foregut endoderm patterning, thyroid cell specification, and subsequent morphogenetic processes in thyroid development. Methods: To begin to address this problem in a systematic way, we used zebrafish embryos to perform a series of in vivo phenotype-driven chemical genetic screens to identify signaling cues regulating early thyroid development. For this purpose, we treated zebrafish embryos during different developmental periods with a panel of small-molecule compounds known to manipulate the activity of major signaling pathways and scored phenotypic deviations in thyroid, endoderm, and cardiovascular development using whole-mount in situ hybridization and transgenic fluorescent reporter models. Results: Systematic assessment of drugged embryos recovered a range of thyroid phenotypes including expansion, reduction or lack of the early thyroid anlage, defective thyroid budding, as well as hypoplastic, enlarged, or overtly disorganized presentation of the thyroid primordium after budding. Our pharmacological screening identified bone morphogenetic protein and fibroblast growth factor signaling as key factors for thyroid specification and early thyroid organogenesis, highlighted the importance of low Wnt activities during early development for thyroid specification, and implicated drug-induced cardiac and vascular anomalies as likely indirect mechanisms causing various forms of thyroid dysgenesis. Conclusions: By integrating the outcome of our screening efforts with previously available information from other model organisms including Xenopus, chicken, and mouse, we conclude that signaling cues regulating thyroid development appear broadly conserved across vertebrates. We therefore expect that observations made in zebrafish can inform mammalian models of thyroid organogenesis to further our understanding of the molecular mechanisms of congenital thyroid diseases.


Asunto(s)
Embrión no Mamífero , Transducción de Señal/genética , Glándula Tiroides/embriología , Pez Cebra/genética , Animales , Proteínas Morfogenéticas Óseas/genética , Desarrollo Embrionario/efectos de los fármacos , Desarrollo Embrionario/genética , Factores de Crecimiento de Fibroblastos/genética , Regulación del Desarrollo de la Expresión Génica , Ensayos Analíticos de Alto Rendimiento , Péptidos y Proteínas de Señalización Intercelular/genética , Organismos Modificados Genéticamente , Fenotipo , Bibliotecas de Moléculas Pequeñas , Disgenesias Tiroideas/genética , Glándula Tiroides/anomalías
20.
J Endocrinol ; 190(1): 157-70, 2006 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16837620

RESUMEN

The uptake of iodide represents the first step in thyroid hormone synthesis by thyroid follicular cells and is mediated by the sodium-iodide symporter (NIS). In mammals, expression of NIS is stimulated by TSH and transcription of the NIS gene involves regulation by the thyroid-specific transcription factors Pax8 and Nkx2.1. In this study, we examined the mRNA expression of NIS, Pax8 and Nkx2.1 in the thyroid gland of Xenopus laevis tadpoles by semi-quantitative reverse transcriptase (RT)-PCR. During spontaneous metamorphosis, NIS mRNA expression was low in premetamorphic tadpoles, increased throughout prometamorphosis, and peaked at climax stage 60. Analysis of TSH beta-subunit (TSHbeta) mRNA in the pituitary of the same tadpoles revealed a close temporal relationship in the expression of the two genes during metamorphosis, suggesting a regulatory role of TSH in the developmental expression of NIS. Treatment of tadpoles with goitrogenic compounds (sodium perchlorate and ethylenethiourea) increased TSHbeta mRNA expression (approximately twofold) and caused thyroid gland hyperplasia, confirming that feedback along the pituitary-thyroid axis was operative. Analysis of gene expression in the thyroid gland revealed that goitrogen treatment was correlated with increased expression of NIS mRNA (approximately 20-fold). In the thyroid gland organ culture experiments, bovine TSH (bTSH; 1 mU/ml) strongly induced NIS mRNA expression. This effect was mimicked by co-culture of thyroid glands with pituitaries from stage 58 tadpoles and by agents that increase intracellular cAMP (forskolin, dibutyryl-cAMP). In addition, it could be shown that thyroid glands of X. laevis tadpoles express Pax8 and Nkx2.1 mRNA in a developmentally regulated manner and that ex vivo treatment of thyroid glands with bTSH, forskolin, and cAMP analogs increased the expression of Pax8 and Nkx2.1 mRNA. This is the first report on developmental profiles and hormonal regulation of thyroid gland gene expression in amphibian tadpoles and, together, results reveal a critical role of TSH in the regulation of NIS mRNA expression in the thyroid gland of X. laevis tadpoles.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , ARN Mensajero/análisis , Simportadores/genética , Glándula Tiroides/metabolismo , Xenopus laevis/metabolismo , Animales , Antitiroideos/farmacología , Bovinos , Etilenotiourea/farmacología , Expresión Génica , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Larva , Metamorfosis Biológica , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Técnicas de Cultivo de Órganos , Factor de Transcripción PAX8 , Factores de Transcripción Paired Box/genética , Factores de Transcripción Paired Box/metabolismo , Percloratos/farmacología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Compuestos de Sodio/farmacología , Glándula Tiroides/efectos de los fármacos , Factor Nuclear Tiroideo 1 , Tirotropina/farmacología , Tirotropina de Subunidad beta/genética , Tirotropina de Subunidad beta/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Transcripción Genética , Proteínas de Xenopus/genética , Proteínas de Xenopus/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA