Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
J Endocrinol ; 232(2): 259-272, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27879339

RESUMEN

Inactivating mutations in the human SLC16A2 gene encoding the thyroid hormone transporter monocarboxylate transporter 8 (MCT8) result in the Allan-Herndon-Dudley syndrome accompanied by severe locomotor deficits. The underlying mechanisms of the associated cerebellar maldevelopment were studied using the chicken as a model. Electroporation of an MCT8-RNAi vector into the cerebellar anlage of a 3-day-old embryo allowed knockdown of MCT8 in Purkinje cell precursors. This resulted in the downregulation of the thyroid hormone-responsive gene RORα and the Purkinje cell-specific differentiation marker LHX1/5 at day 6. MCT8 knockdown also results in a smaller and less complex dendritic tree at day 18 suggesting a pivotal role of MCT8 for cell-autonomous Purkinje cell maturation. Early administration of the thyroid hormone analogue 3,5,3'-triiodothyroacetic acid partially rescued early Purkinje cell differentiation. MCT8-deficient Purkinje cells also induced non-autonomous effects as they led to a reduced granule cell precursor proliferation, a thinner external germinal layer and a loss of PAX6 expression. By contrast, at day 18, the external germinal layer thickness was increased, with an increase in presence of Axonin-1-positive post-mitotic granule cells in the initial stage of radial migration. The concomitant accumulation of presumptive migrating granule cells in the molecular layer, suggests that inward radial migration to the internal granular layer is stalled. In conclusion, early MCT8 deficiency in Purkinje cells results in both cell-autonomous and non-autonomous effects on cerebellar development and indicates that MCT8 expression is essential from very early stages of development, providing a novel insight into the ontogenesis of the Allan-Herndon-Dudley syndrome.


Asunto(s)
Cerebelo/embriología , Transportadores de Ácidos Monocarboxílicos/metabolismo , Neurogénesis/genética , Organogénesis/genética , Células de Purkinje/metabolismo , Animales , Movimiento Celular/genética , Cerebelo/citología , Cerebelo/metabolismo , Embrión de Pollo , Regulación hacia Abajo , Desarrollo Embrionario , Discapacidad Intelectual Ligada al Cromosoma X/genética , Discapacidad Intelectual Ligada al Cromosoma X/metabolismo , Transportadores de Ácidos Monocarboxílicos/genética , Hipotonía Muscular/genética , Hipotonía Muscular/metabolismo , Atrofia Muscular/genética , Atrofia Muscular/metabolismo , Miembro 1 del Grupo F de la Subfamilia 1 de Receptores Nucleares/genética , Miembro 1 del Grupo F de la Subfamilia 1 de Receptores Nucleares/metabolismo , Células de Purkinje/citología
2.
Endocrinology ; 157(6): 2560-74, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27070099

RESUMEN

Thyroid hormone (TH) transmembrane transporters are key regulators of TH availability in target cells where correct TH signaling is essential for normal development. Although the chicken embryo is a valuable model for developmental studies, the only functionally characterized chicken TH transporter so far is the organic anion transporting polypeptide 1C1 (OATP1C1). We therefore cloned the chicken L-type amino acid transporter 1 (LAT1) and the monocarboxylate transporters 8 (MCT8) and 10 (MCT10), and functionally characterized them, together with OATP1C1, in JEG3, COS1, and DF-1 cells. In addition, we used in situ hybridization to study their mRNA expression pattern during development. MCT8 and OATP1C1 are both high affinity transporters for the prohormone T4, whereas receptor-active T3 is preferably transported by MCT8 and MCT10. The latter one shows lower affinity but has a high Vmax and seems to be especially good at T3 export. Also, LAT1 has a lower affinity for its preferred substrate 3,3'-diiodothyronine. Reverse T3 is transported by all 4 TH transporters and is a good export product for OATP1C1. TH transporters are strongly expressed in eye (LAT1, MCT8, MCT10), pancreas (LAT1, MCT10), kidney, and testis (MCT8). Their extensive expression in the central nervous system, especially at the brain barriers, indicates an important role in brain development. In conclusion, we show TH transport by chicken MCT8, MCT10, and LAT1. Together with OATP1C1, these transporters have functional characteristics similar to their mammalian orthologs and are interesting target genes to further elucidate the role of THs during embryonic development.


Asunto(s)
Hormonas Tiroideas/metabolismo , Sistemas de Transporte de Aminoácidos Neutros/metabolismo , Animales , Células COS , Línea Celular Tumoral , Membrana Celular/metabolismo , Pollos , Embrión no Mamífero/metabolismo , Humanos , Inmunohistoquímica , Hibridación in Situ , Transportador de Aminoácidos Neutros Grandes 1/metabolismo , Transportadores de Ácidos Monocarboxílicos/metabolismo , Transportadores de Anión Orgánico/metabolismo
3.
Cerebellum ; 15(6): 710-725, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-26559893

RESUMEN

The cerebellum is a morphologically unique brain structure that requires thyroid hormones (THs) for the correct coordination of key cellular events driving its development. Unravelling the interplay between the multiple factors that can regulate intracellular TH levels is a key step to understanding their role in the regulation of these cellular processes. We therefore investigated the regional/cell-specific expression pattern of TH transporters and deiodinases in the cerebellum using the chicken embryo as a model. In situ hybridisation revealed expression of the TH transporters monocarboxylate transporter 8 (MCT8) and 10 (MCT10), L-type amino acid transporter 1 (LAT1) and organic anion transporting polypeptide 1C1 (OATP1C1) as well as the inactivating type 3 deiodinase (D3) in the fourth ventricle choroid plexus, suggesting a possible contribution of the resulting proteins to TH exchange and subsequent inactivation of excess hormone at the blood-cerebrospinal fluid barrier. Exclusive expression of LAT1 and the activating type 2 deiodinase (D2) mRNA was found at the level of the blood-brain barrier, suggesting a concerted function for LAT1 and D2 in the direct access of active T3 to the developing cerebellum via the capillary endothelial cells. The presence of MCT8 mRNA in Purkinje cells and cerebellar nuclei during the first 2 weeks of embryonic development points to a potential role of this transporter in the uptake of T3 in central neurons. At later stages, together with MCT10, detection of MCT8 signal in close association with the Purkinje cell dendritic tree suggests a role of both transporters in TH signalling during Purkinje cell synaptogenesis. MCT10 was also expressed in late-born cells in the rhombic lip lineage with a clear hybridisation signal in the outer external granular layer, indicating a potential role for MCT10 in the proliferation of granule cell precursors. By contrast, expression of D3 in the first-born rhombic lip-derived population may serve as a buffering mechanism against high T3 levels during early embryonic development, a hypothesis supported by the pattern of expression of a fluorescent TH reporter in this lineage. Overall, this study builds a picture of the TH dependency in multiple cerebellar cell types starting from early embryonic development.


Asunto(s)
Cerebelo/embriología , Cerebelo/metabolismo , Hormonas Tiroideas/metabolismo , Animales , Proteínas Aviares/metabolismo , Barrera Hematoencefálica/embriología , Barrera Hematoencefálica/metabolismo , Linaje de la Célula , Cerebelo/citología , Embrión de Pollo , Electroporación , Regulación del Desarrollo de la Expresión Génica , Inmunohistoquímica , Hibridación in Situ , Proteínas de Transporte de Membrana/metabolismo , Microscopía Fluorescente , Modelos Animales , Neuronas/citología , Neuronas/metabolismo , ARN Mensajero/metabolismo , Sinapsis/metabolismo
4.
Gen Comp Endocrinol ; 214: 30-9, 2015 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-25745816

RESUMEN

Thyroid hormones (THs) are key regulators in the development of the vertebrate brain. Therefore, TH access to the developing brain needs to be strictly regulated. The brain barriers separate the central nervous system from the rest of the body and impose specific transport mechanisms on the exchange of molecules between the general circulation and the nervous system. As such they form ideal structures for regulating TH exchange between the blood and the brain. To investigate the mechanism by which the developing brain regulates TH availability, we investigated the ontogenetic expression profiles of TH transporters, deiodinases and the TH distributor protein transthyretin (TTR) at the brain barriers during embryonic and early postnatal development using the chicken as a model. In situ hybridisation revealed expression of the TH transporters monocarboxylate transporter 8 (MCT8) and 10 (MCT10), organic anion transporting polypeptide 1C1 (OATP1C1) and L-type amino acid transporter 1 (LAT1) and the inactivating type 3 deiodinase (D3) in the choroid plexus which forms the blood-cerebrospinal fluid barrier. This was confirmed by quantitative PCR which additionally indicated strongly increasing expression of TTR as well as detectable expression of the activating type 2 deiodinase (D2) and the (in)activating type 1 deiodinase (D1). In the brain capillaries forming the blood-brain barrier in situ hybridisation showed exclusive expression of LAT1 and D2. The combined presence of LAT1 and D2 in brain capillaries suggests that the blood-brain barrier forms the main route for receptor-active T3 uptake into the embryonic chicken brain. Expression of multiple transporters, deiodinases and TTR in the choroid plexus indicates that the blood-cerebrospinal fluid barrier is also important in regulating early TH availability. The impact of these barrier systems can be deduced from the clear difference in T3 and T4 levels as well as the T3/T4 ratio between the developing brain and the general circulation. We conclude that the tight regulation of TH exchange at the brain barriers from early embryonic stages is one of the factors needed to allow the brain to develop within a relative microenvironment.


Asunto(s)
Barrera Hematoencefálica/metabolismo , Encéfalo/citología , Pollos/crecimiento & desarrollo , Yoduro Peroxidasa/metabolismo , Proteínas de Transporte de Membrana/metabolismo , Transportadores de Anión Orgánico/metabolismo , Hormonas Tiroideas/metabolismo , Animales , Encéfalo/metabolismo , Embrión de Pollo/crecimiento & desarrollo , Embrión de Pollo/metabolismo , Pollos/genética , Pollos/metabolismo , Plexo Coroideo/metabolismo , Femenino , Regulación del Desarrollo de la Expresión Génica , Homeostasis , Hibridación in Situ , Yoduro Peroxidasa/genética , Transportador de Aminoácidos Neutros Grandes 1/genética , Transportador de Aminoácidos Neutros Grandes 1/metabolismo , Proteínas de Transporte de Membrana/genética , Transportadores de Anión Orgánico/genética , Prealbúmina/genética , Prealbúmina/metabolismo , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Glándula Tiroides/embriología , Glándula Tiroides/metabolismo
5.
Front Neurosci ; 9: 66, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25784853

RESUMEN

Thyroid hormones are key players in regulating brain development. Thus, transfer of appropriate quantities of thyroid hormones from the blood into the brain at specific stages of development is critical. The choroid plexus forms the blood-cerebrospinal fluid barrier. In reptiles, birds and mammals, the main protein synthesized and secreted by the choroid plexus is a thyroid hormone distributor protein: transthyretin. This transthyretin is secreted into the cerebrospinal fluid and moves thyroid hormones from the blood into the cerebrospinal fluid. Maximal transthyretin synthesis in the choroid plexus occurs just prior to the period of rapid brain growth, suggesting that choroid plexus-derived transthyretin moves thyroid hormones from blood into cerebrospinal fluid just prior to when thyroid hormones are required for rapid brain growth. The structure of transthyretin has been highly conserved, implying strong selection pressure and an important function. In mammals, transthyretin binds T4 (precursor form of thyroid hormone) with higher affinity than T3 (active form of thyroid hormone). In all other vertebrates, transthyretin binds T3 with higher affinity than T4. As mammals are the exception, we should not base our thinking about the role of transthyretin in the choroid plexus solely on mammalian data. Thyroid hormone transmembrane transporters are involved in moving thyroid hormones into and out of cells and have been identified in many tissues, including the choroid plexus. Thyroid hormones enter the choroid plexus via thyroid hormone transmembrane transporters and leave the choroid plexus to enter the cerebrospinal fluid via either thyroid hormone transmembrane transporters or via choroid plexus-derived transthyretin secreted into the cerebrospinal fluid. The quantitative contribution of each route during development remains to be elucidated. This is part of a review series on ontogeny and phylogeny of brain barrier mechanisms.

6.
Biochim Biophys Acta ; 1849(2): 130-41, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24844179

RESUMEN

BACKGROUND: Thyroid hormones (THs) play an essential role in vertebrate development, acting predominantly via nuclear TH receptors (TRs) which are ligand-dependent transcription factors. Binding of the ligand (predominantly T3) induces a switch from gene activation to gene repression or vice versa. Iodothyronine deiodinases (Ds) and TH transporters are important regulators of intracellular T3 availability and therefore contribute to the control of TR-dependent development. FOCUS: The present review discusses the possible roles of Ds and TH transporters in regulating embryonic and larval (pre-juvenile) TR-dependent development in vertebrates. It focuses mainly on well-known model species for direct and indirect vertebrate development, including zebrafish, Xenopus, chicken and mouse. Data are provided on stage- and tissue/cell-specific changes in expression of Ds and TH transporters. This information is combined with functional data obtained from gain-and-loss of function studies. CONCLUSION: Knockout/knockdown of each type of D has provided strong evidence for their implication in the control of important developmental processes and several D expression patterns and functions have been conserved throughout vertebrate evolution. Knockout/knockdown of the inactivating D3 enzyme indicates that a premature switch from unliganded to liganded TR action is often more detrimental than a delayed one. The majority of ontogenetic studies on TH transporter distribution and function have focused on brain development, showing variable impact of knockout/knockdown depending on the species. Future research in different models using conditional silencing will hopefully further improve our understanding on how TH transporters, Ds and TRs cooperate to regulate TR-mediated impact on vertebrate development. This article is part of a Special Issue entitled: Nuclear receptors in animal development.


Asunto(s)
Receptores de Hormona Tiroidea/fisiología , Hormonas Tiroideas/metabolismo , Anfibios/embriología , Anfibios/genética , Animales , Embrión de Pollo , Regulación del Desarrollo de la Expresión Génica , Humanos , Yoduro Peroxidasa/fisiología , Ratones , Ratones Noqueados , Receptores de Hormona Tiroidea/metabolismo , Pez Cebra/embriología , Pez Cebra/genética
7.
Endocrinology ; 155(4): 1547-59, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24467742

RESUMEN

Exposure to appropriate levels of thyroid hormones (THs) at the right time is of key importance for normal development in all vertebrates. Type 3 iodothyronine deiodinase (D3) is the prime TH-inactivating enzyme, and its expression is highest in the early stages of vertebrate development, implying that it may be necessary to shield developing tissues from overexposure to THs. We used antisense morpholino knockdown to examine the role of D3 during early development in zebrafish. Zebrafish possess 2 D3 genes, dio3a and dio3b. Here, we show that both genes are expressed during development and both contribute to in vivo D3 activity. However, dio3b mRNA levels in embryos are higher, and the effects of dio3b knockdown on D3 activity and on the resulting phenotype are more severe. D3 knockdown induced an overall delay in development, as determined by measurements of otic vesicle length, eye and ear size, and body length. The time of hatching was also severely delayed in D3-knockdown embryos. Importantly, we also observed a severe disturbance of several aspects of development. Swim bladder development and inflation was aberrant as was the development of liver and intestine. Furthermore, D3-knockdown larvae spent significantly less time moving, and both embryos and larvae exhibited perturbed escape responses, suggesting that D3 knockdown affects muscle development and/or functioning. These data indicate that D3 is essential for normal zebrafish embryonic and early larval development and show the value of morpholino knockdown in this model to further elucidate the specific role of D3 in some aspects of vertebrate development.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Yoduro Peroxidasa/genética , Yoduro Peroxidasa/fisiología , Pez Cebra/embriología , Animales , Embrión no Mamífero/fisiología , Técnicas de Silenciamiento del Gen , Hibridación in Situ , Intestinos/embriología , Hígado/embriología , Oligonucleótidos Antisentido/química , Fenotipo , ARN Mensajero/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Hormonas Tiroideas/metabolismo
8.
Gen Comp Endocrinol ; 190: 96-104, 2013 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-23707378

RESUMEN

Thyroid hormones (THs) are crucial elements in vertebrate brain development. They exert their action mainly through binding of 3,5,3'-triiodothyronine (T3) to nuclear receptors that directly influence the expression of TH-regulated genes. Intracellular TH action is therefore dependent on both the availability of T3 and its receptors. TH uptake in cells is regulated by specific TH transporters and local activation and inactivation is regulated by deiodinases. This review provides an overview of the general expression pattern of TH transporters, deiodinases and receptors during embryonic chicken brain development and compares it to the situation in mammals. It is clear that THs and their regulators are present in the embryonic brain from the early stages of development, long before the onset of embryonic thyroid gland functioning. The mechanism of TH uptake across the brain barriers during development is only partly understood. At the developing blood-brain-barrier expression of the TH-activating type 2 deiodinase is closely associated with the blood vessels, but contrary to the situation in (adult) mammals no expression of MCT8 or OATP1C1 TH transporters is found at that level in the developing chicken. At the blood-cerebrospinal fluid-barrier co-expression of the TH-inactivating type 3 deiodinase and MCT8 and OATP1C1 is found in birds and mammals. These comparative data show overlapping patterns, pointing to general mechanisms, but also indicate specific interspecies differences that may help to understand species-specific responses to regulator gene knockout/mutation.


Asunto(s)
Encéfalo/embriología , Hormonas Tiroideas/metabolismo , Animales , Embrión de Pollo , Pollos
9.
J Endocrinol ; 218(1): 105-15, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23608220

RESUMEN

Methimazole (MMI) is an anti-thyroid drug used in the treatment of chronic hyperthyroidism. There is, however, some debate about its use during pregnancy as MMI is known to cross the mammalian placenta and reach the developing foetus. A similar problem occurs in birds, where MMI is deposited in the egg and taken up by the developing embryo. To investigate whether maternally derived MMI can have detrimental effects on embryonic development, we treated laying hens with MMI (0.03% in drinking water) and measured total and reduced MMI contents in the tissues of hens and embryos at different stages of development. In hens, MMI was selectively increased in the thyroid gland, while its levels in the liver and especially brain remained relatively low. Long-term MMI treatment induced a pronounced goitre with a decrease in thyroxine (T4) content but an increase in thyroidal 3,5,3'-triiodothyronine (T3) content. This resulted in normal T3 levels in tissues except in the brain. In chicken embryos, MMI levels were similar in the liver and brain. They gradually decreased during development but always remained above those in the corresponding maternal tissues. Contrary to the situation in hens, T4 availability was only moderately affected in embryos. Peripheral T3 levels were reduced in 14-day-old embryos but normal in 18-day-old embryos, while brain T3 content was decreased at all embryonic stages tested. We conclude that all embryonic tissues are exposed to relatively high doses of MMI and its oxidised metabolites. The effect of maternal MMI treatment on embryonic thyroid hormone availability is most pronounced for brain T3 content, which is reduced throughout the embryonic development period.


Asunto(s)
Antitiroideos/farmacocinética , Desarrollo Embrionario/efectos de los fármacos , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Hipotiroidismo/inducido químicamente , Metimazol/farmacocinética , Glándula Tiroides/efectos de los fármacos , Hormonas Tiroideas/metabolismo , Animales , Antitiroideos/efectos adversos , Antitiroideos/metabolismo , Biotransformación , Encéfalo/efectos de los fármacos , Encéfalo/embriología , Encéfalo/metabolismo , Embrión de Pollo , Pollos , Clara de Huevo/química , Yema de Huevo/química , Femenino , Hipotiroidismo/embriología , Yoduro Peroxidasa/genética , Yoduro Peroxidasa/metabolismo , Riñón/efectos de los fármacos , Riñón/embriología , Riñón/metabolismo , Hígado/efectos de los fármacos , Hígado/embriología , Hígado/metabolismo , Metimazol/efectos adversos , Metimazol/metabolismo , Tamaño de los Órganos/efectos de los fármacos , Oxidación-Reducción , ARN Mensajero/metabolismo , Glándula Tiroides/embriología , Glándula Tiroides/metabolismo , Hormonas Tiroideas/sangre , Distribución Tisular
10.
J Endocrinol ; 215(2): 189-206, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22825922

RESUMEN

Iodothyronine deiodinases are important mediators of thyroid hormone (TH) action. They are present in tissues throughout the body where they catalyse 3,5,3'-triiodothyronine (T(3)) production and degradation via, respectively, outer and inner ring deiodination. Three different types of iodothyronine deiodinases (D1, D2 and D3) have been identified in vertebrates from fish to mammals. They share several common characteristics, including a selenocysteine residue in their catalytic centre, but show also some type-specific differences. These specific characteristics seem very well conserved for D2 and D3, while D1 shows more evolutionary diversity related to its Km, 6-n-propyl-2-thiouracil sensitivity and dependence on dithiothreitol as a cofactor in vitro. The three deiodinase types have an impact on systemic T(3) levels and they all contribute directly or indirectly to intracellular T(3) availability in different tissues. The relative contribution of each of them, however, varies amongst species, developmental stages and tissues. This is especially true for amphibians, where the impact of D1 may be minimal. D2 and D3 expression and activity respond to thyroid status in an opposite and conserved way, while the response of D1 is variable, especially in fish. Recently, a number of deiodinases have been cloned from lower chordates. Both urochordates and cephalochordates possess selenodeiodinases, although they cannot be classified in one of the three vertebrate types. In addition, the cephalochordate amphioxus also expresses a non-selenodeiodinase. Finally, deiodinase-like sequences have been identified in the genome of non-deuterostome organisms, suggesting that deiodination of externally derived THs may even be functionally relevant in a wide variety of invertebrates.


Asunto(s)
Evolución Biológica , Regulación Enzimológica de la Expresión Génica/fisiología , Yoduro Peroxidasa/química , Yoduro Peroxidasa/metabolismo , Animales , Variación Genética , Humanos , Yoduro Peroxidasa/genética , Especificidad de la Especie , Relación Estructura-Actividad
11.
Mol Cell Endocrinol ; 349(2): 289-97, 2012 Feb 26.
Artículo en Inglés | MEDLINE | ID: mdl-22120204

RESUMEN

We used the chick embryo to study the mechanisms regulating intracellular TH availability in developing brain. TH-transporters OATP1C1 and MCT8, and deiodinases D1, D2, and D3 were expressed in a region-specific way, well before the onset of endogenous TH secretion. Between day 4 and 10 of development MCT8 and D2 mRNA levels increased, while OATP1C1 and D3 mRNA levels decreased. D2 and D3 mRNAs were translated into active protein, while no D1 activity was detectable. Injection of THs into the yolk 24h before sampling increased TH levels in the brain and resulted in decreased OATP1C1 and increased MCT8 expression in 4-day-old embryos. A compensatory response in deiodinase activity was only observed at day 8. We conclude that THs are active in the early embryonic brain and TH-transporters and deiodinases can regulate their availability. However, the absence of clear compensatory mechanisms at day 4 makes the brain more vulnerable for changes in maternal TH supply.


Asunto(s)
Encéfalo/metabolismo , Yoduro Peroxidasa/metabolismo , Proteínas de Transporte de Membrana/metabolismo , Glándula Tiroides/metabolismo , Tiroxina/metabolismo , Triyodotironina/metabolismo , Animales , Encéfalo/efectos de los fármacos , Encéfalo/embriología , Embrión de Pollo , Pollos , Desarrollo Embrionario , Regulación del Desarrollo de la Expresión Génica , Yoduro Peroxidasa/genética , Proteínas de Transporte de Membrana/genética , Especificidad de Órganos , Reacción en Cadena de la Polimerasa , ARN Mensajero/análisis , ARN Mensajero/biosíntesis , Glándula Tiroides/embriología , Tiroxina/farmacología , Factores de Tiempo , Triyodotironina/farmacología
12.
J Thyroid Res ; 2011: 402320, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21760979

RESUMEN

Chicken and zebrafish are two model species regularly used to study the role of thyroid hormones in vertebrate development. Similar to mammals, chickens have one thyroid hormone receptor α (TRα) and one TRß gene, giving rise to three TR isoforms: TRα, TRß2, and TRß0, the latter with a very short amino-terminal domain. Zebrafish also have one TRß gene, providing two TRß1 variants. The zebrafish TRα gene has been duplicated, and at least three TRα isoforms are expressed: TRαA1-2 and TRαB are very similar, while TRαA1 has a longer carboxy-terminal ligand-binding domain. All these TR isoforms appear to be functional, ligand-binding receptors. As in other vertebrates, the different chicken and zebrafish TR isoforms have a divergent spatiotemporal expression pattern, suggesting that they also have distinct functions. Several isoforms are expressed from the very first stages of embryonic development and early chicken and zebrafish embryos respond to thyroid hormone treatment with changes in gene expression. Future studies in knockdown and mutant animals should allow us to link the different TR isoforms to specific processes in embryonic development.

13.
Endocrinology ; 150(11): 5171-80, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19819956

RESUMEN

The type 3 iodothyronine deiodinase (D3) is the primary deiodinase that inactivates thyroid hormone. Immunoprecipitation of D3, followed by fluorescent two-dimensional difference gel electrophoresis and mass spectrometry, identified peroxiredoxin 3 (Prx3) as a D3-associated protein. This interaction was confirmed using reverse coimmunoprecipitation, in which pull-down of Prx3 resulted in D3 isolation, and by fluorescence resonance energy transfer between cyan fluorescent protein-D3 and yellow fluorescent protein-Prx3. Prx3 overexpression did not change D3 activity in transfected HEK 293 cells; however, Prx3 knockdown resulted in a 50% decrease in D3-mediated whole-cell deiodination. Notably, D3 activity of cell lysates with dithiothreitol as an exogenous reducing factor and D3 protein levels were not decreased with Prx3 knockdown, indicating that the observed reduction in whole-cell deiodination was not simply due to a decrease in D3 enzyme levels. Prx3 knockdown did not change D3's affinity for T3 because saturation of D3-mediated whole-cell deiodination occurred between 20 and 200 nm T3 both with and without Prx3. Furthermore, the decrease in D3 activity in whole cells was not attributable to nonspecific oxidative stress because pretreatment with the antioxidant N-acetyl cysteine did not reverse the effects of Prx3 knockdown. Thioredoxin, the cofactor needed for Prx3 regeneration, supported D3 microsomal activity; however, Prx3 knockdown did not change D3 activity in this system. In conclusion, knockdown of Prx3 decreases D3 activity in whole cells, whereas absolute levels of D3 are unchanged, consistent with Prx3 playing a rate-limiting role in the regeneration of the D3 enzyme.


Asunto(s)
Yoduro Peroxidasa/metabolismo , Peroxirredoxinas/genética , Peroxirredoxinas/metabolismo , Línea Celular , Técnicas de Silenciamiento del Gen , Halogenación , Humanos , Yoduro Peroxidasa/genética , Unión Proteica , Triyodotironina/metabolismo
14.
Gen Comp Endocrinol ; 163(1-2): 58-62, 2009 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-19063893

RESUMEN

Thyroid hormones (THs) play an important role in vertebrate brain development by stimulating and coordinating cell proliferation, migration and differentiation. Several TH-responsive genes involved in these processes have been identified, but the information is mainly derived from studies of late brain development, while relatively little is known about the more early stages, prior to the onset of embryonic TH secretion. We have chosen the chick embryo to investigate the role of THs in both late and early brain development. T(4) and T(3) are found in chicken brain from the earliest stages tested (day 4). Indirect clues for the involvement of T(3) in brain development are found in the ontogenetic expression profiles of proteins regulating its bioavailability and action, including TH transporters, deiodinases and TH-receptors. All of them are expressed in whole embryos tested on day 2 of incubation and in developing brain tested from day 4 onwards. Their distribution patterns vary over time and according to the brain area and cell type studied. Hypothyroidism induced during the second half of incubation disturbs cell migration in the cerebellum, providing more direct evidence for the requirement for THs during the later stages of brain development. Direct morphological proof for the requirement for THs during the first half of incubation is still missing, but microarray analysis of telencephalon shows a clearly divergent gene expression profile in hypothyroid embryos. In vivo knockdown of TH transporters and deiodinases in chick embryos cultured ex ovo provides an excellent tool to study the role of THs in early brain development in more detail.


Asunto(s)
Encéfalo/embriología , Encéfalo/metabolismo , Pollos/fisiología , Hormonas Tiroideas/fisiología , Animales , Embrión de Pollo , Pollos/crecimiento & desarrollo , Hormonas Tiroideas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA