Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Neurobiol Dis ; 199: 106572, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-38901782

RESUMEN

Within the adult mouse subventricular zone (SVZ), neural stem cells (NSCs) produce neuroblasts and oligodendrocyte precursor cells (OPCs). T3, the active thyroid hormone, influences renewal and commitment of SVZ progenitors. However, how regulators of T3 availability affect these processes is less understood. Using Mct8/Dio2 knockout mice, we investigated the role of MCT8, a TH transporter, and DIO2, the T3-generating enzyme, in regulating adult SVZ-neurogliogenesis. Single-cell RNA-Seq revealed Mct8 expression in various SVZ cell types in WT mice, while Dio2 was enriched in neurons, astrocytes, and quiescent NSCs. The absence of both regulators in the knockout model dysregulated gene expression, increased the neuroblast/OPC ratio and hindered OPC differentiation. Immunostainings demonstrated compromised neuroblast migration reducing their supply to the olfactory bulbs, impairing interneuron differentiation and odor discrimination. These findings underscore the pivotal roles of MCT8 and DIO2 in neuro- and oligodendrogenesis, offering targets for therapeutic avenues in neurodegenerative and demyelinating diseases.


Asunto(s)
Ventrículos Laterales , Ratones Noqueados , Transportadores de Ácidos Monocarboxílicos , Células-Madre Neurales , Neurogénesis , Animales , Ratones , Diferenciación Celular/fisiología , Yoduro Peroxidasa/genética , Yoduro Peroxidasa/metabolismo , Yodotironina Deyodinasa Tipo II , Ventrículos Laterales/metabolismo , Ratones Endogámicos C57BL , Transportadores de Ácidos Monocarboxílicos/genética , Transportadores de Ácidos Monocarboxílicos/metabolismo , Células-Madre Neurales/metabolismo , Neurogénesis/fisiología , Bulbo Olfatorio/metabolismo , Células Precursoras de Oligodendrocitos/metabolismo , Simportadores/genética , Simportadores/metabolismo
2.
Neurobiol Dis ; 200: 106621, 2024 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-39097035

RESUMEN

Allan-Herndon-Dudley syndrome (AHDS) is a rare X-linked disorder that causes severe neurological damage, for which there is no effective treatment. AHDS is due to inactivating mutations in the thyroid hormone transporter MCT8 that impair the entry of thyroid hormones into the brain, resulting in cerebral hypothyroidism. However, the pathophysiology of AHDS is still not fully understood and this is essential to develop therapeutic strategies. Based on evidence suggesting that thyroid hormone deficit leads to alterations in astroglial cells, including gliosis, in this work, we have evaluated astroglial impairments in MCT8 deficiency by means of magnetic resonance imaging, histological, ultrastructural, and immunohistochemical techniques, and by mining available RNA sequencing outputs. Apparent diffusion coefficient (ADC) imaging values obtained from magnetic resonance imaging showed changes indicative of alterations in brain cytoarchitecture in MCT8-deficient patients (n = 11) compared to control subjects (n = 11). Astroglial alterations were confirmed by immunohistochemistry against astroglial markers in autopsy brain samples of an 11-year-old and a 30th gestational week MCT8-deficient subjects in comparison to brain samples from control subjects at similar ages. These findings were validated and further explored in a mouse model of AHDS. Our findings confirm changes in all the astroglial populations of the cerebral cortex in MCT8 deficiency that impact astrocytic metabolic and mitochondrial cellular respiration functions. These impairments arise early in brain development and persist at adult stages, revealing an abnormal distribution, density, morphology of cortical astrocytes, along with altered transcriptome, compatible with an astrogliosis-like phenotype at adult stages. We conclude that astrocytes are potential novel therapeutic targets in AHDS, and we propose ADC imaging as a tool to monitor the progression of neurological impairments and potential effects of treatments in MCT8 deficiency.


Asunto(s)
Astrocitos , Encéfalo , Transportadores de Ácidos Monocarboxílicos , Hipotonía Muscular , Simportadores , Hormonas Tiroideas , Astrocitos/metabolismo , Astrocitos/patología , Animales , Ratones , Humanos , Transportadores de Ácidos Monocarboxílicos/metabolismo , Transportadores de Ácidos Monocarboxílicos/genética , Masculino , Encéfalo/metabolismo , Encéfalo/patología , Hormonas Tiroideas/metabolismo , Niño , Simportadores/metabolismo , Simportadores/genética , Hipotonía Muscular/metabolismo , Hipotonía Muscular/genética , Hipotonía Muscular/patología , Discapacidad Intelectual Ligada al Cromosoma X/genética , Discapacidad Intelectual Ligada al Cromosoma X/metabolismo , Discapacidad Intelectual Ligada al Cromosoma X/patología , Femenino , Adulto , Imagen por Resonancia Magnética/métodos , Atrofia Muscular
3.
Int J Mol Sci ; 24(4)2023 Feb 06.
Artículo en Inglés | MEDLINE | ID: mdl-36834621

RESUMEN

Monocarboxylate transporter 8 (MCT8) and organic anion transporter polypeptide 1C1 (OATP1C1) are thyroid hormone (TH) transmembrane transporters that play an important role in the availability of TH for neural cells, allowing their proper development and function. It is important to define which cortical cellular subpopulations express those transporters to explain why MCT8 and OATP1C1 deficiency in humans leads to dramatic alterations in the motor system. By means of immunohistochemistry and double/multiple labeling immunofluorescence in adult human and monkey motor cortices, we demonstrate the presence of both transporters in long-projection pyramidal neurons and in several types of short-projection GABAergic interneurons in both species, suggesting a critical position of these transporters for modulating the efferent motor system. MCT8 is present at the neurovascular unit, but OATP1C1 is only present in some of the large vessels. Both transporters are expressed in astrocytes. OATP1C1 was unexpectedly found, only in the human motor cortex, inside the Corpora amylacea complexes, aggregates linked to substance evacuation towards the subpial system. On the basis of our findings, we propose an etiopathogenic model that emphasizes these transporters' role in controlling excitatory/inhibitory motor cortex circuits in order to understand some of the severe motor disturbances observed in TH transporter deficiency syndromes.


Asunto(s)
Corteza Motora , Transportadores de Anión Orgánico , Simportadores , Adulto , Humanos , Encéfalo/metabolismo , Interneuronas/metabolismo , Transportadores de Ácidos Monocarboxílicos/metabolismo , Corteza Motora/metabolismo , Transportadores de Anión Orgánico/metabolismo , Péptidos , Células Piramidales/metabolismo , Hormonas Tiroideas
4.
Int J Mol Sci ; 24(11)2023 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-37298594

RESUMEN

Monocarboxylate transporter 8 (MCT8) and organic anion-transporting polypeptide 1C1 (OATP1C1) are thyroid hormone (TH) transmembrane transporters relevant for the availability of TH in neural cells, crucial for their proper development and function. Mutations in MCT8 or OATP1C1 result in severe disorders with dramatic movement disability related to alterations in basal ganglia motor circuits. Mapping the expression of MCT8/OATP1C1 in those circuits is necessary to explain their involvement in motor control. We studied the distribution of both transporters in the neuronal subpopulations that configure the direct and indirect basal ganglia motor circuits using immunohistochemistry and double/multiple labeling immunofluorescence for TH transporters and neuronal biomarkers. We found their expression in the medium-sized spiny neurons of the striatum (the receptor neurons of the corticostriatal pathway) and in various types of its local microcircuitry interneurons, including the cholinergic. We also demonstrate the presence of both transporters in projection neurons of intrinsic and output nuclei of the basal ganglia, motor thalamus and nucleus basalis of Meynert, suggesting an important role of MCT8/OATP1C1 for modulating the motor system. Our findings suggest that a lack of function of these transporters in the basal ganglia circuits would significantly impact motor system modulation, leading to clinically severe movement impairment.


Asunto(s)
Ganglios Basales , Transportadores de Anión Orgánico , Simportadores , Adulto , Humanos , Ganglios Basales/metabolismo , Encéfalo/metabolismo , Interneuronas/metabolismo , Transportadores de Ácidos Monocarboxílicos/genética , Transportadores de Ácidos Monocarboxílicos/metabolismo , Neuronas/metabolismo , Transportadores de Anión Orgánico/metabolismo , Simportadores/genética , Simportadores/metabolismo , Tálamo/metabolismo , Hormonas Tiroideas/metabolismo
5.
Neurobiol Dis ; 162: 105567, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34838669

RESUMEN

Mutations in the thyroid hormone transporter monocarboxylate transporter 8 (MCT8) lead to profound brain alterations, including myelination impairments, in humans. We aimed to further explore the pathophysiological mechanisms underlying the MCT8 deficiency-associated myelination impairments to unravel new biomarkers and therapeutic targets. We have performed brain histological analysis on an MCT8-deficient subject and histological, ultrastructural, and magnetic resonance imaging (MRI) analysis in the brain of a mouse model of the syndrome, lacking MCT8 and enzyme deiodinase type 2 (DIO2, Mct8/Dio2 KO). We have found that the MCT8-deficient subject presents severely reduced myelin lipid and protein staining and increased proportion of small-caliber myelinated axons in detriment of large-caliber ones. Mct8/Dio2 KO mice present myelination impairments and abnormal oligodendroglial development. We conclude that the greater proportion of small-caliber axons and impairments in the oligodendroglia lineage progression arise as potential mechanisms underlying the permanent myelination defects in MCT8-deficiency. Moreover, we present the Mct8/Dio2 KO mouse model, and MRI as a non-invasive biomarker, as highly valuable tools for preclinical studies involving MCT8 deficiency. These findings contribute to the understanding of the pathological mechanisms in MCT8 deficiency and suggest new biomarkers and therapeutic targets to consider therapeutic options for the neurological defects in patients.


Asunto(s)
Transportadores de Ácidos Monocarboxílicos , Simportadores , Animales , Axones/metabolismo , Encéfalo/diagnóstico por imagen , Encéfalo/metabolismo , Humanos , Ratones , Transportadores de Ácidos Monocarboxílicos/genética , Transportadores de Ácidos Monocarboxílicos/metabolismo , Simportadores/genética , Simportadores/metabolismo , Hormonas Tiroideas/genética , Hormonas Tiroideas/metabolismo
6.
Neurobiol Dis ; 174: 105896, 2022 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-36243247

RESUMEN

Inactivating mutations in the specific thyroid hormone transporter monocarboxylate transporter 8 (MCT8) lead to an X-linked rare disease named MCT8 deficiency or Allan-Herndon-Dudley Syndrome. Patients exhibit a plethora of severe endocrine and neurological alterations, with no effective treatment for the neurological symptoms. An optimal mammalian model is essential to explore the pathological mechanisms and potential therapeutic approaches. Here we have generated by CRISPR/Cas9 an avatar mouse model for MCT8 deficiency with a point mutation found in two MCT8-deficient patients (P253L mice). We have predicted by in silico studies that this mutation alters the substrate binding pocket being the probable cause for impairing thyroid hormone transport. We have characterized the phenotype of MCT8-P253L mice and found endocrine alterations similar to those described in patients and in MCT8-deficient mice. Importantly, we detected brain hypothyroidism, structural and functional neurological alterations resembling the patient's neurological impairments. Thus, the P253L mouse provides a valuable model for studying the pathophysiology of MCT8 deficiency and in the future will allow to test therapeutic alternatives such as in vivo gene therapy and pharmacological chaperone therapy to improve the neurological impairments in MCT8 deficiency.


Asunto(s)
Transportadores de Ácidos Monocarboxílicos , Simportadores , Animales , Ratones , Transportadores de Ácidos Monocarboxílicos/genética , Transportadores de Ácidos Monocarboxílicos/metabolismo , Simportadores/genética , Simportadores/metabolismo , Sistemas CRISPR-Cas , Hormonas Tiroideas/metabolismo , Modelos Animales de Enfermedad , Mamíferos/metabolismo
7.
Cereb Cortex ; 28(5): 1783-1793, 2018 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-28407057

RESUMEN

Thyroid hormones (THs, T4 and the transcriptionally active hormone T3) play an essential role in neurodevelopment; however, the mechanisms underlying T3 brain delivery during mice fetal development are not well known. This work has explored the sources of brain T3 during mice fetal development using biochemical, anatomical, and molecular approaches. The findings revealed that during late gestation, a large amount of fetal brain T4 is of maternal origin. Also, in the developing mouse brain, fetal T3 content is regulated through the conversion of T4 into T3 by type-2 deiodinase (D2) activity, which is present from earlier prenatal stages. Additionally, D2 activity was found to be essential to mediate expression of T3-dependent genes in the cerebral cortex, and also necessary to generate the transient cerebral cortex hyperthyroidism present in mice lacking the TH transporter Monocarboxylate transporter 8. Notably, the gene encoding for D2 (Dio2) was mainly expressed at the blood-cerebrospinal fluid barrier (BCSFB). Overall, these data signify that T4 deiodinated by D2 may be the only source of T3 during neocortical development. We therefore propose that D2 activity at the BCSFB converts the T4 transported across the choroid plexus into T3, thus supplying the brain with active hormone to maintain TH homeostasis.


Asunto(s)
Corteza Cerebral , Embrión de Mamíferos/efectos de los fármacos , Embrión de Mamíferos/metabolismo , Regulación del Desarrollo de la Expresión Génica/fisiología , Hormonas Tiroideas/metabolismo , Factores de Edad , Animales , Animales Recién Nacidos , Peso Corporal/fisiología , Corteza Cerebral/embriología , Corteza Cerebral/crecimiento & desarrollo , Corteza Cerebral/metabolismo , Femenino , Edad Gestacional , Yoduro Peroxidasa/deficiencia , Yoduro Peroxidasa/genética , Isótopos de Yodo/metabolismo , Hígado/embriología , Hígado/crecimiento & desarrollo , Hígado/metabolismo , Proteínas de Transporte de Membrana/deficiencia , Proteínas de Transporte de Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Transportadores de Ácidos Monocarboxílicos , Embarazo , ARN Mensajero/metabolismo , Simportadores , Hormonas Tiroideas/farmacología , Tirosina 3-Monooxigenasa/metabolismo , Yodotironina Deyodinasa Tipo II
8.
Cereb Cortex ; 27(5): 2809-2819, 2017 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-27166167

RESUMEN

TaqIA is a polymorphism associated with addictions and dopamine-related traits. It is located in the ankyrin repeat and kinase domain containing 1 gene (ANKK1) nearby the gene for the dopamine D2 receptor (D2R). Since ANKK1 function is unknown, TaqIA-associated traits have been explained only by differences in D2R. Here we report ANKK1 studies in mouse and human brain using quantitative real-time PCR, Western blot, immunohistochemistry, and flow cytometry. ANKK1 mRNA and protein isoforms vary along neurodevelopment in the human and mouse brain. In mouse adult brain ANKK1 is located in astrocytes, nuclei of postmitotic neurons and neural precursors from neurogenic niches. In both embryos and adults, nuclei of neural precursors show significant variation of ANKK1 intensity. We demonstrate a correlation between ANKK1 and the cell cycle. Cell synchronization experiments showed a significant increment of ANKK1-kinase in mitotic cells while ANKK1-kinase overexpression affects G1 and M phase that were found to be modulated by ANKK1 alleles and apomorphine treatment. Furthermore, during embryonic neurogenesis ANKK1 was expressed in slow-dividing neuroblasts and rapidly dividing precursors which are mitotic cells. These results suggest a role of ANKK1 during the cell cycle in neural precursors thus providing biological support to brain structure involvement in the TaqIA-associated phenotypes.


Asunto(s)
Encéfalo/metabolismo , Ciclo Celular/fisiología , Regulación del Desarrollo de la Expresión Génica/genética , Células-Madre Neurales/fisiología , Proteínas Serina-Treonina Quinasas/metabolismo , Adolescente , Factores de Edad , Animales , Animales Recién Nacidos , Encéfalo/embriología , Encéfalo/crecimiento & desarrollo , Diferenciación Celular/fisiología , Línea Celular Tumoral , Embrión de Mamíferos , Feto , Edad Gestacional , Proteína Ácida Fibrilar de la Glía/metabolismo , Humanos , Lactante , Ratones , Persona de Mediana Edad , Neurogénesis/fisiología , Proteínas Serina-Treonina Quinasas/genética , ARN Mensajero/metabolismo , Tubulina (Proteína)/genética , Tubulina (Proteína)/metabolismo
9.
Front Endocrinol (Lausanne) ; 15: 1347802, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38516412

RESUMEN

Over the last decades, thyroid hormones (THs) signaling has been established as a key signaling cue for the proper maintenance of brain functions in adult mammals, including humans. One of the most fascinating roles of THs in the mature mammalian brain is their ability to regulate adult neurogliogenic processes. In this respect, THs control the generation of new neuronal and glial progenitors from neural stem cells (NSCs) as well as their final differentiation and maturation programs. In this review, we summarize current knowledge on the cellular organization of adult rodent neurogliogenic niches encompassing well-established niches in the subventricular zone (SVZ) lining the lateral ventricles, the hippocampal subgranular zone (SGZ), and the hypothalamus, but also less characterized niches in the striatum and the cerebral cortex. We then discuss critical questions regarding how THs availability is regulated in the respective niches in rodents and larger mammals as well as how modulating THs availability in those niches interferes with lineage decision and progression at the molecular, cellular, and functional levels. Based on those alterations, we explore the novel therapeutic avenues aiming at harnessing THs regulatory influences on neurogliogenic output to stimulate repair processes by influencing the generation of either new neurons (i.e. Alzheimer's, Parkinson's diseases), oligodendrocytes (multiple sclerosis) or both (stroke). Finally, we point out future challenges, which will shape research in this exciting field in the upcoming years.


Asunto(s)
Encéfalo , Neurogénesis , Humanos , Adulto , Animales , Neurogénesis/fisiología , Encéfalo/fisiología , Neuronas/fisiología , Hormonas Tiroideas/fisiología , Mamíferos
10.
Eur Thyroid J ; 13(2)2024 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-38417253

RESUMEN

Thyroid hormones play an important role during the development and functioning of the different sensory systems. In order to exert their actions, thyroid hormones need to access their target cells through transmembrane transporter proteins, among which the monocarboxylate transporter 8 (MCT8) stands out for its pathophysiological relevance. Mutations in the gene encoding for MCT8 lead to the Allan-Herndon-Dudley syndrome (AHDS), a rare disease characterised by severe neuromotor and cognitive impairments. The impact of MCT8 deficiency in the neurosensory capacity of AHDS patients is less clear, with only a few patients displaying visual and auditory impairments. In this review we aim to gather data from different animal models regarding thyroid hormone transport and action in the different neurosensory systems that could aid to identify potential neurosensorial alterations in MCT8-deficient patients.


Asunto(s)
Discapacidad Intelectual Ligada al Cromosoma X , Atrofia Muscular , Hormonas Tiroideas , Animales , Humanos , Hormonas Tiroideas/metabolismo , Discapacidad Intelectual Ligada al Cromosoma X/genética , Transporte Biológico , Hipotonía Muscular/genética , Transportadores de Ácidos Monocarboxílicos/genética
11.
Thyroid ; 33(4): 501-510, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36565029

RESUMEN

Introduction: Patients lacking functional monocarboxylate transporter 8 (MCT8), a highly specific thyroid hormone (TH) transporter, present severe psychomotor disabilities. MCT8 deficiency leads to peripheral hyperthyroidism and brain hypothyroidism, the latter due to impaired transport of TH across brain barriers. Available treatments for patients are limited and aim to overcome the limited TH transport across brain barriers. The use of TH analogues such as 3,3',5-triiodothyroacetic acid (TRIAC) that do not require MCT8 to cross the cellular membranes is considered a potential therapy for MCT8 deficiency. Previous studies have shown that systemic administration of TRIAC at therapeutic doses does not increase TRIAC content in the brain, while intracerebroventricular (ICV) administration of therapeutic doses of TRIAC increases TRIAC content in the brain but does not mediate thyromimetic effects. In view of this, we hypothesize that ICV administration of high doses of TRIAC can mediate thyromimetic effects in the brain without worsening the brain hypothyroidism or peripheral hyperthyroidism of patients. Methods: We administered 400 ng/g of body weight per day of ICV TRIAC in a mouse model of MCT8 deficiency: Mct8-/y and deiodinase 2 (Dio2)-/- double knockout mice. The effects of this treatment on TH and TRIAC levels/content in blood and tissues were determined by radioimmunoassay and effects on TH-regulated genes were assessed by real time-quantitative polymerase chain reaction in peripheral and central tissues. Results: ICV administration of high doses of TRIAC ameliorated the peripheral hyperthyroidism. In the brain, this treatment did not further aggravate brain hypothyroidism and increased TRIAC content in several brain regions; however, only moderate thyromimetic activity was observed in restricted brain areas. Conclusion: Administration of high doses of TRIAC by ICV delivery at juvenile stages in a mouse model of MCT8 deficiency is effective in normalizing peripheral hyperthyroidism but exerts minimal thyromimetic activity in the brain.


Asunto(s)
Hipertiroidismo , Hipotiroidismo , Simportadores , Animales , Ratones , Simportadores/genética , Triyodotironina , Hormonas Tiroideas , Encéfalo , Hipertiroidismo/tratamiento farmacológico , Hipotiroidismo/tratamiento farmacológico , Ratones Noqueados , Modelos Animales de Enfermedad , Transportadores de Ácidos Monocarboxílicos/genética
12.
Thyroid ; 33(5): 632-640, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36792926

RESUMEN

Background: Monocarboxylate transporter 8 (MCT8) deficiency is a rare X-linked disease where patients exhibit peripheral hyperthyroidism and cerebral hypothyroidism, which results in severe neurological impairments. These brain defects arise from a lack of thyroid hormones (TH) during critical stages of human brain development. Treatment options for MCT8-deficient patients are limited and none have been able to prevent or ameliorate effectively the neurological impairments. This study explored the effects of the TH agonist sobetirome and its CNS-selective amide prodrug, Sob-AM2, in the treatment of pregnant dams carrying fetuses lacking Mct8 and deiodinase type 2 (Mct8/Dio2 KO), as a murine model for MCT8 deficiency. Methods: Pregnant dams carrying Mct8/Dio2 KO fetuses were treated with 1 mg of sobetirome/kg body weight/day, or 0.3 mg of Sob-AM2/kg body weight/day for 7 days, starting at embryonic day 12.5 (E12.5). As controls, pregnant dams carrying wild-type and pregnant dams carrying Mct8/Dio2 KO fetuses were treated with daily subcutaneous injections of vehicle. Dams TH levels were measured by enzyme-linked immunosorbent assay (ELISA). Samples were extracted at E18.5 and the effect of treatments on the expression of triiodothyronine (T3)-dependent genes was measured in the placenta, fetal liver, and fetal cerebral cortex by real-time polymerase chain reaction. Results: Maternal sobetirome treatment led to spontaneous abortions. Sob-AM2 treatment, however, was able to cross the placental as well as the brain barriers and exert thyromimetic effects in Mct8/Dio2 KO fetal tissues. Sob-AM2 treatment did not affect the expression of the T3-target genes analyzed in the placenta, but it mediated thyromimetic effects in the fetal liver by increasing the expression of Dio1 and Dio3 genes. Interestingly, Sob-AM2 treatment increased the expression of several T3-dependent genes in the brain such as Hr, Shh, Dio3, Kcnj10, Klf9, and Faah in Mct8/Dio2 KO fetuses. Conclusions: Maternal administration of Sob-AM2 can cross the placental barrier and access the fetal tissues, including the brain, in the absence of MCT8, to exert thyromimetic actions by modulating the expression of T3-dependent genes. Therefore, Sob-AM2 has the potential to address the cerebral hypothyroidism characteristic of MCT8 deficiency from fetal stages and to prevent neurodevelopmental alterations in the MCT8-deficient fetal brain.


Asunto(s)
Hipotiroidismo , Profármacos , Simportadores , Animales , Humanos , Ratones , Femenino , Embarazo , Tiroxina/farmacología , Tiroxina/metabolismo , Simportadores/genética , Simportadores/metabolismo , Placenta/metabolismo , Transportadores de Ácidos Monocarboxílicos/genética , Transportadores de Ácidos Monocarboxílicos/metabolismo , Hormonas Tiroideas/farmacología , Hormonas Tiroideas/metabolismo , Triyodotironina/farmacología , Triyodotironina/metabolismo , Hipotiroidismo/metabolismo , Feto/metabolismo , Factores de Transcripción de Tipo Kruppel/metabolismo
13.
Fluids Barriers CNS ; 20(1): 79, 2023 Nov 03.
Artículo en Inglés | MEDLINE | ID: mdl-37924081

RESUMEN

BACKGROUND: The monocarboxylate transporter 8 (MCT8) plays a vital role in maintaining brain thyroid hormone homeostasis. This transmembrane transporter is expressed at the brain barriers, as the blood-brain barrier (BBB), and in neural cells, being the sole known thyroid hormone-specific transporter to date. Inactivating mutations in the MCT8 gene (SLC16A2) cause the Allan-Herndon-Dudley Syndrome (AHDS) or MCT8 deficiency, a rare X-linked disease characterized by delayed neurodevelopment and severe psychomotor disorders. The underlying pathophysiological mechanisms of AHDS remain unclear, and no effective treatments are available for the neurological symptoms of the disease. METHODS: Neurovascular unit ultrastructure was studied by means of transmission electron microscopy. BBB permeability and integrity were evaluated by immunohistochemistry, non-permeable dye infiltration assays and histological staining techniques. Brain blood-vessel density was evaluated by immunofluorescence and magnetic resonance angiography. Finally, angiogenic-related factors expression was evaluated by qRT-PCR. The studies were carried out both in an MCT8 deficient subject and Mct8/Dio2KO mice, an AHDS murine model, and their respective controls. RESULTS: Ultrastructural analysis of the BBB of Mct8/Dio2KO mice revealed significant alterations in neurovascular unit integrity and increased transcytotic flux. We also found functional alterations in the BBB permeability, as shown by an increased presence of peripheral IgG, Sodium Fluorescein and Evans Blue, along with increased brain microhemorrhages. We also observed alterations in the angiogenic process, with reduced blood vessel density in adult mice brain and altered expression of angiogenesis-related factors during brain development. Similarly, AHDS human brain samples showed increased BBB permeability to IgG and decreased blood vessel density. CONCLUSIONS: These findings identify for the first time neurovascular alterations in the MCT8-deficient brain, including a disruption of the integrity of the BBB and alterations in the neurovascular unit ultrastructure as a new pathophysiological mechanism for AHDS. These results open a new field for potential therapeutic targets for the neurological symptoms of these patients and unveils magnetic resonance angiography as a new non-invasive in vivo technique for evaluating the progression of the disease.


Asunto(s)
Discapacidad Intelectual Ligada al Cromosoma X , Simportadores , Animales , Humanos , Ratones , Barrera Hematoencefálica/metabolismo , Inmunoglobulina G , Discapacidad Intelectual Ligada al Cromosoma X/diagnóstico , Discapacidad Intelectual Ligada al Cromosoma X/genética , Discapacidad Intelectual Ligada al Cromosoma X/patología , Transportadores de Ácidos Monocarboxílicos/genética , Transportadores de Ácidos Monocarboxílicos/metabolismo , Hipotonía Muscular/diagnóstico , Hipotonía Muscular/genética , Hipotonía Muscular/metabolismo , Atrofia Muscular/diagnóstico , Atrofia Muscular/genética , Atrofia Muscular/metabolismo , Simportadores/genética , Simportadores/metabolismo , Simportadores/uso terapéutico , Hormonas Tiroideas/metabolismo , Hormonas Tiroideas/uso terapéutico
14.
J Comp Neurol ; 529(1): 234-256, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-30942489

RESUMEN

The cold- and menthol-activated ion channel transient receptor potential channel subfamily M member 8 (TRPM8) is the principal detector of environmental cold in mammalian sensory nerve endings. Although it is mainly expressed in a subpopulation of peripheral sensory neurons, it has also been identified in non-neuronal tissues. Here, we show, by in situ hybridization (ISH) and by the analysis of transgenic reporter expression in two different reporter mouse strains, that TRPM8 is also expressed in the central nervous system. Although it is present at much lower levels than in peripheral sensory neurons, we found cells expressing TRPM8 in restricted areas of the brain, especially in the hypothalamus, septum, thalamic reticular nucleus, certain cortices and other limbic structures, as well as in some specific nuclei in the brainstem. Interestingly, positive fibers were also found traveling through the major limbic tracts, suggesting a role of TRPM8-expressing central neurons in multiple aspects of thermal regulation, including autonomic and behavioral thermoregulation. Additional ISH experiments in rat brain demonstrated a conserved pattern of expression of this ion channel between rodent species. We confirmed the functional activity of this channel in the mouse brain using electrophysiological patch-clamp recordings of septal neurons. These results open a new window in TRPM8 physiology, guiding further efforts to understand potential roles of this molecular sensor within the brain.


Asunto(s)
Regulación de la Temperatura Corporal/fisiología , Encéfalo/metabolismo , Frío , Red Nerviosa/metabolismo , Canales Catiónicos TRPM/biosíntesis , Animales , Frío/efectos adversos , Femenino , Expresión Génica , Masculino , Ratones , Ratones Endogámicos C57BL , Ratas , Ratas Sprague-Dawley , Ratas Transgénicas , Canales Catiónicos TRPM/genética
15.
Front Neurosci ; 14: 380, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32410949

RESUMEN

Allan-Herndon-Dudley syndrome is a rare disease caused by inactivating mutations in the SLC16A2 gene, which encodes the monocarboxylate transporter 8 (MCT8), a transmembrane transporter specific for thyroid hormones (T3 and T4). Lack of MCT8 function produces serious neurological disturbances, most likely due to impaired transport of thyroid hormones across brain barriers during development resulting in severe brain hypothyroidism. Patients also suffer from thyrotoxicity in other organs due to the presence of a high concentration of T3 in the serum. An effective therapeutic strategy should restore thyroid hormone serum levels (both T3 and T4) and should address MCT8 transporter deficiency in brain barriers and neural cells, to enable the access of thyroid hormones to target neural cells. Unfortunately, targeted therapeutic options are currently scarce and their effect is limited to an improvement in the thyrotoxic state, with no sign of any neurological improvement. The use of thyroid hormone analogs such as TRIAC, DITPA, or sobetirome, that do not require MCT8 to cross cell membranes and whose controlled thyromimetic activity could potentially restore the normal function of the affected organs, are being explored to improve the cerebral availability of these analogs. Other strategies aiming to restore the transport of THs through MCT8 at the brain barriers and the cellular membranes include gene replacement therapy and the use of pharmacological chaperones. The design of an appropriate therapeutic strategy in combination with an early diagnosis (at prenatal stages), will be key aspects to improve the devastating alterations present in these patients.

16.
PLoS One ; 15(7): e0236113, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32687511

RESUMEN

Loss of function mutations in the gene encoding the thyroid hormone transporter monocarboxylate transporter 8 (MCT8) lead to severe neurodevelopmental defects in humans associated with a specific thyroid hormone phenotype manifesting high serum 3,5,3'-triiodothyronine (T3) and low thyroxine (T4) levels. Patients present a paradoxical state of peripheral hyperthyroidism and brain hypothyroidism, this last one most likely arising from impaired thyroid hormone transport across the brain barriers. The administration of thyroid hormones by delivery pathways that bypass the brain barriers, such as the intranasal delivery route, offers the possibility to improve the neurological defects of MCT8-deficient patients. In this study, the thyroid hormones T4 and T3 were administrated intranasally in different mouse models of MCT8 deficiency. We have found that, under the present formulation, intranasal administration of thyroid hormones does not increase the content of thyroid hormones in the brain and further raises the peripheral thyroid hormone levels. Our data suggests intranasal delivery of thyroid hormones is not a suitable therapeutic strategy for MCT8 deficiency, although alternative formulations could be considered in the future to improve the nose-to-brain transport.


Asunto(s)
Transportadores de Ácidos Monocarboxílicos/deficiencia , Simportadores/deficiencia , Hormonas Tiroideas/administración & dosificación , Hormonas Tiroideas/farmacología , Administración Intranasal , Animales , Encéfalo/citología , Ratones , Transportadores de Ácidos Monocarboxílicos/genética , Mutación , Transducción de Señal/efectos de los fármacos , Simportadores/genética , Hormonas Tiroideas/sangre
17.
Mol Metab ; 42: 101097, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33049408

RESUMEN

OBJECTIVE: Metainflammation is a chronic low-grade inflammatory state induced by obesity and associated comorbidities, including peripheral insulin resistance. Brown adipose tissue (BAT), a therapeutic target against obesity, is an insulin target tissue sensitive to inflammation. Therefore, it is necessary to find strategies to protect BAT against the effects of inflammation in energy balance. In this study, we explored the impact of moderate sirtuin 1 (SIRT1) overexpression on insulin sensitivity and ß-adrenergic responses in BAT and brown adipocytes (BA) under pro-inflammatory conditions. METHODS: The effect of inflammation on BAT functionality was studied in obese db/db mice and lean wild-type (WT) mice or mice with moderate overexpression of SIRT1 (SIRT1Tg+) injected with a low dose of bacterial lipopolysaccharide (LPS) to mimic endotoxemia. We also conducted studies on differentiated BA (BA-WT and BA-SIRT1Tg+) exposed to a macrophage-derived pro-inflammatory conditioned medium (CM) to evaluate the protection of SIRT1 overexpression in insulin signaling and glucose uptake, mitochondrial respiration, fatty acid oxidation (FAO), and norepinephrine (NE)-mediated-modulation of uncoupling protein-1 (UCP-1) expression. RESULTS: BAT from the db/db mice was susceptible to metabolic inflammation manifested by the activation of pro-inflammatory signaling cascades, increased pro-inflammatory gene expression, tissue-specific insulin resistance, and reduced UCP-1 expression. Impairment of insulin and noradrenergic responses were also found in the lean WT mice upon LPS injection. In contrast, BAT from the mice with moderate overexpression of SIRT1 (SIRT1Tg+) was protected against LPS-induced activation of pro-inflammatory signaling, insulin resistance, and defective thermogenic-related responses upon cold exposure. Importantly, the decline in triiodothyronine (T3) levels in the circulation and intra-BAT after exposure of the WT mice to LPS and cold was markedly attenuated in the SIRT1Tg+ mice. In vitro BA experiments in the two genotypes revealed that upon differentiation with a T3-enriched medium and subsequent exposure to a macrophage-derived pro-inflammatory CM, only BA-SIRT1Tg+ fully recovered insulin and noradrenergic responses. CONCLUSIONS: This study has ascertained the benefit of the moderate overexpression of SIRT1 to confer protection against defective insulin and ß-adrenergic responses caused by BAT inflammation. Our results have potential therapeutic value in combinatorial therapies for BAT-specific thyromimetics and SIRT1 activators to combat metainflammation in this tissue.


Asunto(s)
Tejido Adiposo Pardo/metabolismo , Sirtuina 1/metabolismo , Adipocitos/metabolismo , Adipocitos/fisiología , Adipocitos Marrones/metabolismo , Adipocitos Marrones/fisiología , Tejido Adiposo/metabolismo , Tejido Adiposo Pardo/fisiología , Animales , Metabolismo Energético , Femenino , Expresión Génica/genética , Regulación de la Expresión Génica/genética , Inflamación/prevención & control , Insulina/metabolismo , Resistencia a la Insulina/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Mitocondrias/metabolismo , Obesidad/metabolismo , Receptores Adrenérgicos beta/metabolismo , Sirtuina 1/genética , Sirtuina 1/fisiología , Termogénesis/efectos de los fármacos , Proteína Desacopladora 1/metabolismo
18.
Eur J Neurosci ; 30(4): 679-92, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19686470

RESUMEN

Activity-dependent changes taking place at the hippocampal perforant pathway-dentate gyrus synapse during classical eyeblink conditioning were recorded in adult thyroidectomized (hypothyroid) and control (euthyroid) rats, and in animals treated with thyroid hormones 20 days after thyroidectomy (recovery rats). The aim was to determine the contribution of thyroid hormones and the consequences of adult-onset hypothyroidism to both associative learning and the physiological potentiation of hippocampal synapses during the actual learning process in alert behaving animals. Control and recovery rats presented similar learning curves, whereas hypothyroid animals presented lower values. A single pulse presented to the perforant pathway during the conditioned-unconditioned inter-stimulus interval evoked a monosynaptic field excitatory postsynaptic potential in dentate granule cells (whose slope was linearly related to the rate of acquisition in the control group), but not in hypothyroid and recovery animals. Input-output relationships and long-term potentiation evoked by train stimulation of the perforant pathway were significantly depressed in hypothyroid animals. Thyroid hormone treatment failed to normalize these two neurophysiological abnormalities observed in hypothyroid animals. In contrast, paired-pulse facilitation was not affected by thyroidectomy. The results indicate that thyroid hormone treatment after a short period of adult hypothyroidism helps to restore some hippocampally dependent functions, such as classical conditioning, but not other hippocampal properties, such as the synaptic plasticity evoked during associative learning and during experimentally induced long-term potentiation. The present results have important clinical implications for the handling of patients with adult-onset thyroid diseases.


Asunto(s)
Condicionamiento Palpebral/efectos de los fármacos , Hipocampo/efectos de los fármacos , Plasticidad Neuronal/efectos de los fármacos , Neuronas/efectos de los fármacos , Potenciales Sinápticos/efectos de los fármacos , Hormonas Tiroideas/farmacología , Estimulación Acústica , Análisis de Varianza , Animales , Condicionamiento Palpebral/fisiología , Estimulación Eléctrica , Electrodos Implantados , Electromiografía , Proteínas Transportadoras de GABA en la Membrana Plasmática/metabolismo , Hipocampo/metabolismo , Hipocampo/fisiología , Inmunohistoquímica , Hígado/química , Potenciación a Largo Plazo/efectos de los fármacos , Potenciación a Largo Plazo/fisiología , Masculino , Plasticidad Neuronal/fisiología , Neuronas/metabolismo , Neuronas/fisiología , Parvalbúminas/metabolismo , Ratas , Ratas Wistar , Receptores de N-Metil-D-Aspartato/metabolismo , Procesamiento de Señales Asistido por Computador , Sinapsis/efectos de los fármacos , Sinapsis/fisiología , Potenciales Sinápticos/fisiología , Tiroidectomía , Tiroxina/análisis
19.
Mol Cell Biol ; 26(6): 2317-26, 2006 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16508007

RESUMEN

dickkopf (dkk) genes encode a small family of secreted Wnt antagonists, except for dkk3, which is divergent and whose function is poorly understood. Here, we describe the generation and characterization of dkk3 mutant mice. dkk3-deficient mice are viable and fertile. Phenotypic analysis shows no major alterations in organ morphology, physiology, and most clinical chemistry parameters. Since Dkk3 was proposed to function as thyroid hormone binding protein, we have analyzed deiodinase activities, as well as thyroid hormone levels. Mutant mice are euthyroid, and the data do not support a relationship of dkk3 with thyroid hormone metabolism. Altered phenotypes in dkk3 mutant mice were observed in the frequency of NK cells, immunoglobulin M, hemoglobin, and hematocrit levels, as well as lung ventilation. Furthermore, dkk3-deficient mice display hyperactivity.


Asunto(s)
Conducta Animal/fisiología , Sistema Inmunológico/fisiología , Péptidos y Proteínas de Señalización Intercelular/genética , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Ventilación Pulmonar/genética , Proteínas Adaptadoras Transductoras de Señales , Animales , Antígenos Helmínticos/genética , Antígenos Helmínticos/inmunología , Eritrocitos/patología , Femenino , Inmunoglobulina M/sangre , Péptidos y Proteínas de Señalización Intercelular/inmunología , Yoduro Peroxidasa/metabolismo , Pulmón/fisiopatología , Masculino , Ratones , Ratones Mutantes , Tiroxina/metabolismo , Triyodotironina/metabolismo , Yodotironina Deyodinasa Tipo II
20.
PLoS One ; 14(12): e0226017, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31809508

RESUMEN

Patients lacking the thyroid hormone (TH) transporter MCT8 present abnormal serum levels of TH: low thyroxine and high triiodothyronine. They also have severe neurodevelopmental defects resulting from cerebral hypothyroidism, most likely due to impaired TH transport across the brain barriers. The use of TH analogs, such as triiodothyroacetic acid (TRIAC), that can potentially access the brain in the absence of MCT8 and restore at least a subset of cerebral TH actions could improve the neurological defects in these patients. We hypothesized that direct administration of TRIAC into the brain by intracerebroventricular delivery to mice lacking MCT8 could bypass the restriction at the brain barriers and mediate TH action without causing hypermetabolism. We found that intracerebroventricular administration of therapeutic doses of TRIAC does not increase further plasma triiodothyronine or further decrease plasma thyroxine levels and does not alter TH content in the cerebral cortex. Although TRIAC content increased in the brain, it did not induce TH-mediated actions on selected target genes. Our data suggest that intracerebroventricular delivery of TRIAC has the ability to target the brain in the absence of MCT8 and should be further investigated to address its potential therapeutic use in MCT8 deficiency.


Asunto(s)
Corteza Cerebral/metabolismo , Transportadores de Ácidos Monocarboxílicos/genética , Simportadores/genética , Hormonas Tiroideas/metabolismo , Triyodotironina/análogos & derivados , Animales , Femenino , Infusiones Intraventriculares , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Transportadores de Ácidos Monocarboxílicos/deficiencia , Simportadores/deficiencia , Hormonas Tiroideas/química , Tiroxina/sangre , Triyodotironina/administración & dosificación , Triyodotironina/sangre
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA