Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 107
Filtrar
1.
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
2.
Am J Hum Genet ; 109(3): 518-532, 2022 03 03.
Artículo en Inglés | MEDLINE | ID: mdl-35108495

RESUMEN

Cell adhesion molecules are membrane-bound proteins predominantly expressed in the central nervous system along principal axonal pathways with key roles in nervous system development, neural cell differentiation and migration, axonal growth and guidance, myelination, and synapse formation. Here, we describe ten affected individuals with bi-allelic variants in the neuronal cell adhesion molecule NRCAM that lead to a neurodevelopmental syndrome of varying severity; the individuals are from eight families. This syndrome is characterized by developmental delay/intellectual disability, hypotonia, peripheral neuropathy, and/or spasticity. Computational analyses of NRCAM variants, many of which cluster in the third fibronectin type III (Fn-III) domain, strongly suggest a deleterious effect on NRCAM structure and function, including possible disruption of its interactions with other proteins. These findings are corroborated by previous in vitro studies of murine Nrcam-deficient cells, revealing abnormal neurite outgrowth, synaptogenesis, and formation of nodes of Ranvier on myelinated axons. Our studies on zebrafish nrcamaΔ mutants lacking the third Fn-III domain revealed that mutant larvae displayed significantly altered swimming behavior compared to wild-type larvae (p < 0.03). Moreover, nrcamaΔ mutants displayed a trend toward increased amounts of α-tubulin fibers in the dorsal telencephalon, demonstrating an alteration in white matter tracts and projections. Taken together, our study provides evidence that NRCAM disruption causes a variable form of a neurodevelopmental disorder and broadens the knowledge on the growing role of the cell adhesion molecule family in the nervous system.


Asunto(s)
Trastornos del Neurodesarrollo , Enfermedades del Sistema Nervioso Periférico , Animales , Axones/metabolismo , Adhesión Celular/genética , Moléculas de Adhesión Celular/genética , Moléculas de Adhesión Celular/metabolismo , Moléculas de Adhesión Celular Neuronal , Humanos , Ratones , Hipotonía Muscular/genética , Hipotonía Muscular/metabolismo , Espasticidad Muscular/metabolismo , Trastornos del Neurodesarrollo/genética , Trastornos del Neurodesarrollo/metabolismo , Pez Cebra/genética , Pez Cebra/metabolismo
3.
J Coll Physicians Surg Pak ; 32(12): SS134-SS136, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36597316

RESUMEN

Pompe disease, also known as Glycogen Storage Disease Type II, is a rare disorder of glucose metabolism caused by congenital acid alpha-glucosidase (GAA) deficiency. A large amount of glycogen accumulates in the lysosomes, causing these to swell and rupture. Its incidence is about 1 in 40,000 to 1 in 50,000 newborns. The main features are hypotonia and cardiomyopathy. Only a few clinical cases of Pompe disease have been reported, and appendicular torsion has rarely been observed. Herein, we report a case of Pompe disease combined with appendicular torsion, both of which were diagnosed on autopsy pathology. The clinical diagnosis of this disease is difficult in developing countries, and it is mostly misdiagnosed as other types of heart disease. Once the clinical symptoms worsen, most of them die within a short period. Therefore, screening for neonatal genetic metabolic diseases for early diagnosis and treatment should be carried out. Key Words: Glycogen storage disease type II, Metabolic disease, Enzyme replacement therapy, Neonatal screening.


Asunto(s)
Cardiomiopatías , Enfermedad del Almacenamiento de Glucógeno Tipo II , Enfermedades del Recién Nacido , Humanos , Recién Nacido , Enfermedad del Almacenamiento de Glucógeno Tipo II/complicaciones , Enfermedad del Almacenamiento de Glucógeno Tipo II/diagnóstico , Enfermedad del Almacenamiento de Glucógeno Tipo II/genética , alfa-Glucosidasas/genética , alfa-Glucosidasas/metabolismo , alfa-Glucosidasas/uso terapéutico , Hipotonía Muscular/tratamiento farmacológico , Hipotonía Muscular/metabolismo , Cardiomiopatías/tratamiento farmacológico , Lisosomas/metabolismo , Lisosomas/patología
4.
Mol Genet Metab ; 135(1): 109-113, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34969638

RESUMEN

BACKGROUND AND OBJECTIVES: MCT8 deficiency is a rare genetic leukoencephalopathy caused by a defect of thyroid hormone transport across cell membranes, particularly through blood brain barrier and into neural cells. It is characterized by a complex neurological presentation, signs of peripheral thyrotoxicosis and cerebral hypothyroidism. Movement disorders (MDs) have been frequently mentioned in this condition, but not systematically studied. METHODS: Each patient recruited was video-recorded during a routine outpatient visit according to a predefined protocol. The presence and the type of MDs were evaluated. The type of MD was blindly scored by two child neurologists experts in inherited white matter diseases and in MD. Dystonia was scored according to Burke-Fahn-Marsden Dystonia Rating Scale (BFMDRS). When more than one MD was present, the predominant one was scored. RESULTS: 27 patients were included through a multicenter collaboration. In many cases we saw a combination of different MDs. Hypokinesia was present in 25/27 patients and was the predominant MD in 19. It was often associated with hypomimia and global hypotonia. Dystonia was observed in 25/27 patients, however, in a minority of cases (5) it was deemed the predominant MD. In eleven patients, exaggerated startle reactions and/or other paroxysmal non-epileptic events were observed. CONCLUSION: MDs are frequent clinical features of MCT8 deficiency, possibly related to the important role of thyroid hormones in brain development and functioning of normal dopaminergic circuits of the basal ganglia. Dystonia is common, but usually mild to moderate in severity, while hypokinesia was the predominant MD in the majority of patients.


Asunto(s)
Discapacidad Intelectual Ligada al Cromosoma X , Trastornos del Movimiento , Simportadores , Humanos , Discapacidad Intelectual Ligada al Cromosoma X/genética , Transportadores de Ácidos Monocarboxílicos/genética , Trastornos del Movimiento/genética , Hipotonía Muscular/complicaciones , Hipotonía Muscular/genética , Hipotonía Muscular/metabolismo , Atrofia Muscular/complicaciones , Atrofia Muscular/genética , Atrofia Muscular/metabolismo , Simportadores/genética
5.
Horm Res Paediatr ; 94(3-4): 81-104, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34091447

RESUMEN

The current differential diagnosis for a short child with low insulin-like growth factor I (IGF-I) and a normal growth hormone (GH) peak in a GH stimulation test (GHST), after exclusion of acquired causes, includes the following disorders: (1) a decreased spontaneous GH secretion in contrast to a normal stimulated GH peak ("GH neurosecretory dysfunction," GHND) and (2) genetic conditions with a normal GH sensitivity (e.g., pathogenic variants of GH1 or GHSR) and (3) GH insensitivity (GHI). We present a critical appraisal of the concept of GHND and the role of 12- or 24-h GH profiles in the selection of children for GH treatment. The mean 24-h GH concentration in healthy children overlaps with that in those with GH deficiency, indicating that the previously proposed cutoff limit (3.0-3.2 µg/L) is too high. The main advantage of performing a GH profile is that it prevents about 20% of false-positive test results of the GHST, while it also detects a low spontaneous GH secretion in children who would be considered GH sufficient based on a stimulation test. However, due to a considerable burden for patients and the health budget, GH profiles are only used in few centres. Regarding genetic causes, there is good evidence of the existence of Kowarski syndrome (due to GH1 variants) but less on the role of GHSR variants. Several genetic causes of (partial) GHI are known (GHR, STAT5B, STAT3, IGF1, IGFALS defects, and Noonan and 3M syndromes), some responding positively to GH therapy. In the final section, we speculate on hypothetical causes.


Asunto(s)
Enanismo Hipofisario , Enanismo , Hormona de Crecimiento Humana/metabolismo , Factor I del Crecimiento Similar a la Insulina/deficiencia , Hipotonía Muscular , Síndrome de Noonan , Columna Vertebral/anomalías , Niño , Preescolar , Diagnóstico Diferencial , Enanismo/diagnóstico , Enanismo/genética , Enanismo/metabolismo , Enanismo Hipofisario/diagnóstico , Enanismo Hipofisario/genética , Enanismo Hipofisario/metabolismo , Hormona de Crecimiento Humana/genética , Humanos , Factor I del Crecimiento Similar a la Insulina/metabolismo , Hipotonía Muscular/diagnóstico , Hipotonía Muscular/genética , Hipotonía Muscular/metabolismo , Síndrome de Noonan/diagnóstico , Síndrome de Noonan/genética , Síndrome de Noonan/metabolismo , Columna Vertebral/metabolismo
6.
Cell Rep ; 35(2): 108963, 2021 04 13.
Artículo en Inglés | MEDLINE | ID: mdl-33852835

RESUMEN

The assembly pathways of mitochondrial respirasome (supercomplex I+III2+IV) are not fully understood. Here, we show that an early sub-complex I assembly, rather than holo-complex I, is sufficient to initiate mitochondrial respirasome assembly. We find that a distal part of the membrane arm of complex I (PD-a module) is a scaffold for the incorporation of complexes III and IV to form a respirasome subcomplex. Depletion of PD-a, rather than other complex I modules, decreases the steady-state levels of complexes III and IV. Both HEK293T cells lacking TIMMDC1 and patient-derived cells with disease-causing mutations in TIMMDC1 showed accumulation of this respirasome subcomplex. This suggests that TIMMDC1, previously known as a complex-I assembly factor, may function as a respirasome assembly factor. Collectively, we provide a detailed, cooperative assembly model in which most complex-I subunits are added to the respirasome subcomplex in the lateral stages of respirasome assembly.


Asunto(s)
Complejo III de Transporte de Electrones/genética , Complejo IV de Transporte de Electrones/genética , Complejo I de Transporte de Electrón/genética , Mitocondrias/genética , Proteínas del Complejo de Importación de Proteínas Precursoras Mitocondriales/genética , Animales , Linfocitos B , Línea Celular Transformada , Discapacidades del Desarrollo/genética , Discapacidades del Desarrollo/metabolismo , Discapacidades del Desarrollo/patología , Complejo I de Transporte de Electrón/antagonistas & inhibidores , Complejo I de Transporte de Electrón/metabolismo , Complejo III de Transporte de Electrones/antagonistas & inhibidores , Complejo III de Transporte de Electrones/metabolismo , Complejo IV de Transporte de Electrones/antagonistas & inhibidores , Complejo IV de Transporte de Electrones/metabolismo , Embrión no Mamífero , Regulación del Desarrollo de la Expresión Génica , Células HEK293 , Humanos , Mitocondrias/metabolismo , Membranas Mitocondriales/metabolismo , Proteínas del Complejo de Importación de Proteínas Precursoras Mitocondriales/deficiencia , Morfolinos/genética , Morfolinos/metabolismo , Hipotonía Muscular/genética , Hipotonía Muscular/metabolismo , Hipotonía Muscular/patología , Fosforilación Oxidativa , Pez Cebra
7.
Hum Mol Genet ; 29(21): 3516-3531, 2021 01 06.
Artículo en Inglés | MEDLINE | ID: mdl-33105479

RESUMEN

Neurodevelopmental disorder with microcephaly, hypotonia and variable brain anomalies (NMIHBA) is an autosomal recessive neurodevelopmental and neurodegenerative disorder characterized by global developmental delay and severe intellectual disability. Microcephaly, progressive cortical atrophy, cerebellar hypoplasia and delayed myelination are neurological hallmarks in affected individuals. NMIHBA is caused by biallelic variants in PRUNE1 encoding prune exopolyphosphatase 1. We provide in-depth clinical description of two affected siblings harboring compound heterozygous variant alleles, c.383G > A (p.Arg128Gln), c.520G > T (p.Gly174*) in PRUNE1. To gain insights into disease biology, we biochemically characterized missense variants within the conserved N-terminal aspartic acid-histidine-histidine (DHH) motif and provide evidence that they result in the destabilization of protein structure and/or loss of exopolyphosphatase activity. Genetic ablation of Prune1 results in midgestational lethality in mice, associated with perturbations to embryonic growth and vascular development. Our findings suggest that NMIHBA results from hypomorphic variant alleles in humans and underscore the potential key role of PRUNE1 exopolyphoshatase activity in neurodevelopment.


Asunto(s)
Ácido Anhídrido Hidrolasas/deficiencia , Discapacidad Intelectual/patología , Microcefalia/patología , Hipotonía Muscular/patología , Mutación , Trastornos del Neurodesarrollo/patología , Monoéster Fosfórico Hidrolasas/genética , Alelos , Animales , Preescolar , Femenino , Humanos , Lactante , Discapacidad Intelectual/etiología , Discapacidad Intelectual/metabolismo , Masculino , Ratones , Microcefalia/etiología , Microcefalia/metabolismo , Hipotonía Muscular/etiología , Hipotonía Muscular/metabolismo , Trastornos del Neurodesarrollo/etiología , Trastornos del Neurodesarrollo/metabolismo , Linaje , Fenotipo
8.
Thyroid ; 31(6): 985-993, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33307956

RESUMEN

Background: The monocarboxylate transporter 8 (Mct8) protein is a primary thyroxine (T4) and triiodothyronine (T3) (thyroid hormone [TH]) transporter. Mutations of the MCT8-encoding, SLC16A2 gene alter thyroid function and TH metabolism and severely impair neurodevelopment (Allan-Herndon-Dudley syndrome [AHDS]). Mct8-deficient mice manifest thyroid alterations but lack neurological signs. It is believed that Mct8 deficiency in mice is compensated by T4 transport through the Slco1c1-encoded organic anion transporter polypeptide 1c1 (Oatp1c1). This allows local brain generation of sufficient T3 by the Dio2-encoded type 2 deiodinase, thus preventing brain hypothyroidism. The Slc16a2/Slco1c1 (MO) and Slc16a2/Dio2 (MD) double knockout (KO) mice lacking T4 and T3 transport, or T3 transport and T4 deiodination, respectively, should be appropriate models of AHDS. Our goal was to compare the cerebral hypothyroidism of systemic hypothyroidism (SH) caused by thyroid gland blockade with that present in the double KO mice. Methods: We performed RNA sequencing by using RNA from the cerebral cortex and striatum of SH mice and the double KO mice on postnatal days 21-23. Real-time polymerase chain reaction was used to confirm RNA-Seq results in replicate biological samples. Cell type involvement was assessed from cell type-enriched genes. Functional genomic differences were analyzed by functional node activity based on a probabilistic graphical model. Results: Each of the three conditions gave a different pattern of gene expression, with partial overlaps. SH gave a wider and highest variation of gene expression than MD or MO. This was partially due to secondary gene responses to hypothyroidism. The set of primary transcriptional T3 targets showed a tighter overlap, but quantitative gene responses indicated that the gene responses in SH were more severe than in MD or MO. Examination of cell type-enriched genes indicated cellular differences between the three conditions. Conclusions: The results indicate that the neurological impairment of AHDS is too severe to be fully explained by TH deprivation only.


Asunto(s)
Encéfalo/metabolismo , Expresión Génica , Hipotiroidismo/genética , Yoduro Peroxidasa/genética , Discapacidad Intelectual Ligada al Cromosoma X/genética , Transportadores de Ácidos Monocarboxílicos/genética , Hipotonía Muscular/genética , Atrofia Muscular/genética , Proteínas de Transporte de Catión Orgánico/genética , Simportadores/genética , Tiroxina/metabolismo , Triyodotironina/metabolismo , Animales , Encéfalo/fisiopatología , Corteza Cerebral/metabolismo , Perfilación de la Expresión Génica , Hipotiroidismo/metabolismo , Hipotiroidismo/fisiopatología , Yoduro Peroxidasa/metabolismo , Discapacidad Intelectual Ligada al Cromosoma X/metabolismo , Discapacidad Intelectual Ligada al Cromosoma X/fisiopatología , Ratones , Ratones Noqueados , Transportadores de Ácidos Monocarboxílicos/metabolismo , Hipotonía Muscular/metabolismo , Hipotonía Muscular/fisiopatología , Atrofia Muscular/metabolismo , Atrofia Muscular/fisiopatología , Neostriado/metabolismo , Proteínas de Transporte de Catión Orgánico/metabolismo , Simportadores/metabolismo , Yodotironina Deyodinasa Tipo II
9.
Genes (Basel) ; 11(12)2020 12 09.
Artículo en Inglés | MEDLINE | ID: mdl-33316910

RESUMEN

Ring chromosome 8 (r(8)) is one of the least frequent ring chromosomes. Usually, maternal chromosome 8 forms a ring, which can be lost from cells due to mitotic instability. The 8q24 region contains the imprinted KCNK9 gene, which is expressed from the maternal allele. Heterozygous KCNK9 mutations are associated with the imprinting disorder Birk-Barel syndrome. Here, we report a 2.5-year-old boy with developmental delay, microcephaly, dysmorphic features, diffuse muscle hypotonia, feeding problems, motor alalia and noncoarse neurogenic type of disturbance of muscle electrogenesis, partially overlapping with Birk-Barel syndrome phenotype. Cytogenetic analysis of lymphocytes revealed his karyotype to be 46,XY,r(8)(p23q24.3)[27]/45,XY,-8[3]. A de novo 7.9 Mb terminal 8p23.3p23.1 deletion, a 27.1 Mb 8p23.1p11.22 duplication, and a 4.4 Mb intact segment with a normal copy number located between them, as well as a 154-kb maternal LINGO2 gene deletion (9p21.2) with unknown clinical significance were identified by aCGH + SNP array. These aberrations were confirmed by real-time PCR. According to FISH analysis, the 8p23.1-p11.22 duplication was inverted. The ring chromosome originated from maternal chromosome 8. Targeted massive parallel sequencing did not reveal the KCNK9 mutations associated with Birk-Barel syndrome. Our data allow to assume that autosomal monosomy with inactive allele of imprinted gene arising from the loss of a ring chromosome in some somatic cells may be an etiological mechanism of mosaic imprinting disorders, presumably with less severe phenotype.


Asunto(s)
Anomalías Craneofaciales/genética , Discapacidad Intelectual/genética , Hipotonía Muscular/genética , Preescolar , Deleción Cromosómica , Cromosomas Humanos Par 8/genética , Cromosomas Humanos Par 8/metabolismo , Anomalías Craneofaciales/metabolismo , Impresión Genómica/genética , Humanos , Discapacidad Intelectual/metabolismo , Cariotipo , Cariotipificación/métodos , Masculino , Proteínas de la Membrana/genética , Mosaicismo , Hipotonía Muscular/metabolismo , Mutación/genética , Proteínas del Tejido Nervioso/genética , Fenotipo , Canales de Potasio de Dominio Poro en Tándem/genética , Cromosomas en Anillo
10.
Mol Med Rep ; 22(5): 3895-3903, 2020 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-33000225

RESUMEN

Potassium­channel tetramerization-domain-containing 1 (KCTD1) mutations are reported to result in scalp­ear­nipple syndrome. These mutations occur in the conserved broad­complex, tramtrack and bric a brac domain, which is associated with inhibited transcriptional activity. However, the mechanisms of KCTD1 mutants have not previously been elucidated; thus, the present study aimed to investigate whether KCTD1 mutants affect their interaction with transcription factor AP­2α and their regulation of the Wnt pathway. Results from the present study demonstrated that none of the ten KCTD1 mutants had an inhibitory effect on the transcriptional activity of AP­2α. Co­immunoprecipitation assays demonstrated that certain mutants exhibited changeable localization compared with the nuclear localization of wild­type KCTD1, but no KCTD1 mutant interacted with AP­2α. Almost all KCTD1 mutants, except KCTD1 A30E and H33Q, exhibited differential inhibitory effects on regulating TOPFLASH luciferase reporter activity. In addition, the interaction region of KCTD1 to the PY motif (amino acids 59­62) in AP­2α was identified. KCTD1 exhibited no suppressive effects on the transcriptional activity of the AP­2α P59A mutant, resulting in Char syndrome, a genetic disorder characterized by a distinctive facial appearance, heart defect and hand abnormalities, by altered protein cellular localization that abolished protein interactions. However, the P59A, P60A, P61R and 4A AP­2α mutants inhibited TOPFLASH reporter activity. Moreover, AP­2α and KCTD1 inhibited ß­catenin expression levels and SW480 cell viability. The present study thus identified a putative mechanism of disease­related KCTD1 mutants and AP­2α mutants by disrupting their interaction with the wildtype proteins AP­2α and KCTD1 and influencing the regulation of the Wnt/ß­catenin pathway.


Asunto(s)
Anomalías Múltiples/metabolismo , Proteínas Co-Represoras/metabolismo , Conducto Arterioso Permeable/metabolismo , Oído Externo/anomalías , Cara/anomalías , Dedos/anomalías , Hipospadias/metabolismo , Hipotonía Muscular/metabolismo , Proteínas Mutantes/metabolismo , Pezones/anomalías , Cuero Cabelludo/anomalías , Factor de Transcripción AP-2/metabolismo , Vía de Señalización Wnt/genética , beta Catenina/metabolismo , Anomalías Múltiples/genética , Supervivencia Celular/genética , Proteínas Co-Represoras/genética , Conducto Arterioso Permeable/genética , Oído Externo/metabolismo , Células HEK293 , Células HeLa , Humanos , Hipospadias/genética , Inmunoprecipitación , Hipotonía Muscular/genética , Mutación , Pezones/metabolismo , Fenotipo , Unión Proteica , Cuero Cabelludo/metabolismo , Factor de Transcripción AP-2/genética , Transfección
11.
Invest Ophthalmol Vis Sci ; 61(11): 18, 2020 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-32915983

RESUMEN

Purpose: Cohen syndrome (CS) is a rare genetic disorder caused by variants of the VPS13B gene. CS patients are affected with a severe form of retinal dystrophy, and in several cases cataracts also develop. The purpose of this study was to investigate the mechanisms and risk factors for cataract in CS, as well as to report on cataract surgeries in CS patients. Methods: To understand how VPS13B is associated with visual impairments in CS, we generated the Vps13b∆Ex3/∆Ex3 mouse model. Mice from 1 to 3 months of age were followed by ophthalmoscopy and slit-lamp examinations. Phenotypes were investigated by histology, immunohistochemistry, and western blot. Literature analysis was performed to determine specific characteristic features of cataract in CS and to identify potential genotype-phenotype correlations. Results: Cataracts rapidly developed in 2-month-old knockout mice and were present in almost all lenses at 3 months. Eye fundi appeared normal until cataract development. Lens immunostaining revealed that cataract formation was associated with the appearance of large vacuoles in the cortical area, epithelial-mesenchymal transition, and fibrosis. In later stages, cataracts became hypermature, leading to profound retinal remodeling due to inflammatory events. Literature analysis showed that CS-related cataracts display specific features compared to other forms of retinitis pigmentosa-related cataracts, and their onset is modified by additional genetic factors. Corroboratively, we were able to isolate a subline of the Vps13b∆Ex3/∆Ex3 model with delayed cataract onset. Conclusions: VPS13B participates in lens homeostasis, and the CS-related cataract development dynamic is linked to additional genetic factors.


Asunto(s)
Catarata/genética , Dedos/anomalías , Regulación de la Expresión Génica , Homeostasis/genética , Discapacidad Intelectual/complicaciones , Cristalino/metabolismo , Microcefalia/complicaciones , Hipotonía Muscular/complicaciones , Miopía/complicaciones , Obesidad/complicaciones , ARN/genética , Degeneración Retiniana/complicaciones , Proteínas de Transporte Vesicular/genética , Animales , Western Blotting , Catarata/etiología , Catarata/metabolismo , Discapacidades del Desarrollo/complicaciones , Discapacidades del Desarrollo/genética , Discapacidades del Desarrollo/metabolismo , Modelos Animales de Enfermedad , Discapacidad Intelectual/genética , Discapacidad Intelectual/metabolismo , Cristalino/patología , Ratones Endogámicos C57BL , Ratones Noqueados , Microcefalia/genética , Microcefalia/metabolismo , Hipotonía Muscular/genética , Hipotonía Muscular/metabolismo , Miopía/genética , Miopía/metabolismo , Obesidad/genética , Obesidad/metabolismo , Degeneración Retiniana/genética , Degeneración Retiniana/metabolismo , Proteínas de Transporte Vesicular/biosíntesis
12.
Mol Brain ; 13(1): 69, 2020 05 06.
Artículo en Inglés | MEDLINE | ID: mdl-32375900

RESUMEN

Significant clinical symptoms of Cohen syndrome (CS), a rare autosomal recessive disorder, include intellectual disability, facial dysmorphism, postnatal microcephaly, retinal dystrophy, and intermittent neutropenia. CS has been associated with mutations in the VPS13B (vacuolar protein sorting 13 homolog B) gene, which regulates vesicle-mediated protein sorting and transport; however, the cellular mechanism underlying CS pathogenesis in patient-derived neurons remains uncertain. This report states that autophagic vacuoles accumulate in CS fibroblasts and the axonal terminals of CS patient-specific induced pluripotent stem cells (CS iPSC)-derived neurons; additionally, autophagic flux was significantly increased in CS-derived neurons compared to control neurons. VPS13B knockout HeLa cell lines generated using the CRISPR/Cas9 genome editing system showed significant upregulation of autophagic flux, indicating that VSP13B may be associated with autophagy in CS. Transcriptomic analysis focusing on the autophagy pathway revealed that genes associated with autophagosome organization were dysregulated in CS-derived neurons. ATG4C is a mammalian ATG4 paralog and a crucial regulatory component of the autophagosome biogenesis/recycling pathway. ATG4C was significantly upregulated in CS-derived neurons, indicating that autophagy is upregulated in CS neurons. The autophagy pathway in CS neurons may be associated with the pathophysiology exhibited in the neural network of CS patients.


Asunto(s)
Autofagosomas/metabolismo , Autofagia/genética , Fibroblastos/metabolismo , Dedos/anomalías , Células Madre Pluripotentes Inducidas/metabolismo , Discapacidad Intelectual/metabolismo , Microcefalia/metabolismo , Hipotonía Muscular/metabolismo , Miopía/metabolismo , Neuronas/metabolismo , Obesidad/metabolismo , Degeneración Retiniana/metabolismo , Proteínas de Transporte Vesicular/genética , Autofagosomas/genética , Autofagosomas/ultraestructura , Proteínas Relacionadas con la Autofagia/genética , Proteínas Relacionadas con la Autofagia/metabolismo , Axones/metabolismo , Cisteína Endopeptidasas/genética , Cisteína Endopeptidasas/metabolismo , Discapacidades del Desarrollo/metabolismo , Discapacidades del Desarrollo/fisiopatología , Fibroblastos/patología , Fibroblastos/ultraestructura , Dedos/fisiopatología , Técnicas de Inactivación de Genes , Células HeLa , Humanos , Células Madre Pluripotentes Inducidas/patología , Discapacidad Intelectual/fisiopatología , Microcefalia/fisiopatología , Microscopía Electrónica , Hipotonía Muscular/fisiopatología , Mutación Missense , Miopía/fisiopatología , Red Nerviosa/fisiología , Neuronas/patología , Obesidad/fisiopatología , Degeneración Retiniana/fisiopatología , Regulación hacia Arriba , Vacuolas/metabolismo
13.
Exp Clin Endocrinol Diabetes ; 128(6-07): 414-422, 2020 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-32242326

RESUMEN

Despite its first description more than 75 years ago, effective treatment for "Allan-Herndon-Dudley-Syndrome (AHDS)", an X-linked thyroid hormone transporter defect, is unavailable. Mutations in the SLC16A2 gene have been discovered to be causative for AHDS in 2004, but a comprehensive understanding of the function of the encoded protein, monocarboxylate transporter 8 (MCT8), is incomplete. Patients with AHDS suffer from neurodevelopmental delay, as well as extrapyramidal (dystonia, chorea, athetosis), pyramidal (spasticity), and cerebellar symptoms (ataxia). This suggests an affection of the pyramidal tracts, basal ganglia, and cerebellum, most likely already during fetal brain development. The function of other brain areas relevant for mood, behavior, and vigilance seems to be intact. An optimal treatment strategy should thus aim to deliver T3 to these relevant structures at the correct time points during development. A potential therapeutic strategy meeting these needs might be the delivery of T3 via a "Trojan horse mechanism" by which T3 is delivered into target cells by a thyroid hormone transporter independent T3 internalization.


Asunto(s)
Ganglios Basales , Cerebelo , Discapacidad Intelectual Ligada al Cromosoma X/tratamiento farmacológico , Discapacidad Intelectual Ligada al Cromosoma X/metabolismo , Discapacidad Intelectual Ligada al Cromosoma X/fisiopatología , Transportadores de Ácidos Monocarboxílicos/metabolismo , Hipotonía Muscular/tratamiento farmacológico , Hipotonía Muscular/metabolismo , Hipotonía Muscular/fisiopatología , Atrofia Muscular/tratamiento farmacológico , Atrofia Muscular/metabolismo , Atrofia Muscular/fisiopatología , Tractos Piramidales , Triyodotironina/administración & dosificación , Triyodotironina/metabolismo , Ganglios Basales/efectos de los fármacos , Ganglios Basales/fisiopatología , Cerebelo/efectos de los fármacos , Cerebelo/fisiopatología , Humanos , Tractos Piramidales/efectos de los fármacos , Tractos Piramidales/fisiopatología
14.
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
15.
Thyroid ; 30(8): 1205-1216, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32188347

RESUMEN

Background: Tachycardia, cardiac hypertrophy, and elevated body temperature are major signs of systemic hyperthyroidism, which are considered to reflect the excessive thyroid hormone (TH) action in the respective peripheral tissues. However, recent observations indicate that the central actions of TH also contribute substantially to cardiovascular regulation and thermogenesis. Methods: In this study, we dissect the individual contributions of peripheral TH action versus the central effects in body temperature regulation and cardiovascular functions by taking advantage of mice lacking the TH transporters monocarboxylate transporter 8 (MCT8) and organic anion transporting polypeptide 1C1 (OATP1C1) (M/O double knock-out [dko]), which exhibit elevated serum triiodothyronine (T3) levels while their brain is in a profoundly hypothyroid state. We compared these animals with wild-type (WT) mice that were treated orally with T3 to achieve similarly elevated serum T3 levels, but are centrally hyperthyroid. For the studies, we used radiotelemetry, infrared thermography, gene expression profiling, Western blot analyses, and enzyme linked immunosorbent assays (ELISA) assays. Results: Our analyses revealed mild hyperthermia and cardiac hypertrophy in T3-treated WT mice but not in M/O dko animals, suggesting that central actions of TH are required for these hyperthyroid phenotypes. Although the average heart rate was unaffected in either model, the M/O dko exhibited an altered heart rate frequency distribution with tachycardic bursts in active periods and bradycardic episodes during resting time, demonstrating that the stabilization of heart rate by the autonomic nervous system can be impaired in centrally hypothyroid animals. Conclusions: Our studies unravel distinct phenotypical traits of hyperthyroidism that depend on an intact central nervous system, and provide valuable insight into the cardiovascular pathology of the Allan-Herndon-Dudley syndrome, a condition caused by the lack of MCT8 in humans.


Asunto(s)
Cardiomegalia/metabolismo , Fiebre/metabolismo , Frecuencia Cardíaca , Hipotiroidismo/complicaciones , Hormonas Tiroideas/metabolismo , Animales , Cardiomegalia/prevención & control , Cruzamientos Genéticos , Ensayo de Inmunoadsorción Enzimática , Perfilación de la Expresión Génica , Glucógeno/metabolismo , Lipólisis , Hígado/metabolismo , Masculino , Discapacidad Intelectual Ligada al Cromosoma X/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Transportadores de Ácidos Monocarboxílicos/genética , Hipotonía Muscular/metabolismo , Atrofia Muscular/metabolismo , Fenotipo , Telemetría , Termogénesis , Termografía , Factores de Tiempo , Triyodotironina/sangre
16.
Am J Hum Genet ; 106(2): 272-279, 2020 02 06.
Artículo en Inglés | MEDLINE | ID: mdl-32004445

RESUMEN

Recent studies have identified both recessive and dominant forms of mitochondrial disease that result from ATAD3A variants. The recessive form includes subjects with biallelic deletions mediated by non-allelic homologous recombination. We report five unrelated neonates with a lethal metabolic disorder characterized by cardiomyopathy, corneal opacities, encephalopathy, hypotonia, and seizures in whom a monoallelic reciprocal duplication at the ATAD3 locus was identified. Analysis of the breakpoint junction fragment indicated that these 67 kb heterozygous duplications were likely mediated by non-allelic homologous recombination at regions of high sequence identity in ATAD3A exon 11 and ATAD3C exon 7. At the recombinant junction, the duplication allele produces a fusion gene derived from ATAD3A and ATAD3C, the protein product of which lacks key functional residues. Analysis of fibroblasts derived from two affected individuals shows that the fusion gene product is expressed and stable. These cells display perturbed cholesterol and mitochondrial DNA organization similar to that observed for individuals with severe ATAD3A deficiency. We hypothesize that the fusion protein acts through a dominant-negative mechanism to cause this fatal mitochondrial disorder. Our data delineate a molecular diagnosis for this disorder, extend the clinical spectrum associated with structural variation at the ATAD3 locus, and identify a third mutational mechanism for ATAD3 gene cluster variants. These results further affirm structural variant mutagenesis mechanisms in sporadic disease traits, emphasize the importance of copy number analysis in molecular genomic diagnosis, and highlight some of the challenges of detecting and interpreting clinically relevant rare gene rearrangements from next-generation sequencing data.


Asunto(s)
ATPasas Asociadas con Actividades Celulares Diversas/genética , Colesterol/metabolismo , Duplicación de Gen , Recombinación Homóloga , Proteínas de la Membrana/genética , Mitocondrias/patología , Enfermedades Mitocondriales/patología , Proteínas Mitocondriales/genética , ATPasas Asociadas con Actividades Celulares Diversas/química , Secuencia de Aminoácidos , Encefalopatías/etiología , Encefalopatías/metabolismo , Encefalopatías/patología , Cardiomiopatías/etiología , Cardiomiopatías/metabolismo , Cardiomiopatías/patología , Opacidad de la Córnea/etiología , Opacidad de la Córnea/metabolismo , Opacidad de la Córnea/patología , Variaciones en el Número de Copia de ADN , Femenino , Reordenamiento Génico , Humanos , Lactante , Recién Nacido , Masculino , Proteínas de la Membrana/química , Mitocondrias/genética , Mitocondrias/metabolismo , Enfermedades Mitocondriales/genética , Enfermedades Mitocondriales/metabolismo , Proteínas Mitocondriales/química , Hipotonía Muscular/etiología , Hipotonía Muscular/metabolismo , Hipotonía Muscular/patología , Mutación , Conformación Proteica , Convulsiones/etiología , Convulsiones/metabolismo , Convulsiones/patología , Homología de Secuencia
17.
Nat Commun ; 11(1): 480, 2020 01 24.
Artículo en Inglés | MEDLINE | ID: mdl-31980599

RESUMEN

Mutations in the actively expressed, maternal allele of the imprinted KCNK9 gene cause Birk-Barel intellectual disability syndrome (BBIDS). Using a BBIDS mouse model, we identify here a partial rescue of the BBIDS-like behavioral and neuronal phenotypes mediated via residual expression from the paternal Kcnk9 (Kcnk9pat) allele. We further demonstrate that the second-generation HDAC inhibitor CI-994 induces enhanced expression from the paternally silenced Kcnk9 allele and leads to a full rescue of the behavioral phenotype suggesting CI-994 as a promising molecule for BBIDS therapy. Thus, these findings suggest a potential approach to improve cognitive dysfunction in a mouse model of an imprinting disorder.


Asunto(s)
Anomalías Craneofaciales/genética , Anomalías Craneofaciales/metabolismo , Histonas/metabolismo , Discapacidad Intelectual/genética , Discapacidad Intelectual/metabolismo , Hipotonía Muscular/genética , Hipotonía Muscular/metabolismo , Canales de Potasio/genética , Animales , Conducta Animal , Benzamidas , Encéfalo/metabolismo , Anomalías Craneofaciales/tratamiento farmacológico , Modelos Animales de Enfermedad , Femenino , Técnicas de Silenciamiento del Gen , Impresión Genómica , Inhibidores de Histona Desacetilasas/farmacología , Humanos , Discapacidad Intelectual/tratamiento farmacológico , Locus Coeruleus/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Hipotonía Muscular/tratamiento farmacológico , Mutación , Fenotipo , Fenilendiaminas/farmacología , Canales de Potasio/deficiencia , Canales de Potasio/metabolismo , Regulación hacia Arriba/efectos de los fármacos
18.
Exp Clin Endocrinol Diabetes ; 128(6-07): 423-427, 2020 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-31724131

RESUMEN

Thyroid hormone (TH) transporters are required for cellular transmembrane passage of TH and are thus mandatory for proper TH metabolism and action. Consequently, inactivating mutations in TH transporters such as MCT8 or OATP1C1 can cause tissue- specific changes in TH homeostasis. As the most prominent example, patients with MCT8 mutations exhibit elevated serum T3 levels, whereas their CNS appear to be in a TH deficient state. Here, we will briefly summarize recent studies of mice lacking Mct8 alone or in combination with the TH transporters Mct10 or Oatp1c1 that shed light on many aspects and pathogenic events underlying global MCT8 deficiency and also underscore the contribution of Mct10 and Oatp1c1 in tissue-specific TH transport processes. Moreover, development of conditional knock-out mice that allow a cell-specific inactivation of TH transporters in distinct tissues, disclosed cell-specific changes in TH signaling, thereby highlighting the pathophysiological significance of local control of TH action.


Asunto(s)
Discapacidad Intelectual Ligada al Cromosoma X/metabolismo , Transportadores de Ácidos Monocarboxílicos/metabolismo , Hipotonía Muscular/metabolismo , Atrofia Muscular/metabolismo , Transportadores de Anión Orgánico/metabolismo , Tiroxina/metabolismo , Triyodotironina/metabolismo , Animales , Humanos
19.
Endocr Rev ; 41(2)2020 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-31754699

RESUMEN

Thyroid hormone transporters at the plasma membrane govern intracellular bioavailability of thyroid hormone. Monocarboxylate transporter (MCT) 8 and MCT10, organic anion transporting polypeptide (OATP) 1C1, and SLC17A4 are currently known as transporters displaying the highest specificity toward thyroid hormones. Structure-function studies using homology modeling and mutational screens have led to better understanding of the molecular basis of thyroid hormone transport. Mutations in MCT8 and in OATP1C1 have been associated with clinical disorders. Different animal models have provided insight into the functional role of thyroid hormone transporters, in particular MCT8. Different treatment strategies for MCT8 deficiency have been explored, of which thyroid hormone analogue therapy is currently applied in patients. Future studies may reveal the identity of as-yet-undiscovered thyroid hormone transporters. Complementary studies employing animal and human models will provide further insight into the role of transporters in health and disease. (Endocrine Reviews 41: 1 - 55, 2020).


Asunto(s)
Transporte Biológico/fisiología , Proteínas de Transporte de Membrana/fisiología , Discapacidad Intelectual Ligada al Cromosoma X , Transportadores de Ácidos Monocarboxílicos/fisiología , Hipotonía Muscular , Atrofia Muscular , Transportadores de Anión Orgánico/fisiología , Simportadores/fisiología , Hormonas Tiroideas/metabolismo , Animales , Humanos , Proteínas de Transporte de Membrana/deficiencia , Proteínas de Transporte de Membrana/genética , Discapacidad Intelectual Ligada al Cromosoma X/genética , Discapacidad Intelectual Ligada al Cromosoma X/metabolismo , Discapacidad Intelectual Ligada al Cromosoma X/fisiopatología , Discapacidad Intelectual Ligada al Cromosoma X/terapia , Transportadores de Ácidos Monocarboxílicos/deficiencia , Transportadores de Ácidos Monocarboxílicos/genética , Hipotonía Muscular/genética , Hipotonía Muscular/metabolismo , Hipotonía Muscular/fisiopatología , Hipotonía Muscular/terapia , Atrofia Muscular/genética , Atrofia Muscular/metabolismo , Atrofia Muscular/fisiopatología , Atrofia Muscular/terapia , Transportadores de Anión Orgánico/deficiencia , Transportadores de Anión Orgánico/genética , Simportadores/deficiencia , Simportadores/genética , Hormonas Tiroideas/uso terapéutico
20.
Thyroid ; 29(10): 1499-1510, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31436139

RESUMEN

Background: Mutations in the thyroid hormone (TH) transporter monocarboxylate transporter 8 (MCT8) cause MCT8 deficiency, characterized by severe intellectual and motor disability and abnormal serum thyroid function tests. Various Mct8 knock-out mouse models as well as mct8 knock-out and knockdown zebrafish models are used as a disease model for MCT8 deficiency. Although important for model eligibility, little is known about the functional characteristics of the MCT8 orthologues in these species. Therefore, we here compared the functional characteristics of mouse (mm) MCT8 and zebrafish (dr) Mct8 to human (hs) MCT8. Methods: We performed extensive transport studies in COS-1 and JEG-3 cells transiently transfected with hsMCT8, drMct8, and mmMCT8. Protein expression levels and subcellular localization were assessed by immunoblotting, surface biotinylation, and immunocytochemistry. Sequence alignment and structural modeling were used to interpret functional differences between the orthologues. Results: hsMCT8, drMct8, and mmMCT8 all facilitated the uptake and efflux of 3,3'-diiodothyronine (3,3'-T2), rT3, triiodothyronine (T3), and thyroxine (T4), although the initial uptake rates of drMct8 were 1.5-4.0-fold higher than for hsMCT8 and mmMCT8. drMct8 exhibited 3-50-fold lower apparent IC50 values than hsMCT8 and mmMCT8 for all tested substrates, and substrate preference of drMct8 (3,3'-T2, T3 > T4 > rT3) differed from hsMCT8 and mmMCT8 (T3 > T4 > rT3, 3,3'-T2). Compared with hsMCT8 and mmMCT8, cis-inhibition studies showed that T3 uptake by drMct8 was inhibited at a lower concentration and by a broader spectrum of TH metabolites. Total and cell surface expression levels of drMct8 and hsMCT8 were equal and both significantly exceeded those of mmMCT8. Structural modeling located most non-conserved residues outside the substrate pore, except for H192 in hsMCT8, which is replaced by a glutamine in drMct8. However, a H192Q substituent of hsMCT8 did not alter its transporter characteristics. Conclusion: Our studies substantiate the eligibility of mice and zebrafish models for human MCT8 deficiency. However, differences in the intrinsic transporter properties of MCT8 orthologues may exist, which should be realized when comparing MCT8 deficiency in different in vivo models. Moreover, our findings may indicate that the protein domains outside the substrate channel may play a role in substrate selection and protein stability.


Asunto(s)
Transportadores de Ácidos Monocarboxílicos/metabolismo , Simportadores/metabolismo , Animales , Células COS , Línea Celular Tumoral , Chlorocebus aethiops , Diyodotironinas/metabolismo , Modelos Animales de Enfermedad , Técnicas de Silenciamiento del Gen , Humanos , Immunoblotting , Inmunohistoquímica , Técnicas In Vitro , Discapacidad Intelectual Ligada al Cromosoma X/genética , Discapacidad Intelectual Ligada al Cromosoma X/metabolismo , Ratones , Ratones Noqueados , Modelos Moleculares , Hipotonía Muscular/genética , Hipotonía Muscular/metabolismo , Atrofia Muscular/genética , Atrofia Muscular/metabolismo , Alineación de Secuencia , Tiroxina/metabolismo , Triyodotironina/metabolismo , Triyodotironina Inversa/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...