Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Nutr Bull ; 48(4): 559-571, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37905391

RESUMEN

Childhood dyslipidaemia is associated with the occurrence of cardiovascular diseases in adulthood, so evaluating whether an individual has a genetic predisposition to this pathology is of great importance for early action of prevention and treatment. This study aimed to evaluate the association between the FTO (rs9939609), MC4R (rs17782313) and MTMR9 (rs2293855) polymorphisms, the obesity-related genetic risk score and atherogenic risk in Brazilian children. This is a cross-sectional study conducted in 544 children aged 4-9 years in the city of Viçosa, Minas Gerais state, Brazil. The single nucleotide polymorphisms rs9939609, rs17782313 and rs2293855, were identified by the system TaqMan SNP genotyping and the obesity-related genetic risk score was determined. The lipid profile (serum total cholesterol [TC], high density lipoprotein [HDL] cholesterol, low density lipoprotein [LDL] cholesterol, triglycerides) was analysed and the atherogenic indices (Castelli I and II indices), atherogenic coefficient (AC), lipoprotein combined index (LCI) and plasma atherogenic index (PAI) were calculated. A semi-structured questionnaire was applied, obtaining data on the sociodemographic, economic and lifestyle characteristics of the children. Weight and height measurements were performed in all children, and body composition was evaluated by Dual-Energy X-ray Absorptiometry (DXA). 55.5% of the sample had dyslipidaemia, while 28.5% of the sample had at least one polymorphism and 2.2% had three polymorphisms. Children with the AG/AA genotypes in the rs2293855 polymorphism had lower HDL cholesterol levels and higher TC/HDL cholesterol, LDL/HDL cholesterol ratios and AC. Those with one or more polymorphisms (rs9939609, rs17782313 and rs2293855) in the genetic risk score had lower HDL cholesterol levels and higher TC/HDL cholesterol ratios, AC, LCI and PAI. In conclusion, the risk allele of the rs2293855 polymorphism and a higher obesity-related genetic risk score were positively associated with higher atherogenic risk in Brazilian children.


Asunto(s)
Dislipidemias , Obesidad , Niño , Humanos , HDL-Colesterol , Genotipo , Estudios Transversales , Índice de Masa Corporal , Polimorfismo de Nucleótido Simple/genética , Colesterol , Lipoproteínas HDL/genética , Dislipidemias/epidemiología , Proteínas Tirosina Fosfatasas no Receptoras/genética , Dioxigenasa FTO Dependiente de Alfa-Cetoglutarato/genética
2.
Oncogene ; 39(18): 3693-3709, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32152405

RESUMEN

Caveolin-1 (CAV1) enhanced migration, invasion, and metastasis of cancer cells is inhibited by co-expression of the glycoprotein E-cadherin. Although the two proteins form a multiprotein complex that includes ß-catenin, it remained unclear how this would contribute to blocking the metastasis promoting function of CAV1. Here, we characterized by mass spectrometry the protein composition of CAV1 immunoprecipitates from B16F10 murine melanoma cells expressing or not E-cadherin. The novel protein tyrosine phosphatase PTPN14 was identified by mass spectrometry analysis exclusively in co-immunoprecipitates of CAV1 with E-cadherin. Interestingly, PTPN14 is implicated in controlling metastasis, but only few known PTPN14 substrates exist. We corroborated by western blotting experiments that PTPN14 and CAV1 co-inmunoprecipitated in the presence of E-cadherin in B16F10 melanoma and other cancer cells. Moreover, the CAV1(Y14F) mutant protein was shown to co-immunoprecipitate with PTPN14 even in the absence of E-cadherin, and overexpression of PTPN14 reduced CAV1 phosphorylation on tyrosine-14, as well as suppressed CAV1-enhanced cell migration, invasion and Rac-1 activation in B16F10, metastatic colon [HT29(US)] and breast cancer (MDA-MB-231) cell lines. Finally, PTPN14 overexpression in B16F10 cells reduced the ability of CAV1 to induce metastasis in vivo. In summary, we identify here CAV1 as a novel substrate for PTPN14 and show that overexpression of this phosphatase suffices to reduce CAV1-induced metastasis.


Asunto(s)
Cadherinas/genética , Caveolina 1/genética , Melanoma Experimental/genética , Proteínas Tirosina Fosfatasas no Receptoras/genética , Animales , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular/genética , Femenino , Regulación Neoplásica de la Expresión Génica/genética , Humanos , Melanoma Experimental/patología , Ratones , Invasividad Neoplásica , Metástasis de la Neoplasia , Fosforilación/genética , beta Catenina/genética
3.
Sci Rep ; 9(1): 11621, 2019 08 12.
Artículo en Inglés | MEDLINE | ID: mdl-31406157

RESUMEN

Carotid intima-media thickness (cIMT) is a subclinical marker for atherosclerosis. Previously, we reported a quantitative trait locus (QTL) for total cIMT on chromosome 14q and identified PRiMA1, FOXN3 and CCDC88C as candidate genes using a common variants (CVs)-based approach. Herein, we further evaluated the genetic contribution of the QTL to cIMT by resequencing. We sequenced all exons within the QTL and genomic regions of PRiMA1, FOXN3 and CCDC88C in Dominican families with evidence for linkage to the QTL. Unrelated Dominicans from the Northern Manhattan Study (NOMAS) were used for validation. Single-variant-based and gene-based analyses were performed for CVs and rare variants (RVs). The strongest evidence for association with CVs was found in PRiMA1 (p = 8.2 × 10-5 in families, p = 0.01 in NOMAS at rs12587586), and in the five-gene cluster SPATA7-PTPN21-ZC3H14-EML5-TTC8 locus (p = 1.3 × 10-4 in families, p = 0.01 in NOMAS at rs2274736). No evidence for association with RVs was found in PRiMA1. The top marker from previous study in PRiMA1 (rs7152362) was associated with fewer atherosclerotic events (OR = 0.67; p = 0.02 in NOMAS) and smaller cIMT (ß = -0.58, p = 2.8 × 10-4 in Family). Within the five-gene cluster, evidence for association was found for exonic RVs (p = 0.02 in families, p = 0.28 in NOMAS), which was enriched among RVs with higher functional potentials (p = 0.05 in NOMAS for RVs in the top functional tertile). In summary, targeted resequencing provided validation and novel insights into the genetic architecture of cIMT, suggesting stronger effects for RVs with higher functional potentials. Furthermore, our data support the clinical relevance of CVs associated with subclinical atherosclerosis.


Asunto(s)
Aterosclerosis/genética , Grosor Intima-Media Carotídeo , Ligamiento Genético , Proteínas de la Membrana/genética , Proteínas del Tejido Nervioso/genética , Aterosclerosis/patología , Proteínas del Citoesqueleto/genética , Proteínas de Unión al ADN/genética , República Dominicana , Femenino , Humanos , Escala de Lod , Masculino , Proteínas Asociadas a Microtúbulos/genética , Proteínas de Unión a Poli(A)/genética , Polimorfismo de Nucleótido Simple , Proteínas Tirosina Fosfatasas no Receptoras/genética , Sitios de Carácter Cuantitativo
4.
Medicina (B Aires) ; 78(6): 436-439, 2018.
Artículo en Español | MEDLINE | ID: mdl-30504111

RESUMEN

Lafora's disease is infrequent. However, it is one of the most common causes of progressive myoclonus epilepsy. We present the case of a 19-year-old woman, without comorbidities and normal development that started at 8 years with seizures and that from 15 years, had progressive cognitive deterioration. She was admitted to our institution with a diagnosis of super refractory status epilepticus. The diagnosis of Lafora's disease was made through pathological anatomy, later a genetic test was performed that reported a pathogenic variant of the EPM2A gene, confirming the diagnosis. We present a cause of progressive myoclonic epilepsy, with an ominous prognosis and a treatment oriented to palliative measures, so it is important to analyze the differential diagnoses with other entities, in order to establish a prognosis, offer better quality of life, adequate medical care and provide genetic counseling to family members.


Asunto(s)
Enfermedad de Lafora/complicaciones , Epilepsias Mioclónicas Progresivas/etiología , Biopsia , Diagnóstico Diferencial , Electroencefalografía , Femenino , Humanos , Enfermedad de Lafora/genética , Enfermedad de Lafora/patología , Mutación/genética , Epilepsias Mioclónicas Progresivas/genética , Proteínas Tirosina Fosfatasas no Receptoras , Adulto Joven
5.
Medicina (B.Aires) ; Medicina (B.Aires);78(6): 436-439, Dec. 2018. ilus, tab
Artículo en Español | LILACS | ID: biblio-976142

RESUMEN

La enfermedad de Lafora es infrecuente; sin embargo, es una de las causas más comunes de epilepsia mioclónica progresiva. Presentamos el caso de una mujer de 19 años sin comorbilidades y pautas madurativas normales, que inició a los 8 años con convulsiones y que a partir de los 15 años agregó deterioro cognitivo progresivo. Fue internada en nuestra institución con diagnóstico de estatus epiléptico super refractario. Se diagnosticó enfermedad de Lafora, confirmada por la anatomía patológica, y posteriormente se realizó un test genético que informó una variante patogénica del gen EPM2A, que confirmó el diagnóstico. Presentamos una causa de epilepsia mioclónica progresiva, con un pronóstico ominoso y un tratamiento orientado a medidas paliativas, por lo que es importante analizar los diagnósticos diferenciales con otras entidades, a fin de establecer un pronóstico, ofrecer mejor calidad de vida, asistencia médica adecuada y brindar asesoría genética a los familiares.


Lafora's disease is infrequent. However, it is one of the most common causes of progressive myoclonus epilepsy. We present the case of a 19-year-old woman, without comorbidities and normal development that started at 8 years with seizures and that from 15 years, had progressive cognitive deterioration. She was admitted to our institution with a diagnosis of super refractory status epilepticus. The diagnosis of Lafora's disease was made through pathological anatomy, later a genetic test was performed that reported a pathogenic variant of the EPM2A gene, confirming the diagnosis. We present a cause of progressive myoclonic epilepsy, with an ominous prognosis and a treatment oriented to palliative measures, so it is important to analyze the differential diagnoses with other entities, in order to establish a prognosis, offer better quality of life, adequate medical care and provide genetic counseling to family members.


Asunto(s)
Humanos , Femenino , Adulto Joven , Epilepsias Mioclónicas Progresivas/etiología , Enfermedad de Lafora/complicaciones , Biopsia , Epilepsias Mioclónicas Progresivas/genética , Enfermedad de Lafora/genética , Enfermedad de Lafora/patología , Diagnóstico Diferencial , Electroencefalografía , Proteínas Tirosina Fosfatasas no Receptoras , Mutación/genética
6.
Biochim Biophys Acta Mol Basis Dis ; 1864(6 Pt A): 2010-2020, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29577983

RESUMEN

Oxidative stress affects the survival and function of neurons. Hence, they have a complex and highly regulated machinery to handle oxidative changes. The dysregulation of this antioxidant machinery is associated with a wide range of neurodegenerative conditions. Therefore, we evaluated signaling alterations, synaptic properties and behavioral performance in 2 and 6-month-old heterozygous manganese superoxide dismutase knockout mice (SOD2+/- mice). We found that their low antioxidant capacity generated direct oxidative damage in proteins, lipids, and DNA. However, only 6-month-old heterozygous knockout mice presented behavioral impairments. On the other hand, synaptic plasticity, synaptic strength and NMDA receptor (NMDAR) dependent postsynaptic potentials were decreased in an age-dependent manner. We also analyzed the phosphorylation state of the NMDAR subunit GluN2B. We found that while the levels of GluN2B phosphorylated on tyrosine 1472 (synaptic form) remain unchanged, we detected increased levels of GluN2B phosphorylated on tyrosine 1336 (extrasynaptic form), establishing alterations in the synaptic/extrasynaptic ratio of GluN2B. Additionally, we found increased levels of two phosphatases associated with dephosphorylation of p-1472: striatal-enriched protein tyrosine phosphatase (STEP) and phosphatase and tensin homolog deleted on chromosome Ten (PTEN). Moreover, we found decreased levels of p-CREB, a master transcription factor activated by synaptic stimulation. In summary, we describe mechanisms by which glutamatergic synapses are altered under oxidative stress conditions. Our results uncovered new putative therapeutic targets for conditions where NMDAR downstream signaling is altered. This work also contributes to our understanding of processes such as synapse formation, learning, and memory in neuropathological conditions.


Asunto(s)
Envejecimiento/fisiología , N-Metilaspartato/metabolismo , Neuronas/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Superóxido Dismutasa/metabolismo , Factores de Edad , Animales , Conducta Animal/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Animales , Enfermedades Neurodegenerativas/patología , Plasticidad Neuronal/fisiología , Estrés Oxidativo/fisiología , Fosfohidrolasa PTEN/metabolismo , Fosforilación/fisiología , Proteínas Tirosina Fosfatasas no Receptoras/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/fisiología , Superóxido Dismutasa/genética , Sinapsis/metabolismo
7.
Pediatr Neurol ; 58: 107-12, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-26995067

RESUMEN

BACKGROUND: Myotubular myopathy is a rare X-linked congenital myopathy characterized by marked neonatal hypotonia and respiratory insufficiency, facial and ocular involvement, and muscle biopsy with prominent central nuclei in the majority of muscle fibers. It is caused by mutations in MTM1, which codes for the phosphoinositides phosphatase myotubularin. In this work, we established and detailed a new cohort of six patients at the clinical, histologic, and genetic levels. PATIENTS AND METHODS: Patients were recruited after screening 3065 muscle biopsy reports from two large biopsy banks in Sao Paulo, Brazil from the years 2008 to 2013, and from referrals to a neuromuscular outpatient clinic between 2011 and 2013. We reviewed biopsy slides, evaluated patients, and Sanger sequenced MTM1 in the families. RESULTS: All patients but one had classic phenotypes with a stable course after a severe onset. Two patients died suddenly from hypovolemic shock. Muscle biopsies had been performed in five patients, all of whom showed a classic pattern with a predominance of centrally located nuclei and increased oxidative activity in the center of the fibers. Two patients showed necklace fibers, and two families had novel truncating mutations in MTM1. CONCLUSIONS: X-linked myotubular myopathy is rare in the Brazilian population. Necklace fibers might be more prevalent in this condition than previously reported. Direct Sanger sequencing of MTM1 on clinical suspicion avoids the need of a muscle biopsy.


Asunto(s)
Músculo Esquelético/patología , Miopatías Estructurales Congénitas/genética , Miopatías Estructurales Congénitas/patología , Proteínas Tirosina Fosfatasas no Receptoras/genética , Biopsia , Brasil , Niño , Preescolar , Estudios de Cohortes , Análisis Mutacional de ADN , Cara/anomalías , Humanos , Lactante , Masculino , Miopatías Estructurales Congénitas/epidemiología , Miopatías Estructurales Congénitas/fisiopatología , Fenotipo
8.
Epilepsy Res ; 108(9): 1501-10, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25246353

RESUMEN

Lafora disease (LD) is an autosomal recessive progressive myoclonus epilepsy with classic adolescent onset of stimuli sensitive seizures. Patients typically deteriorate rapidly with dementia, ataxia, vegetative failure and death by 25 years of age. LD is caused by homozygous mutations in EPM2A or EPM2B genes. We found four novel mutations in EPM2A - three in exon 4 (Q247X, H265R G279C) and one in exon 1 (Y86D) - and a previously described mutation in exon 4 (R241X). These five EPM2A mutations were found in four index cases and affected relatives. Patient 1 with classic LD was doubly heterozygous for H265R and R241X in exon 4; while Patient 2, who also had classic LD, was homozygous for Q247X in exon 4. Patient 3 with classic LD was homozygous for Y86D in exon 1, but the same mutation in his affected brother manifested an atypical earlier childhood onset. For the first time, we describe a later onset and slower progression of EPM2A-deficient LD seen in Patient 4 and her three sisters who were doubly heterozygous for R241X and G279C in exon 4. In these sisters, seizures started later at 21 to 28 years of age and progressed slowly with patients living beyond 30 years of age. Our observations suggest that variations in phenotypes of EPM2A-deficient LD, like an earlier childhood or adolescent or later adult onset with a rapid or slower course, depend on a second modifying factor separate from pathogenicity or exon location of EPM2A mutations. A modifying gene amongst the patient's genetic background or environmental factors may condition age of onset and rapid or slow progression of LD.


Asunto(s)
Enfermedad de Lafora/genética , Mutación/genética , Proteínas Tirosina Fosfatasas no Receptoras/genética , Adulto , Análisis Mutacional de ADN , Electroencefalografía , Salud de la Familia , Femenino , Humanos , Enfermedad de Lafora/diagnóstico , Masculino
9.
Neuromuscul Disord ; 22(6): 541-5, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22264517

RESUMEN

X-linked myotubular myopathy due to mutations in the MTM1 gene is classically characterized by a severe neonatal phenotype and a typical muscle biopsy presenting globular and centrally located nuclei in muscle myofibers. Recently, four patients with mild late-onset form have been described, a male with a hemizygous mutation and three females with heterozygous mutations in the MTM1 gene. The muscle biopsies were performed at 13-35 years of age and a new histological marker, the necklace fibers, was described. Here, we report two siblings with the pathogenic c.664 C>T mutation in the MTM1 gene, presenting a severe muscle weakness and respiratory impairment requiring ventilatory support since the first months of life until death, at the age of 36 months and 5 months. In the older brother the muscle biopsy, performed at the age of 30 months, showed almost 100% of necklace fibers, which were not present in the younger one submitted to muscle biopsy at 5 months of age. Our findings confirm the necklace fibers can be a histopathological finding of MTM1 myopathies, even in the severe neonatal form, and suggest that the necklace fibers appear or increase in number over time.


Asunto(s)
Debilidad Muscular/patología , Músculo Esquelético/patología , Miopatías Estructurales Congénitas/patología , Proteínas Tirosina Fosfatasas no Receptoras/genética , Preescolar , Resultado Fatal , Humanos , Lactante , Masculino , Debilidad Muscular/genética , Miopatías Estructurales Congénitas/genética , Hermanos
10.
Int J Biochem Cell Biol ; 43(9): 1402-11, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21689781

RESUMEN

The transfer of cholesterol to mitochondria, which might involve the phosphorylation of proteins, is the rate-limiting step in human placental steroidogenesis. Protein kinase A (PKA) activity and its role in progesterone synthesis by human placental mitochondria were assessed in this study. The results showed that PKA and phosphotyrosine phosphatase D1 are associated with syncytiotrophoblast mitochondrial membrane by an anchoring kinase cAMP protein-121. The ³²P-labeled of four major proteins was analyzed. The specific inhibitor of PKA, H89, decreased progesterone synthesis in mitochondria while in mitochondrial steroidogenic contact sites protein-phosphorylation was diminished, suggesting that PKA plays a role in placental hormone synthesis. In isolated mitochondria, PKA activity was unaffected by the addition of cAMP suggesting a constant activity of this kinase in the syncytiotrophoblast. The presence of PKA and phosphotyrosine phosphatase D1 anchored to mitochondria by an anchoring kinase cAMP protein-121 indicated that syncytiotrophoblast mitochondria contain a full phosphorylation/dephosphorylation system.


Asunto(s)
Proteínas Quinasas Dependientes de AMP Cíclico/antagonistas & inhibidores , Isoquinolinas/farmacología , Mitocondrias/efectos de los fármacos , Placenta/efectos de los fármacos , Progesterona/biosíntesis , Sulfonamidas/farmacología , Trofoblastos/efectos de los fármacos , Proteínas de Anclaje a la Quinasa A/metabolismo , Pruebas de Enzimas , Femenino , Humanos , Mitocondrias/enzimología , Mitocondrias/metabolismo , Consumo de Oxígeno , Fosfoproteínas/metabolismo , Fosforilación , Placenta/citología , Placenta/metabolismo , Embarazo , Estabilidad Proteica , Proteínas Tirosina Fosfatasas no Receptoras/metabolismo , Trofoblastos/enzimología , Trofoblastos/metabolismo
13.
Curr Opin Neurol ; 15(5): 553-61, 2002 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-12351999

RESUMEN

PURPOSE OF REVIEW: Considerable progress has been made in molecular genetic research and in identifying the underlying pathogenesis of congenital myopathies, with implications for genetic counseling. Therefore an overview of such advances in the last two years is most timely and relevant for a more precise delineation of these disorders. RECENT FINDINGS: New mutations have been described on the ryanodine receptor gene, including the carboxyl-terminus region, and experimental models developed to explain their role in central core disease. Phenotype-genotype correlations for nemaline myopathy have improved our understanding of those related to gene mutations. In multi-minicore disease, collaborative studies support genetic heterogeneity and autosomal-recessive inheritance. Research on X-linked myotubular myopathies has revealed a high percentage of mothers of sporadic cases as carriers. Although not initially included within the congenital myopathies, desmin-related or myofibrillar myopathies are described here because they are closely related to other congenital myopathies with intracytoplasmic inclusions. Western blot for myotubularin and desmin has been proposed as a useful diagnostic test for both X-linked myotubular myopathy and desmin-related myopathy, and in-vitro and mouse models for the latter have provided insights into its pathogenesis. Several entities still await genetic characterization. Here we focus on clinical features, inheritance, and molecular genetics. SUMMARY: Advances in immunohistochemistry and molecular genetics in congenital muscular dystrophies have enriched our knowledge of this heterogeneous group of disorders, leading to more accurate classification and differentiation between the various congenital myopathies.


Asunto(s)
Mutación , Miopatías Estructurales Congénitas/genética , Miopatías Estructurales Congénitas/fisiopatología , Canal Liberador de Calcio Receptor de Rianodina/genética , Animales , Desmina/metabolismo , Diagnóstico Diferencial , Genotipo , Humanos , Hialina/metabolismo , Cuerpos de Inclusión/metabolismo , Distrofias Musculares/diagnóstico , Miopatías Nemalínicas/genética , Miopatías Nemalínicas/fisiopatología , Miopatías Estructurales Congénitas/diagnóstico , Miopatía del Núcleo Central/genética , Miopatía del Núcleo Central/fisiopatología , Fenotipo , Proteínas Tirosina Fosfatasas/metabolismo , Proteínas Tirosina Fosfatasas no Receptoras
14.
J Pediatr ; 134(2): 206-14, 1999 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-9931531

RESUMEN

OBJECTIVES: X-linked myotubular myopathy (MTM1) is a rare developmental disorder of skeletal muscle characterized by the presence of central nuclei in biopsy specimens from affected male subjects. Until recently, the disorder was usually fatal within the first year of life. This study was undertaken to determine the outcome in long-term survivors (>1 year of age) with MTM1. METHODS: Clinical data were obtained on 55 male subjects from 49 independent North American families for which a mutation was identified in the X-linked myotubularin gene by direct genomic sequencing. Medical records were reviewed and families were interviewed to ascertain features at birth, length of survival, developmental milestones, and medical complications. RESULTS: Seventy-four percent (26 of 35) of the affected male subjects over the age of 1 year are living (range, 1 to 27 years); 80% remain completely or partially ventilator-dependent. In the absence of significant hypoxia, cognitive development is normal, and the muscle disorder appears nonprogressive. Several patients have had other medical problems not previously reported to be associated with MTM1. These include pyloric stenosis (4 male subjects from 3 families), spherocytosis (2 patients), gallstones (4 patients), kidney stones or nephrocalcinosis (2 patients), a vitamin K responsive bleeding diathesis (2 patients), and height >/=90% for age (40% of the patients). Six patients have had biochemical evidence of liver dysfunction, and 2 patients died after significant liver hemorrhage. CONCLUSIONS: These data suggest that the prognosis for X-linked MTM may not be as poor as previously reported. However, at least some long-term survivors appear at risk for medical complications involving other organ systems, and patients should be carefully monitored for these potentially life-threatening complications. The pleiotropic symptoms demonstrated in these patients strongly suggest that the function of the MTM1 protein is not limited to developing muscle cells.


Asunto(s)
Ligamiento Genético , Enfermedades Musculares/genética , Proteínas Tirosina Fosfatasas/genética , Cromosoma X , Adolescente , Adulto , Niño , Preescolar , Cognición , Enfermedades Gastrointestinales/etiología , Crecimiento , Enfermedades Hematológicas/etiología , Humanos , Lactante , Masculino , Músculo Esquelético/ultraestructura , Enfermedades Musculares/complicaciones , Enfermedades Musculares/fisiopatología , Mutación , Pronóstico , Proteínas Tirosina Fosfatasas no Receptoras , Desempeño Psicomotor , Enfermedades Respiratorias/etiología , Sobrevivientes
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA