Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 892
Filtrar
1.
Am J Physiol Cell Physiol ; 322(3): C382-C394, 2022 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-35044855

RESUMEN

Sarcolipin (SLN) is a small regulatory protein that inhibits the sarco(endo)plasmic reticulum Ca2+-ATPase (SERCA) pump. When bound to SERCA, SLN reduces the apparent Ca2+ affinity of SERCA and uncouples SERCA Ca2+ transport from its ATP consumption. As such, SLN plays a direct role in altering skeletal muscle relaxation and energy expenditure. Interestingly, the expression of SLN is dynamic during times of muscle adaptation, in that large increases in SLN content are found in response to development, atrophy, overload, and disease. Several groups have suggested that increases in SLN, especially in dystrophic muscle, are deleterious as it may reduce muscle function and exacerbate already abhorrent intracellular Ca2+ levels. However, there is also significant evidence to show that increased SLN content is a beneficial adaptive mechanism that protects the SERCA pump and activates Ca2+ signaling and adaptive remodeling during times of cell stress. In this review, we first discuss the role for SLN in healthy muscle during both development and overload, where SLN has been shown to activate Ca2+ signaling to promote mitochondrial biogenesis, fiber-type shifts, and muscle hypertrophy. Then, with respect to muscle disease, we summarize the discrepancies in the literature as to whether SLN upregulation is adaptive or maladaptive in nature. This review is the first to offer the concept of SLN hormesis in muscle disease, wherein both too much and too little SLN are detrimental to muscle health. Finally, the underlying mechanisms which activate SLN upregulation are discussed, specifically acknowledging a potential positive feedback loop between SLN and Ca2+ signaling molecules.


Asunto(s)
Desarrollo de Músculos , Proteínas Musculares/metabolismo , Músculo Esquelético/enzimología , Atrofia Muscular/enzimología , Distrofias Musculares/enzimología , Proteolípidos/metabolismo , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/metabolismo , Animales , Señalización del Calcio , Humanos , Mitocondrias Musculares/metabolismo , Mitocondrias Musculares/patología , Músculo Esquelético/patología , Músculo Esquelético/fisiopatología , Atrofia Muscular/patología , Atrofia Muscular/fisiopatología , Distrofias Musculares/patología , Distrofias Musculares/fisiopatología
2.
Mol Cell ; 79(3): 504-520.e9, 2020 08 06.
Artículo en Inglés | MEDLINE | ID: mdl-32707033

RESUMEN

Protein kinases are essential for signal transduction and control of most cellular processes, including metabolism, membrane transport, motility, and cell cycle. Despite the critical role of kinases in cells and their strong association with diseases, good coverage of their interactions is available for only a fraction of the 535 human kinases. Here, we present a comprehensive mass-spectrometry-based analysis of a human kinase interaction network covering more than 300 kinases. The interaction dataset is a high-quality resource with more than 5,000 previously unreported interactions. We extensively characterized the obtained network and were able to identify previously described, as well as predict new, kinase functional associations, including those of the less well-studied kinases PIM3 and protein O-mannose kinase (POMK). Importantly, the presented interaction map is a valuable resource for assisting biomedical studies. We uncover dozens of kinase-disease associations spanning from genetic disorders to complex diseases, including cancer.


Asunto(s)
Redes Reguladoras de Genes , Enfermedades Genéticas Congénitas/genética , Neoplasias/genética , Proteínas Quinasas/genética , Proteínas Serina-Treonina Quinasas/genética , Proteínas Proto-Oncogénicas/genética , Biología Computacional/métodos , Conjuntos de Datos como Asunto , Regulación de la Expresión Génica , Ontología de Genes , Enfermedades Genéticas Congénitas/enzimología , Enfermedades Genéticas Congénitas/patología , Humanos , Redes y Vías Metabólicas/genética , Anotación de Secuencia Molecular , Distrofias Musculares/enzimología , Distrofias Musculares/genética , Distrofias Musculares/patología , Neoplasias/enzimología , Neoplasias/patología , Enfermedades Neurodegenerativas/enzimología , Enfermedades Neurodegenerativas/genética , Enfermedades Neurodegenerativas/patología , Mapeo de Interacción de Proteínas/métodos , Proteínas Quinasas/química , Proteínas Quinasas/clasificación , Proteínas Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas/química , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/química , Proteínas Proto-Oncogénicas/metabolismo , Transducción de Señal
3.
PLoS One ; 13(5): e0197254, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29758057

RESUMEN

Ankyrin repeat and kinase domain containing 1 (ANKK1) gene has been widely related to neuropsychiatry disorders. The localization of ANKK1 in neural progenitors and its correlation with the cell cycle has suggested its participation in development. However, ANKK1 functions still need to be identified. Here, we have further characterized the ANKK1 localization in vivo and in vitro, by using immunolabeling, quantitative real-time PCR and Western blot in the myogenic lineage. Histologic investigations in mice and humans revealed that ANKK1 is expressed in precursors of embryonic and adult muscles. In mice embryos, ANKK1 was found in migrating myotubes where it shows a polarized cytoplasmic distribution, while proliferative myoblasts and satellite cells show different isoforms in their nuclei and cytoplasm. In vitro studies of ANKK1 protein isoforms along the myogenic progression showed the decline of nuclear ANKK1-kinase until its total exclusion in myotubes. In adult mice, ANKK1 was expressed exclusively in the Fast-Twitch muscles fibers subtype. The induction of glycolytic metabolism in C2C12 cells with high glucose concentration or treatment with berberine caused a significant increase in the ANKK1 mRNA. Similarly, C2C12 cells under hypoxic conditions caused the increase of nuclear ANKK1. These results altogether show a relationship between ANKK1 gene regulation and the metabolism of muscles during development and in adulthood. Finally, we found ANKK1 expression in regenerative fibers of muscles from dystrophic patients. Future studies in ANKK1 biology and the pathological response of muscles will reveal whether this protein is a novel muscle disease biomarker.


Asunto(s)
Células Musculares/enzimología , Músculo Esquelético/enzimología , Proteínas Serina-Treonina Quinasas/metabolismo , Células Madre/enzimología , Adulto , Animales , Hipoxia de la Célula/fisiología , Línea Celular , Núcleo Celular/enzimología , Proliferación Celular , Niño , Citoplasma/enzimología , Femenino , Humanos , Lactante , Masculino , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Persona de Mediana Edad , Células Musculares/citología , Células Musculares/patología , Músculo Esquelético/citología , Músculo Esquelético/crecimiento & desarrollo , Músculo Esquelético/patología , Distrofias Musculares/enzimología , Distrofias Musculares/patología , Células Madre/citología , Células Madre/patología
4.
Pediatrics ; 141(Suppl 5): S510-S514, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29610182

RESUMEN

X-linked recessive mutations in the dystrophin gene are one of the most common causes of inherited neuromuscular disorders in humans. Duchenne muscular dystrophy, the most common phenotype, and Becker muscular dystrophy are often recognizable by certain clinical features; however, less frequent presentations require a higher degree of suspicion. In this article, we describe a series of 6 children (4 boys, 2 girls) referred to a tertiary pediatric neuromuscular clinic for isolated elevated creatine kinase levels (range: 720-7000 IU/L) identified on initial assessment for otherwise unexplained transaminase elevations (n = 2), a social communication disorder (n = 3), and exertional myalgia and/or rhabdomyolysis (n = 1). There was no preceding family history of neuromuscular disease. One boy had an additional history of severe cerebral palsy and cyclical vomiting, and 1 girl had a history of maternal hepatitis C. There was no significant weakness at presentation, and the majority remained stable over a prolonged period of follow-up (age range at last follow-up: 9-16 years). All 6 children were found to carry dystrophin gene mutations resulting in milder phenotypes. This series highlights that dystrophinopathies may not uncommonly present with features distinct from the classic Duchenne muscular dystrophy and Becker muscular dystrophy phenotypes in both boys and girls. Pediatricians should be aware of such atypical presentations to initiate a timely and adequate diagnostic process. Establishing the correct genetic diagnosis of a dystrophinopathy is important to allow appropriate genetic counseling, to implement relevant surveillance and management strategies, and to avoid unnecessary investigations in search of an incorrect alternative diagnosis.


Asunto(s)
Enfermedades Genéticas Ligadas al Cromosoma X/diagnóstico , Distrofias Musculares/diagnóstico , Niño , Preescolar , Creatina Quinasa/sangre , Distrofina/genética , Femenino , Estudios de Seguimiento , Genes Recesivos , Genes Ligados a X , Asesoramiento Genético , Enfermedades Genéticas Ligadas al Cromosoma X/enzimología , Enfermedades Genéticas Ligadas al Cromosoma X/genética , Humanos , Masculino , Distrofias Musculares/enzimología , Distrofias Musculares/genética , Mutación , Fenotipo
5.
Curr Neurovasc Res ; 14(1): 60-64, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-27915985

RESUMEN

Congenital Muscular Dystrophy type 1D (CMD1D) is characterized by an abnormal glycosylation of α-DG (α-dystroglycan) and is associated to the central nervous system (CNS) abnormalities such as cognitive impairment. The purpose of the research was to evaluate the blood-brain barrier permeability (BBB) permeability and matrix metalloproteinases (MMP) -2 and -9 in adult Largemyd-/- mice in order to understand the physiopathology of brain involvement during the CMD1D process. To this aim, we used adult homozygous Largemyd-/- (mutation in Large), heterozygous Largemyd+/- as well as wild-type (C57BL/6) mice. The animals were submitted to the evaluation of BBB permeability and MMP-2 and MMP-9 in striatum, hippocampus and cerebral cortex. There was an increase in BBB permeability in the hippocampus and striatum associated with an increase in the protein levels of MMP-2 in the cerebral cortex and striatum and MMP-9 in the hippocampus in adult Largemyd-/- mice. Our results suggest that the pathophysiologic processes can be associated to the action of MMPs and BBB disruption and that the BBB breakdown is relevant to the perpetuation of brain inflammation and can be related to brain dysfunction observed in CMD1D patients.


Asunto(s)
Barrera Hematoencefálica/enzimología , Encéfalo/anomalías , Discapacidad Intelectual/enzimología , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Distrofias Musculares/enzimología , Animales , Encéfalo/enzimología , Modelos Animales de Enfermedad , Activación Enzimática , Hipocampo/metabolismo , Ratones Endogámicos C57BL , Ratones Transgénicos , Permeabilidad
6.
Int J Biochem Cell Biol ; 79: 469-477, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27458054

RESUMEN

TRIM32 is a member of the TRIpartite Motif family characterised by the presence of an N-terminal three-domain-module that includes a RING domain, which confers E3 ubiquitin ligase activity, one or two B-box domains and a Coiled-Coil region that mediates oligomerisation. Several TRIM32 substrates were identified including muscular proteins and proteins involved in cell cycle regulation and cell motility. As ubiquitination is a versatile post-translational modification that can affect target turnover, sub-cellular localisation or activity, it is likely that diverse substrates may be differentially affected by TRIM32-mediated ubiquitination, reflecting its multi-faceted roles in muscle physiology, cancer and immunity. With particular relevance for muscle physiology, mutations in TRIM32 are associated with autosomal recessive Limb-Girdle Muscular Dystrophy 2H, a muscle-wasting disease with variable clinical spectrum ranging from almost asymptomatic to wheelchair-bound patients. In this review, we will focus on the ability of TRIM32 to mark specific substrates for proteasomal degradation discussing how the TRIM32-proteasome axis may (i) be important for muscle homeostasis and for the pathogenesis of muscular dystrophy; and (ii) define either an oncogenic or tumour suppressive role for TRIM32 in the context of different types of cancer.


Asunto(s)
Distrofias Musculares/enzimología , Neoplasias/enzimología , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Humanos , Músculos/enzimología , Músculos/fisiopatología , Distrofias Musculares/fisiopatología
7.
J Hum Genet ; 61(8): 753-9, 2016 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-27193224

RESUMEN

Protein O-mannosyltransferase 1 (POMT1) is a glycosyltransferase involved in α-dystroglycan glycosylation. POMT1 mutations cause a wide spectrum of clinical conditions from Walker-Warburg syndrome (WWS), which involves muscle, eye and brain abnormalities, to mild forms of limb-girdle muscular dystrophy with mental retardation. We aimed to elucidate the impact of different POMT1 mutations on the clinical phenotype. We report five Chinese patients with POMT1 mutations: one had a typical clinical manifestation of WWS, and the other four were diagnosed with congenital muscular dystrophy with mental retardation of varying severity. We analyzed the influence of the POMT1 mutations on POMT activity by assaying the patients' muscles and cultured skin fibroblasts. We demonstrated different levels of decreased POMT activity that correlated highly with decreased α-dystroglycan glycosylation. Our results suggest that POMT activity is inversely proportional to clinical severity, and demonstrate that skin fibroblasts can be used for differential diagnosis of patients with α-dystroglycanopathies. We have provided clinical, histological, enzymatic and genetic evidence of POMT1 involvement in five unrelated Chinese patients.


Asunto(s)
Estudios de Asociación Genética , Genotipo , Manosiltransferasas/genética , Manosiltransferasas/metabolismo , Mutación , Fenotipo , Adolescente , Niño , Preescolar , Análisis Mutacional de ADN , Ecocardiografía , Activación Enzimática , Femenino , Humanos , Lactante , Imagen por Resonancia Magnética , Masculino , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Distrofias Musculares/diagnóstico , Distrofias Musculares/enzimología , Distrofias Musculares/genética , Embarazo , Diagnóstico Prenatal
8.
Cell Rep ; 14(9): 2209-2223, 2016 Mar 08.
Artículo en Inglés | MEDLINE | ID: mdl-26923585

RESUMEN

Glycosylation is an essential post-translational modification that underlies many biological processes and diseases. α-dystroglycan (α-DG) is a receptor for matrix and synaptic proteins that causes muscular dystrophy and lissencephaly upon its abnormal glycosylation (α-dystroglycanopathies). Here we identify the glycan unit ribitol 5-phosphate (Rbo5P), a phosphoric ester of pentose alcohol, in α-DG. Rbo5P forms a tandem repeat and functions as a scaffold for the formation of the ligand-binding moiety. We show that enzyme activities of three major α-dystroglycanopathy-causing proteins are involved in the synthesis of tandem Rbo5P. Isoprenoid synthase domain-containing (ISPD) is cytidine diphosphate ribitol (CDP-Rbo) synthase. Fukutin and fukutin-related protein are sequentially acting Rbo5P transferases that use CDP-Rbo. Consequently, Rbo5P glycosylation is defective in α-dystroglycanopathy models. Supplementation of CDP-Rbo to ISPD-deficient cells restored α-DG glycosylation. These findings establish the molecular basis of mammalian Rbo5P glycosylation and provide insight into pathogenesis and therapeutic strategies in α-DG-associated diseases.


Asunto(s)
Proteínas de la Membrana/fisiología , Distrofias Musculares/enzimología , Pentosafosfatos/metabolismo , Procesamiento Proteico-Postraduccional , Proteínas/fisiología , Secuencia de Aminoácidos , Conformación de Carbohidratos , Secuencia de Carbohidratos , Glicosilación , Células HEK293 , Humanos , Distrofias Musculares/genética , Mutación , Nucleotidiltransferasas/genética , Pentosiltransferasa
9.
Pediatr Neurol ; 54: 49-54, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26548592

RESUMEN

BACKGROUND: Clinical presentation with motor delay, proximal weakness, and learning difficulties raise the possibility of a dystrophinopathy, dystroglycanopathy, or myotonic dystrophy. This differential should also include the more recently described choline kinase beta-related muscular dystrophy. This condition is typically characterized by large and abnormally distributed mitochondria on muscle biopsy, which can distinguish this condition from the other muscle conditions in the differential. METHODS: We present a boy with choline kinase beta mutations with relatively mild clinical manifestations, including proximal weakness, learning difficulties and elevated creatine kinase. Investigations included muscle magnetic resonance imaging (MRI) with T1 axial sequences through thigh and calves, and needle muscle biopsy of the left vastus lateralis muscle. RESULTS: MRI showed involvement mainly of the quadriceps femoris, sartorius, and adductor magnus, with selective sparing of the gracilis, hamstrings, and adductor longus and brevis. Muscle biopsy revealed chronic dystrophic features. Oxidative stains demonstrated enlarged mitochondria accentuated peripherally or present diffusely in a few fibres giving a coarsely stippled appearance. A homozygous C.722A>G (p.Asn241Ser) mutation was detected in exon 6 of the CHKB gene. CONCLUSION: This selective pattern of skeletal muscle involvement might be helpful for identifying other patients with this condition, even in the absence of diagnostic muscle pathology.


Asunto(s)
Colina Quinasa/genética , Imagen por Resonancia Magnética , Músculo Esquelético/patología , Distrofias Musculares/genética , Distrofias Musculares/patología , Niño , Diagnóstico Diferencial , Humanos , Pierna/patología , Imagen por Resonancia Magnética/métodos , Masculino , Distrofias Musculares/diagnóstico , Distrofias Musculares/enzimología
10.
Dis Markers ; 2015: 543282, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26063958

RESUMEN

BACKGROUND: Differentiation among types of muscular dystrophy (MD) has remained challenging. In this retrospective study, we sought to develop a methodology for differentiation of MD types using analysis of serum enzyme profiles. METHODS: The serum levels of enzymes from 232 patients, including 120 with DMD, 36 with BMD, 36 with FSHD, 46 with LGMD, and 11 with EDMD, were evaluated. RESULTS: The characteristic profiles of serum enzymes facilitated differentiation of these five types of MD. DMD was characterized by simultaneous elevation of ALT, AST, LDH, and ALP; BMD and LGMD were characterized by elevation of ALT, AST, and LDH; and FSHD and EDMD were characterized by a lack of abnormal serum enzyme levels. We further developed discriminant functions to distinguish BMD and LGMD. For LGMD, LGMD2B patients had significantly higher ALP levels than non-LGMD2B patients (98 ± 59 U/L versus 45 ± 9 U/L, resp., p < 0.05). CONCLUSIONS: Our approach enabled the determination of MD subtypes using serum enzyme profiles prior to genetic testing, which will increase the chance a mutation will be found in the first gene analyzed.


Asunto(s)
Alanina Transaminasa/sangre , Aspartato Aminotransferasas/sangre , L-Lactato Deshidrogenasa/sangre , Distrofias Musculares/clasificación , Distrofias Musculares/enzimología , Diagnóstico Diferencial , Femenino , Humanos , Masculino , Distrofias Musculares/sangre , Estudios Retrospectivos
11.
Matrix Biol ; 44-46: 130-7, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25701226

RESUMEN

Dystrophin is a cytosolic protein belonging to a membrane-spanning glycoprotein complex, called dystrophin-glycoprotein complex (DGC) that is expressed in many tissues, especially in skeletal muscle and in the nervous system. The DGC connects the cytoskeleton to the extracellular matrix and, although none of the proteins of the DGC displays kinase or phosphatase activity, it is involved in many signal transduction pathways. Mutations in some components of the DGC are linked to many forms of inherited muscular dystrophies. In particular, a mutation in the dystrophin gene, leading to a complete loss of the protein, provokes one of the most prominent muscular dystrophies, the Duchenne muscular dystrophy, which affects 1 out of 3500 newborn males. What is observed in these circumstances, is a dramatic alteration of the expression levels of a multitude of metalloproteinases (MMPs), a family of extracellular Zn(2+)-dependent endopeptidases, in particular of MMP-2 and MMP-9, also called gelatinases. Indeed, the enzymatic activity of MMP-2 and MMP-9 on dystroglycan, an important member of the DGC, plays a significant role also in physiological processes taking place in the central and peripheral nervous system. This mini-review discusses the role of MMP-2 and MMP-9, in physiological as well as pathological processes involving members of the DGC.


Asunto(s)
Distroglicanos/metabolismo , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Animales , Sistema Nervioso Central/enzimología , Regulación Enzimológica de la Expresión Génica , Humanos , Distrofias Musculares/enzimología , Sistema Nervioso Periférico/enzimología
12.
J Biol Chem ; 289(41): 28138-48, 2014 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-25138275

RESUMEN

Mutations in the LARGE gene have been identified in congenital muscular dystrophy (CMD) patients with brain abnormalities. Both LARGE and its paralog, LARGE2 (also referred to as GYLTL1B) are bifunctional glycosyltransferases with xylosyltransferase (Xyl-T) and glucuronyltransferase (GlcA-T) activities, and are capable of forming polymers consisting of [-3Xyl-α1,3GlcAß1-] repeats. LARGE-dependent modification of α-dystroglycan (α-DG) with these polysaccharides is essential for the ability of α-DG to act as a receptor for ligands in the extracellular matrix. Here we report on the endogenous enzymatic activities of LARGE and LARGE2 in mice and humans, using a newly developed assay for GlcA-T activity. We show that normal mouse and human cultured cells have endogenous LARGE GlcA-T, and that this activity is absent in cells from the Large(myd) (Large-deficient) mouse model of muscular dystrophy, as well as in cells from CMD patients with mutations in the LARGE gene. We also demonstrate that GlcA-T activity is significant in the brain, heart, and skeletal muscle of wild-type and Large2(-/-) mice, but negligible in the corresponding tissues of the Large(myd) mice. Notably, GlcA-T activity is substantial, though reduced, in the kidneys of both the Large(myd) and Large2(-/-) mice, consistent with the observation of α-DG/laminin binding in these contexts. This study is the first to test LARGE activity in samples as small as cryosections and, moreover, provides the first direct evidence that not only LARGE, but also LARGE2, is vital to effective functional modification of α-DG in vivo.


Asunto(s)
Distroglicanos/metabolismo , Glicosiltransferasas/metabolismo , Laminina/metabolismo , Distrofias Musculares/enzimología , N-Acetilglucosaminiltransferasas/metabolismo , Animales , Sitios de Unión , Encéfalo/enzimología , Encéfalo/patología , Células Cultivadas , Niño , Modelos Animales de Enfermedad , Distroglicanos/genética , Pruebas de Enzimas , Femenino , Fibroblastos/enzimología , Fibroblastos/patología , Regulación de la Expresión Génica , Glicosiltransferasas/genética , Humanos , Riñón/enzimología , Riñón/patología , Laminina/genética , Ratones , Ratones Noqueados , Músculo Esquelético/enzimología , Músculo Esquelético/patología , Distrofias Musculares/genética , Distrofias Musculares/patología , Miocardio/enzimología , Miocardio/patología , N-Acetilglucosaminiltransferasas/genética , Especificidad de Órganos , Unión Proteica
13.
Free Radic Biol Med ; 75: 40-7, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25017965

RESUMEN

Although mitochondrial dysfunction and oxidative stress have been proposed to play a crucial role in several types of muscular dystrophy (MD), whether a causal link between these two alterations exists remains an open question. We have documented that mitochondrial dysfunction through opening of the permeability transition pore plays a key role in myoblasts from patients as well as in mouse models of MD, and that oxidative stress caused by monoamine oxidases (MAO) is involved in myofiber damage. In the present study we have tested whether MAO-dependent oxidative stress is a causal determinant of mitochondrial dysfunction and apoptosis in myoblasts from patients affected by collagen VI myopathies. We find that upon incubation with hydrogen peroxide or the MAO substrate tyramine myoblasts from patients upregulate MAO-B expression and display a significant rise in reactive oxygen species (ROS) levels, with concomitant mitochondrial depolarization. MAO inhibition by pargyline significantly reduced both ROS accumulation and mitochondrial dysfunction, and normalized the increased incidence of apoptosis in myoblasts from patients. Thus, MAO-dependent oxidative stress is causally related to mitochondrial dysfunction and cell death in myoblasts from patients affected by collagen VI myopathies, and inhibition of MAO should be explored as a potential treatment for these diseases.


Asunto(s)
Apoptosis/efectos de los fármacos , Mitocondrias/patología , Inhibidores de la Monoaminooxidasa/farmacología , Monoaminooxidasa/biosíntesis , Mioblastos/patología , Adulto , Células Cultivadas , Niño , Preescolar , Colágeno Tipo VI/genética , Humanos , Peróxido de Hidrógeno/farmacología , Monoaminooxidasa/metabolismo , Distrofias Musculares/enzimología , Mioblastos/enzimología , Mioblastos/metabolismo , Estrés Oxidativo/efectos de los fármacos , Pargilina/farmacología , Tiramina/farmacología
15.
J Med Genet ; 51(4): 275-82, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24556084

RESUMEN

BACKGROUND: Congenital muscular dystrophies (CMD) with hypoglycosylation of α-dystroglycan are clinically and genetically heterogeneous disorders that are often associated with brain malformations and eye defects. Presently, 16 proteins are known whose dysfunction impedes glycosylation of α-dystroglycan and leads to secondary dystroglycanopathy. OBJECTIVE: To identify the cause of CMD with secondary merosin deficiency, hypomyelination and intellectual disability in two siblings from a consanguineous family. METHODS: Autozygosity mapping followed by whole exome sequencing and immunochemistry were used to discover and verify a new genetic defect in two siblings with CMD. RESULTS: We identified a homozygous missense mutation (c.325C>T, p.Q109*) in protein O-mannosyl kinase (POMK) that encodes a glycosylation-specific kinase (SGK196) required for function of the dystroglycan complex. The protein was absent from skeletal muscle and skin fibroblasts of the patients. In patient muscle, ß-dystroglycan was normally expressed at the sarcolemma, while α-dystroglycan failed to do so. Further, we detected co-localisation of POMK with desmin at the costameres in healthy muscle, and a substantial loss of desmin from the patient muscle. CONCLUSIONS: Homozygous truncating mutations in POMK lead to CMD with secondary merosin deficiency, hypomyelination and intellectual disability. Loss of desmin suggests that failure of proper α-dystroglycan glycosylation impedes the binding to extracellular matrix proteins and also affects the cytoskeleton.


Asunto(s)
Pérdida Auditiva/complicaciones , Discapacidad Intelectual/complicaciones , Laminina/deficiencia , Distrofias Musculares/congénito , Mutación/genética , Vaina de Mielina/patología , Proteínas Quinasas/genética , Adolescente , Secuencia de Bases , Niño , Preescolar , Análisis Mutacional de ADN , Familia , Femenino , Pérdida Auditiva/enzimología , Pérdida Auditiva/genética , Humanos , Inmunohistoquímica , Lactante , Recién Nacido , Discapacidad Intelectual/enzimología , Discapacidad Intelectual/genética , Masculino , Datos de Secuencia Molecular , Músculo Esquelético/patología , Distrofias Musculares/complicaciones , Distrofias Musculares/enzimología , Distrofias Musculares/genética , Linaje , Adulto Joven
16.
Antioxid Redox Signal ; 20(11): 1673-6, 2014 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-24341409

RESUMEN

Not only double, Janus face, but numerous appearances characterize heme oxygenase-1 (HO-1), an inducible enzyme which main role is to degrade heme. Recently, the noncanonical functions of HO-1 have particularly attracted researchers' attention. Indeed, understanding the enzyme-independent activities of HO-1 can provide additional chances for translational application of research on HO-1. In this Forum, eight reviews and two original articles describe a plethora of mechanisms in which this pleiotropic protein is involved. Further understanding of HO-1 functions is of particular significance for elucidating the pathology of various human diseases and providing rationale for novel therapies.


Asunto(s)
Hemo-Oxigenasa 1/fisiología , Animales , Regulación de la Expresión Génica , Humanos , Distrofias Musculares/tratamiento farmacológico , Distrofias Musculares/enzimología , Neoplasias/tratamiento farmacológico , Neoplasias/enzimología , Neovascularización Patológica/tratamiento farmacológico , Neovascularización Patológica/enzimología
17.
Chembiochem ; 14(18): 2392-402, 2013 Dec 16.
Artículo en Inglés | MEDLINE | ID: mdl-24318691

RESUMEN

Dystroglycanopathies form a subgroup of muscular dystrophies that arise from defects in enzymes that are implicated in the recently elucidated O-mannosylation pathway, thereby resulting in underglycosylation of α-dystroglycan. The emerging identification of additional brain proteins modified by O-mannosylation provides a broader context for interpreting the range of neurological consequences associated with dystroglycanopathies. This form of glycosylation is associated with protein mucin-like domains that present numerous serine and threonine residues as possible sites for modification. Furthermore, the O-Man glycans coexist in this region with O-GalNAc glycans (conventionally associated with such protein sequences), thus resulting in a complex glycoconjugate landscape. Sorting out the relationships between the various molecular defects in glycosylation and the modes of disease presentation, as well as the regulatory interplay among the O-Man glycans and the effects on other modes of glycosylation in the same domain, is challenging. Here we provide a perspective on chemical biology approaches employing synthetic and analytical methods to address these questions.


Asunto(s)
Distroglicanos/metabolismo , Glicoconjugados/metabolismo , Manosa/metabolismo , Distrofias Musculares/metabolismo , Secuencia de Aminoácidos , Animales , Secuencia de Carbohidratos , Distroglicanos/química , Glicoconjugados/química , Glicopéptidos/química , Glicopéptidos/metabolismo , Humanos , Manosa/química , Datos de Secuencia Molecular , Distrofias Musculares/enzimología , Distrofias Musculares/terapia
18.
Curr Opin Neurol ; 26(5): 536-43, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23945283

RESUMEN

PURPOSE OF REVIEW: Recessive mutations in CHKB cause a megaconial congenital muscular dystrophy whose most characteristic feature is mitochondrial enlargement at the periphery of muscle fibers and loss of mitochondria in the center of muscle fibers. This review will summarize clinicopathological features, genetic cause, and biochemical abnormalities of the disease, trying to decipher the mechanism of this complex disorder. RECENT FINDINGS: Since our report of CHKB mutations found in 15 cases with megaconial congenital muscular dystrophy from Japanese, Turkish, and British populations, we have further identified two British and one French patients. One African-American patient has also been reported by another group. All patients have relatively homogenous phenotype although severity varies to some extent. The peculiar distribution pattern of enlarged mitochondria on muscle section seems to be due to a compensatory mechanism after the elimination of functionally defective mitochondria by mitophagy. SUMMARY: CHKB encodes choline kinase ß, an enzyme that catalyzes the first de-novo biosynthetic step of phosphatidylcholine, the most abundant phospholipid in the eukaryotic membrane. The identification of a new muscle disease caused by the defect in phospholipid metabolism will pave the way for a novel biological pathway that connects phospholipid metabolism, mitochondria biology, and muscular dystrophy.


Asunto(s)
Colina Quinasa/genética , Mitocondrias/genética , Distrofias Musculares/enzimología , Mutación/genética , Animales , Colina Quinasa/metabolismo , Humanos , Mitocondrias/enzimología , Distrofias Musculares/congénito , Distrofias Musculares/patología , Fenotipo
19.
Am J Hum Genet ; 92(3): 354-65, 2013 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-23453667

RESUMEN

Mutations in several known or putative glycosyltransferases cause glycosylation defects in α-dystroglycan (α-DG), an integral component of the dystrophin glycoprotein complex. The hypoglycosylation reduces the ability of α-DG to bind laminin and other extracellular matrix ligands and is responsible for the pathogenesis of an inherited subset of muscular dystrophies known as the dystroglycanopathies. By exome and Sanger sequencing we identified two individuals affected by a dystroglycanopathy with mutations in ß-1,3-N-acetylgalactosaminyltransferase 2 (B3GALNT2). B3GALNT2 transfers N-acetyl galactosamine (GalNAc) in a ß-1,3 linkage to N-acetyl glucosamine (GlcNAc). A subsequent study of a separate cohort of individuals identified recessive mutations in four additional cases that were all affected by dystroglycanopathy with structural brain involvement. We show that functional dystroglycan glycosylation was reduced in the fibroblasts and muscle (when available) of these individuals via flow cytometry, immunoblotting, and immunocytochemistry. B3GALNT2 localized to the endoplasmic reticulum, and this localization was perturbed by some of the missense mutations identified. Moreover, knockdown of b3galnt2 in zebrafish recapitulated the human congenital muscular dystrophy phenotype with reduced motility, brain abnormalities, and disordered muscle fibers with evidence of damage to both the myosepta and the sarcolemma. Functional dystroglycan glycosylation was also reduced in the b3galnt2 knockdown zebrafish embryos. Together these results demonstrate a role for B3GALNT2 in the glycosylation of α-DG and show that B3GALNT2 mutations can cause dystroglycanopathy with muscle and brain involvement.


Asunto(s)
Distroglicanos/genética , Distrofias Musculares/genética , Mutación , N-Acetilgalactosaminiltransferasas/genética , Animales , Encéfalo/enzimología , Encéfalo/metabolismo , Línea Celular , Distroglicanos/metabolismo , Retículo Endoplásmico/enzimología , Retículo Endoplásmico/genética , Retículo Endoplásmico/metabolismo , Femenino , Fibroblastos/enzimología , Fibroblastos/metabolismo , Predisposición Genética a la Enfermedad , Glicosilación , Humanos , Lactante , Masculino , Músculo Esquelético/enzimología , Músculo Esquelético/metabolismo , Distrofias Musculares/enzimología , Distrofias Musculares/metabolismo , N-Acetilgalactosaminiltransferasas/metabolismo , Pez Cebra
20.
J Biol Chem ; 287(34): 28526-36, 2012 Aug 17.
Artículo en Inglés | MEDLINE | ID: mdl-22715095

RESUMEN

The severe phenotypic effects of altered glycosylation in the congenital muscular dystrophies, including Walker-Warburg syndrome, muscle-eye-brain disease, Fukuyama congenital muscular dystrophy, and congenital muscular dystrophy 1D, are caused by mutations resulting in altered glycans linked to proteins through O-linked mannose. A glycosyltransferase that branches O-Man, N-acetylglucosaminyltransferase Vb (GnT-Vb), is highly expressed in neural tissues. To understand the expression and function of GnT-Vb, we studied its expression during neuromorphogenesis and generated GnT-Vb null mice. A paralog of GnT-Vb, N-acetylglucosaminyltransferase (GnT-V), is expressed in many tissues and brain, synthesizing N-linked, ß1,6-branched glycans, but its ability to synthesize O-mannosyl-branched glycans is unknown; conversely, although GnT-Vb can synthesize N-linked glycans in vitro, its contribution to their synthesis in vivo is unknown. Our results showed that deleting both GnT-V and GnT-Vb results in the total loss of both N-linked and O-Man-linked ß1,6-branched glycans. GnT-V null brains lacked N-linked, ß1,6-glycans but had normal levels of O-Man ß1,6-branched structures, showing that GnT-Vb could not compensate for the loss of GnT-V. By contrast, GnT-Vb null brains contained normal levels of N-linked ß1,6-glycans but low levels of some O-Man ß1,6-branched glycans. Therefore, GnT-V could partially compensate for GnT-Vb activity in vivo. We found no apparent change in α-dystroglycan binding of glycan-specific antibody IIH6C4 or binding to laminin in GnT-Vb null mice. These results demonstrate that GnT-V is involved in synthesizing branched O-mannosyl glycans in brain, but the function of these branched O-mannosyl structures is unresolved using mice that lack these glycosyltransferases.


Asunto(s)
Encéfalo/enzimología , Regulación Enzimológica de la Expresión Génica , N-Acetilglucosaminiltransferasas/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Polisacáridos/metabolismo , Animales , Glicosilación , Humanos , Ratones , Ratones Noqueados , Distrofias Musculares/enzimología , Distrofias Musculares/genética , N-Acetilglucosaminiltransferasas/genética , Proteínas del Tejido Nervioso/genética , Polisacáridos/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...