Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 265
Filtrar
1.
J Cell Biol ; 220(12)2021 12 06.
Artigo em Inglês | MEDLINE | ID: mdl-34633413

RESUMO

The cavin proteins are essential for caveola biogenesis and function. Here, we identify a role for the muscle-specific component, Cavin4, in skeletal muscle T-tubule development by analyzing two vertebrate systems, mouse and zebrafish. In both models, Cavin4 localized to T-tubules, and loss of Cavin4 resulted in aberrant T-tubule maturation. In zebrafish, which possess duplicated cavin4 paralogs, Cavin4b was shown to directly interact with the T-tubule-associated BAR domain protein Bin1. Loss of both Cavin4a and Cavin4b caused aberrant accumulation of interconnected caveolae within the T-tubules, a fragmented T-tubule network enriched in Caveolin-3, and an impaired Ca2+ response upon mechanical stimulation. We propose a role for Cavin4 in remodeling the T-tubule membrane early in development by recycling caveolar components from the T-tubule to the sarcolemma. This generates a stable T-tubule domain lacking caveolae that is essential for T-tubule function.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas de Membrana/metabolismo , Proteínas Musculares/metabolismo , Músculo Esquelético/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Sarcolema/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Proteínas de Peixe-Zebra/metabolismo , Peixe-Zebra/metabolismo , Animais , Cavéolas/metabolismo , Linhagem Celular , Embrião não Mamífero/metabolismo , Imageamento Tridimensional , Camundongos Endogâmicos C57BL , Fibras Musculares Esqueléticas/metabolismo , Fibras Musculares Esqueléticas/ultraestrutura , Músculo Esquelético/ultraestrutura , Ligação Proteica , Sarcolema/ultraestrutura , Peixe-Zebra/embriologia
2.
Neurogenetics ; 22(2): 117-125, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33811585

RESUMO

We report a multiplex family with extended multisystem neurological phenotype associated with a CRYAB variant. Two affected siblings were evaluated with whole exome sequencing, muscle biopsy, laser microdissection, and mass spectrometry-based proteomic analysis. Both patients and their mother manifested a combination of early-onset cataracts, cardiomyopathy, cerebellar ataxia, optic atrophy, cognitive impairment, and myopathy. Whole exome sequencing identified a heterozygous c.458C>T variant mapped to the C-terminal extension domain of the Alpha-crystallin B chain, disrupting its function as a molecular chaperone and its ability to suppress protein aggregation. In accordance with the molecular findings, muscle biopsies revealed subsarcolemmal deposits that appeared dark with H&E and trichrome staining were negative for the other routine histochemical staining and for amyloid with the Congo-red stain. Electron microscopy demonstrated that the deposits were composed of numerous parallel fibrils. Laser microdissection and mass spectrometry-based proteomic analysis revealed that the inclusions are almost exclusively composed of crystallized chaperones/heat shock proteins. Moreover,  a structural model suggests that Ser153 could be involved in monomer stabilization, dimer association, and possible binding of partner proteins. We propose that our report potentially expands the complex phenotypic spectrum of alpha B-crystallinopathies with possible effect of a CRYAB variant on the central nervous system.


Assuntos
Cardiomiopatia Hipertrófica/genética , Catarata/genética , Ataxia Cerebelar/genética , Disfunção Cognitiva/genética , Atrofia Óptica/genética , Sarcolema/ultraestrutura , Cadeia B de alfa-Cristalina/genética , Sequência de Aminoácidos , Biópsia , Morte Súbita Cardíaca/etiologia , Feminino , Humanos , Corpos de Inclusão/ultraestrutura , Judeus/genética , Masculino , Pessoa de Meia-Idade , Modelos Moleculares , Debilidade Muscular/genética , Músculo Esquelético/patologia , Fenótipo , Conformação Proteica , Sequenciamento do Exoma
3.
PLoS Genet ; 16(11): e1009179, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-33175853

RESUMO

Gene therapy approaches for DMD using recombinant adeno-associated viral (rAAV) vectors to deliver miniaturized (or micro) dystrophin genes to striated muscles have shown significant progress. However, concerns remain about the potential for immune responses against dystrophin in some patients. Utrophin, a developmental paralogue of dystrophin, may provide a viable treatment option. Here we examine the functional capacity of an rAAV-mediated microutrophin (µUtrn) therapy in the mdx4cv mouse model of DMD. We found that rAAV-µUtrn led to improvement in dystrophic histopathology & mostly restored the architecture of the neuromuscular and myotendinous junctions. Physiological studies of tibialis anterior muscles indicated peak force maintenance, with partial improvement of specific force. A fundamental question for µUtrn therapeutics is not only can it replace critical functions of dystrophin, but whether full-length utrophin impacts the therapeutic efficacy of the smaller, highly expressed µUtrn. As such, we found that µUtrn significantly reduced the spacing of the costameric lattice relative to full-length utrophin. Further, immunostaining suggested the improvement in dystrophic pathophysiology was largely influenced by favored correction of fast 2b fibers. However, unlike µUtrn, µdystrophin (µDys) expression did not show this fiber type preference. Interestingly, µUtrn was better able to protect 2a and 2d fibers in mdx:utrn-/- mice than in mdx4cv mice where the endogenous full-length utrophin was most prevalent. Altogether, these data are consistent with the role of steric hindrance between full-length utrophin & µUtrn within the sarcolemma. Understanding the stoichiometry of this effect may be important for predicting clinical efficacy.


Assuntos
Terapia Genética/métodos , Fibras Musculares Esqueléticas/patologia , Distrofia Muscular de Duchenne/terapia , Utrofina/uso terapêutico , Animais , Dependovirus/genética , Modelos Animais de Doenças , Distrofina/genética , Técnicas de Transferência de Genes , Vetores Genéticos/genética , Células HEK293 , Humanos , Camundongos , Camundongos Endogâmicos mdx , Microscopia Eletrônica , Contração Muscular , Fibras Musculares Esqueléticas/citologia , Fibras Musculares Esqueléticas/ultraestrutura , Músculo Esquelético , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/patologia , Junção Neuromuscular/patologia , Junção Neuromuscular/ultraestrutura , Sarcolema/patologia , Sarcolema/ultraestrutura , Utrofina/genética
4.
Int J Mol Sci ; 21(2)2020 Jan 14.
Artigo em Inglês | MEDLINE | ID: mdl-31947691

RESUMO

The arrhythmogenic potential of ß1-adrenoceptor autoantibodies (ß1-AA), as well as antiarrhythmic properties of omega-3 in heart diseases, have been reported while underlying mechanisms are poorly understood. We aimed to test our hypothesis that omega-3 (eicosapentaenoic acid-EPA, docosahexaenoic acid-DHA) may inhibit matrix metalloproteinase (MMP-2) activity to prevent cleavage of ß1-AR and formation of ß1-AA resulting in attenuation of pro-arrhythmic connexin-43 (Cx43) and protein kinase C (PKC) signaling in the diseased heart. We have demonstrated that the appearance and increase of ß1-AA in blood serum of male and female 12-month-old spontaneously hypertensive rats (SHR) was associated with an increase of inducible ventricular fibrillation (VF) comparing to normotensive controls. In contrast, supplementation of hypertensive rats with omega-3 for two months suppressed ß1-AA levels and reduced incidence of VF. Suppression of ß1-AA was accompanied by a decrease of elevated myocardial MMP-2 activity, preservation of cardiac cell membrane integrity and Cx43 topology. Moreover, omega-3 abrogated decline in expression of total Cx43 as well as its phosphorylated forms at serine 368 along with PKC-ε, while decreased pro-fibrotic PKC-δ levels in hypertensive rat heart regardless the sex. The implication of MMP-2 in the action of omega-3 was also demonstrated in cultured cardiomyocytes in which desensitization of ß1-AR due to permanent activation of ß1-AR with isoproterenol was prevented by MMP-2 inhibitor or EPA. Collectively, these data support the notion that omega-3 via suppression of ß1-AA mechanistically controlled by MMP-2 may attenuate abnormal of Cx43 and PKC-ε signaling; thus, abolish arrhythmia substrate and protect rats with an advanced stage of hypertension from malignant arrhythmias.


Assuntos
Antiarrítmicos/farmacologia , Arritmias Cardíacas/etiologia , Autoanticorpos/imunologia , Autoantígenos/imunologia , Ácidos Graxos Ômega-3/farmacologia , Hipertensão/complicações , Receptores Adrenérgicos beta 1/imunologia , Animais , Arritmias Cardíacas/tratamento farmacológico , Arritmias Cardíacas/metabolismo , Arritmias Cardíacas/fisiopatologia , Biomarcadores , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Membrana Celular/ultraestrutura , Conexina 43/metabolismo , Modelos Animais de Doenças , Suscetibilidade a Doenças , Ácidos Graxos Ômega-3/metabolismo , Feminino , Masculino , Metaloproteinase 2 da Matriz/metabolismo , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/ultraestrutura , Proteína Quinase C-épsilon/metabolismo , Ratos , Ratos Endogâmicos SHR , Sarcolema/metabolismo , Sarcolema/ultraestrutura , Fibrilação Ventricular/tratamento farmacológico , Fibrilação Ventricular/etiologia , Fibrilação Ventricular/fisiopatologia
5.
Metabolism ; 89: 18-26, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30253140

RESUMO

BACKGROUND: Skeletal muscle mitochondrial content and function appear to be altered in obesity. Mitochondria in muscle are found in well-defined regions within cells, and they are arranged in a way that form distinct subpopulations of subsarcolemmal (SS) and intermyofibrillar (IMF) mitochondria. We sought to investigate differences in the proteomes of SS and IMF mitochondria between lean subjects and subjects with obesity. METHODS: We performed comparative proteomic analyses on SS and IMF mitochondria isolated from muscle samples obtained from lean subjects and subjects with obesity. Mitochondria were isolated using differential centrifugation, and proteins were subjected to label-free quantitative tandem mass spectrometry analyses. Collected data were evaluated for abundance of mitochondrial proteins using spectral counting. The Reactome pathway database was used to determine metabolic pathways that are altered in obesity. RESULTS: Among proteins, 73 and 41 proteins showed different (mostly lower) expression in subjects with obesity in the SS and IMF mitochondria, respectively (false discovery rate-adjusted P ≤ 0.05). We specifically found an increase in proteins forming the tricarboxylic acid cycle and electron transport chain (ETC) complex II, but a decrease in proteins forming protein complexes I and III of the ETC and adenosine triphosphate (ATP) synthase in subjects with obesity in the IMF, but not SS, mitochondria. Obesity was associated with differential effects on metabolic pathways linked to protein translation in the SS mitochondria and ATP formation in the IMF mitochondria. CONCLUSIONS: Obesity alters the expression of mitochondrial proteins regulating key metabolic processes in skeletal muscle, and these effects are distinct to mitochondrial subpopulations located in different regions of the muscle fibers. TRIAL REGISTRATION: ClinicalTrials.gov (NCT01824173).


Assuntos
Mitocôndrias Musculares/ultraestrutura , Proteínas Mitocondriais/metabolismo , Obesidade/metabolismo , Complexos de ATP Sintetase/metabolismo , Adulto , Feminino , Voluntários Saudáveis , Humanos , Masculino , Redes e Vias Metabólicas , Mitocôndrias Musculares/metabolismo , Mitocôndrias Musculares/patologia , Fibras Musculares Esqueléticas/metabolismo , Fibras Musculares Esqueléticas/ultraestrutura , Obesidade/patologia , Proteômica , Sarcolema/metabolismo , Sarcolema/ultraestrutura , Frações Subcelulares/metabolismo , Frações Subcelulares/ultraestrutura , Espectrometria de Massas em Tandem
6.
Dis Model Mech ; 11(4)2018 04 16.
Artigo em Inglês | MEDLINE | ID: mdl-29666144

RESUMO

Skeletal muscle atrophy is the most prominent feature of amyotrophic lateral sclerosis (ALS), an adult-onset neurodegenerative disease of motor neurons. However, the contribution of skeletal muscle to disease progression remains elusive. Our previous studies have shown that intrathecal injection of cerebrospinal fluid from sporadic ALS patients (ALS-CSF) induces several degenerative changes in motor neurons and glia of neonatal rats. Here, we describe various pathologic events in the rat extensor digitorum longus muscle following intrathecal injection of ALS-CSF. Adenosine triphosphatase staining and electron microscopic (EM) analysis revealed significant atrophy and grouping of type 2 fibres in ALS-CSF-injected rats. Profound neuromuscular junction (NMJ) damage, such as fragmentation accompanied by denervation, were revealed by α-bungarotoxin immunostaining. Altered expression of key NMJ proteins, rapsyn and calpain, was also observed by immunoblotting. In addition, EM analysis showed sarcolemmal folding, Z-line streaming, structural alterations of mitochondria and dilated sarcoplasmic reticulum. The expression of trophic factors was affected, with significant downregulation of vascular endothelial growth factor (VEGF), marginal reduction in insulin-like growth factor-1 (IGF-1), and upregulation of brain-derived neurotrophic factor (BDNF) and glial-derived neurotrophic factor (GDNF). However, motor neurons might be unable to harness the enhanced levels of BDNF and GDNF, owing to impaired NMJs. We propose that ALS-CSF triggers motor neuronal degeneration, resulting in pathological changes in the skeletal muscle. Muscle damage further aggravates the motor neuronal pathology, because of the interdependency between them. This sets in a vicious cycle, leading to rapid and progressive loss of motor neurons, which could explain the relentless course of ALS.This article has an associated First Person interview with the first author of the paper.


Assuntos
Esclerose Lateral Amiotrófica/líquido cefalorraquidiano , Músculo Esquelético/patologia , Esclerose Lateral Amiotrófica/patologia , Animais , Animais Recém-Nascidos , Atrofia , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Modelos Animais de Doenças , Regulação para Baixo , Fator Neurotrófico Derivado de Linhagem de Célula Glial/metabolismo , Humanos , Fator de Crescimento Insulin-Like I/metabolismo , Junção Neuromuscular/patologia , Estresse Oxidativo , Ratos Wistar , Sarcolema/patologia , Sarcolema/ultraestrutura , Regulação para Cima , Fator A de Crescimento do Endotélio Vascular/metabolismo
7.
Muscle Nerve ; 58(2): 286-292, 2018 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-29603301

RESUMO

INTRODUCTION: GNE myopathy is an adult-onset muscle disorder characterized by impaired sialylation of (muscle) glycans, detectable by lectin histochemistry. We describe a standardized method to quantify (lectin-) fluorescence in muscle sections, applicable for diagnosis and response to therapy for GNE myopathy. METHODS: Muscle sections were fluorescently labeled with the sialic acid-binding Sambucus nigra agglutinin (SNA) lectin and antibodies to sarcolemma residence protein caveolin-3 (CAV-3). Entire tissue sections were imaged in tiles and fluorescence was quantified. RESULTS: SNA fluorescence co-localizing with CAV-3 was ∼50% decreased in GNE myopathy biopsies compared with muscle-matched controls, confirming previous qualitative results. DISCUSSION: This quantitative fluorescence method can accurately determine sialylation status of GNE myopathy muscle biopsies. This method is adaptable for expression of other membrane-associated muscle proteins, and may be of benefit for disorders in which therapeutic changes in expression are subtle and difficult to assess by other methods. Muscle Nerve 58: 286-292, 2018.


Assuntos
Miopatias Distais/patologia , Lectinas , Músculo Esquelético/patologia , Adulto , Caveolina 3/genética , Miopatias Distais/genética , Feminino , Corantes Fluorescentes , Humanos , Processamento de Imagem Assistida por Computador , Masculino , Microscopia Confocal , Pessoa de Meia-Idade , Lectinas de Plantas , Proteínas Inativadoras de Ribossomos , Sarcolema/patologia , Sarcolema/ultraestrutura
8.
J Cell Sci ; 130(5): 841-852, 2017 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-28104817

RESUMO

The multi-C2 domain protein dysferlin localizes to the plasma membrane and the T-tubule system in skeletal muscle; however, its physiological mode of action is unknown. Mutations in the DYSF gene lead to autosomal recessive limb-girdle muscular dystrophy type 2B and Miyoshi myopathy. Here, we show that dysferlin has membrane tubulating capacity and that it shapes the T-tubule system. Dysferlin tubulates liposomes, generates a T-tubule-like membrane system in non-muscle cells, and links the recruitment of phosphatidylinositol 4,5-bisphosphate to the biogenesis of the T-tubule system. Pathogenic mutant forms interfere with all of these functions, indicating that muscular wasting and dystrophy are caused by the dysferlin mutants' inability to form a functional T-tubule membrane system.


Assuntos
Proteínas de Membrana/metabolismo , Proteínas Musculares/metabolismo , Distrofias Musculares/metabolismo , Sarcolema/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Células COS , Cálcio/metabolismo , Caveolina 3/metabolismo , Chlorocebus aethiops , Dinaminas/metabolismo , Disferlina , Células HeLa , Humanos , Proteínas de Membrana/deficiência , Camundongos Knockout , Proteínas Musculares/deficiência , Distrofias Musculares/patologia , Proteínas do Tecido Nervoso/metabolismo , Fenótipo , Fosfatidilinositol 4,5-Difosfato/metabolismo , Condicionamento Físico Animal , Ligação Proteica , Sarcolema/ultraestrutura , Proteínas Supressoras de Tumor/metabolismo
9.
J Struct Biol ; 194(3): 375-82, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-27016283

RESUMO

The 30kDa N-BAR domain of the human Bin1 protein is essential for the generation of skeletal muscle T-tubules. By electron cryo-microscopy and electron cryo-tomography with a direct electron detector, we found that Bin1-N-BAR domains assemble into scaffolds of low long-range order that form flexible membrane tubules. The diameter of the tubules closely matches the curved shape of the N-BAR domain, which depends on the composition of the target membrane. These insights are fundamental to our understanding of T-tubule formation and function in human skeletal muscle.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/química , Proteínas Nucleares/química , Domínios Proteicos/fisiologia , Multimerização Proteica , Sarcolema/ultraestrutura , Proteínas Supressoras de Tumor/química , Microscopia Crioeletrônica , Humanos , Proteínas de Membrana/metabolismo , Membranas/ultraestrutura , Músculo Esquelético/química , Músculo Esquelético/ultraestrutura , Tomografia
10.
J Neuropathol Exp Neurol ; 75(2): 148-55, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26733584

RESUMO

Cylindrical spirals (CSs) are rare but distinct subsarcolemmal accumulations in skeletal muscle fibers. To date, CSs have been reported in only 16 patients with a variety of neuromuscular conditions. The origin and composition of CSs are unknown, although there are some morphologic similarities between CSs and tubular aggregates (TAs). To clarify the nature of CSs, we characterized the sarcoplasmic reticulum (SR) and other intracellular membrane system proteins in CSs of muscle biopsies from 2 adult Chinese siblings. Immunohistochemical studies revealed subsarcolemmal immunoreactivity for sarco/endoplasmic reticulum Ca2þ-ATPase 1 (SERCA 1) in the longitudinal SR, but no immunoreactivity for calsequestrin in the terminal cisternae or type 1 ryanodine receptor (RYR1) in the junctional SR. Muscles biopsied from 2 patients with TAs showed immunoreactivity not only for SERCA1 but also for other SR proteins, including calsequestrin and RYR1. CSs exhibited no immunoreactivity for the Golgi apparatus marker GM130, the nuclear membrane emerin, desmin, the autophagosome marker LC3, the lysosomal membrane marker LAMP2, dystrophin, or myosin. Our results suggest CSs may originate only from the longitudinal SR, whereas TAs are composed of both the junctional and longitudinal SR. Immunochemical staining with antibodies against calsequestrin and RYR1 help to distinguish these 2 pathological alterations.


Assuntos
Fibras Musculares Esqueléticas/ultraestrutura , Músculo Esquelético/ultraestrutura , Sarcolema/ultraestrutura , Retículo Sarcoplasmático/ultraestrutura , Adulto , Povo Asiático , Canais de Cálcio/biossíntese , Canais de Cálcio/genética , Moléculas de Adesão Celular/biossíntese , Moléculas de Adesão Celular/genética , Feminino , Humanos , Imuno-Histoquímica , Masculino , Proteínas de Membrana/biossíntese , Proteínas de Membrana/genética , Pessoa de Meia-Idade , Fibras Musculares Esqueléticas/patologia , Músculo Esquelético/patologia , Proteínas de Neoplasias/biossíntese , Proteínas de Neoplasias/genética , Doenças Neuromusculares/genética , Doenças Neuromusculares/patologia , Proteína ORAI1 , Sarcolema/genética , Sarcolema/patologia , Retículo Sarcoplasmático/genética , Retículo Sarcoplasmático/patologia , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/biossíntese , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/genética , Molécula 1 de Interação Estromal , Molécula 2 de Interação Estromal
11.
Neuropathology ; 35(6): 575-81, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26094647

RESUMO

Distal myopathies are a group of clinically and pathologically overlapping muscle diseases that are genetically complex and can represent a diagnostic challenge. Laing early-onset distal myopathy (MPD1) is a form of distal myopathy caused by mutations in the MYH7 gene, which encodes the beta myosin heavy chain protein expressed in type 1 skeletal muscle fibers and cardiac myocytes. Here, we present a case of genetically confirmed MPD1 with a typical clinical presentation but distinctive light microscopic and ultrastructural findings on muscle biopsy. A 39-year-old professional male cellist presented with a bilateral foot drop that developed by age 8; analysis of the family pedigree showed an autosomal dominant pattern of inheritance. The physical exam demonstrated bilateral weakness of ankle dorsiflexors, toe extensors and finger extensors; creatine kinase level was normal. Biopsy of the quadriceps femoris muscle showed predominance and hypotrophy of type 1 fibers, hybrid fibers with co-expression of slow and fast myosin proteins (both in highly atrophic and normal size range), moth-eaten fibers and mini-cores, lack of rimmed vacuoles and rare desmin-positive eosinophilic sarcoplasmic inclusions. In addition to these abnormalities often observed in MPD1, the biopsy demonstrated frequent clefted fibers with complex sarcolemmal invaginations; on ultrastructural examination, these structures closely mimicked myotendinous junctions but were present away from the tendon and were almost exclusively found in type 1 fibers. Sequencing analysis of the MYH7 gene in the index patient and other affected family members demonstrated a previously described heterozygous c.4522_4524delGAG (p.Glu1508del) mutation. This case widens the pathologic spectrum of MPD1 and highlights the pathologic and clinical variability that can accompany the same genetic mutation, suggesting a significant role for modifier genes in MPD1 pathogenesis.


Assuntos
Miopatias Distais/patologia , Sarcolema/ultraestrutura , Adulto , Humanos , Masculino , Microscopia Eletrônica de Transmissão , Músculo Esquelético/ultraestrutura , Linhagem
12.
ABCD (São Paulo, Impr.) ; 28(1): 28-31, 2015. graf
Artigo em Inglês | LILACS | ID: lil-742748

RESUMO

BACKGROUND: The surgical treatment of advanced megaesophagus has no consensus, being esophagectomy the more commonly used method. Since it has high morbimortality - inconvenient for benign disease -, in recent years an alternative has been introduced: the esophageal mucosal resection. AIM: To compare early and late results of the two techniques evaluating the operative time, length of ICU stay; postoperative hospitalization; total hospitalization; intra- and postoperative complication rates; mortality; and long-term results. METHODS: Were evaluated retrospectively 40 charts, 23 esophagectomies and 17 mucosectomies. In assessing postoperative results, interviews were conducted by using a specific questionnaire. RESULTS: Comparing the means of esophagectomy and mucosal resection, respectively, the data were: 1) surgical time - 310.2 min and 279.7 min (p> 0.05); 2) length of stay in ICU - 5 days and 2.53 days (p <0.05); 3) total time of hospitalization - 24.25 days and 20.76 days (p> 0.05); 4) length of hospital stay after surgery - 19.05 days and 14.94 days (p> 0.05); 5) presence of intraoperative complications - 65% and 18% (p <0.05); 6) the presence of postoperative complications - 65% and 35% (p> 0.05). In the assessment of late postoperative score (range 0-10) esophagectomy (n = 5) obtained 8.8 points and 8.8 points also got mucosal resection (n = 5). CONCLUSIONS: Esophageal mucosal resection proved to be good alternative for surgical treatment of megaesophagus. It was advantageous in the immediate postoperative period by presenting a lower average time in operation, the total hospitalization, ICU staying and complications rate. In the late postoperative period, the result was excellent and good in both operations. .


RACIONAL: O tratamento cirúrgico do megaesôfago avançado não é consensual sendo mais comumente usada a esofagectomia. Por tratar-se de técnica que apresenta maior morbimortalidade e empregada em doença benigna, foi introduzida nos últimos anos, como alternativa, a mucosectomia esofágica. OBJETIVO: Comparar os resultados imediatos e tardios das duas técnicas avaliando-se os tempos operatório, de internação em UTI, de internação do pós-operatório, de internação total; taxas de complicações intra-operatórias e pós-operatórias; taxa de mortalidade; e resultados a longo prazo. MÉTODOS: Foram avaliados 40 prontuários, retrospectivamente, sendo 23 esofagectomias e 17 mucosectomias. Na avaliação dos resultados pós-operatórios, foram realizadas entrevistas, mediante uso de questionário específico. RESULTADOS: Comparando-se as médias da esofagectomia e mucosectomia, respectivamente, os dados foram: 1) tempo cirúrgico - 310,2 min e 279,7 min (p>0,05); 2) tempo de internação em UTI - 5 dias e 2,53 dias (p<0,05); 3) tempo de internação total - 24,25 dias e 20,76 dias (p>0,05); 4) tempo de internação após a operação - 19,05 dias e 14,94 dias (p>0,05); 5) presença de complicações intra-operatórias - 65% e 18% (p<0,05); 6) presença de complicações pós-operatórias imediatas - 65% e 35% (p>0,05). Na avaliação do escore pós-operatório tardio (escala 0-10) a esofagectomia (n=5) obteve 8,8 pontos e também 8,8 pontos obteve a mucosectomia (n=5). CONCLUSÕES: A mucosectomia esofágica mostrou-se boa alternativa no tratamento cirúrgico do megaesôfago avançado. Foi vantajosa no pós-operatório imediato por apresentar menor média de tempo na operação, na internação total, na UTI e no índice de complicações. No pós-operatório tardio, o resultado foi excelente e bom nas duas operações. .


Assuntos
Animais , Masculino , Camundongos , Metabolismo Energético , /metabolismo , Insulina/metabolismo , Contração Miocárdica/fisiologia , Miócitos Cardíacos/metabolismo , Transdução de Sinais/fisiologia , Hipóxia/metabolismo , Células Cultivadas , Clatrina/metabolismo , /genética , Camundongos Transgênicos , Miócitos Cardíacos/citologia , Transporte Proteico/fisiologia , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Sarcolema/metabolismo , Sarcolema/ultraestrutura
13.
J Am Heart Assoc ; 3(4)2014 Aug 21.
Artigo em Inglês | MEDLINE | ID: mdl-25146704

RESUMO

BACKGROUND: Chronic hypersecretion of the pancreatic hormone amylin is common in humans with obesity or prediabetic insulin resistance and induces amylin aggregation and proteotoxicity in the pancreas. We recently showed that hyperamylinemia also affects the cardiovascular system. Here, we investigated whether amylin aggregates interact directly with cardiac myocytes and whether controlling hyperamylinemia protects the heart. METHODS AND RESULTS: By Western blot, we found abundant amylin aggregates in lysates of cardiac myocytes from obese patients, but not in controls. Aggregated amylin was elevated in failing hearts, suggesting a role in myocyte injury. Using rats overexpressing human amylin in the pancreas (HIP rats) and control myocytes incubated with human amylin, we show that amylin aggregation at the sarcolemma induces oxidative stress and Ca(2+) dysregulation. In time, HIP rats developed cardiac hypertrophy and left-ventricular dilation. We then tested whether metabolites with antiaggregation properties, such as eicosanoid acids, limit myocardial amylin deposition. Rats were treated with an inhibitor of soluble epoxide hydrolase, the enzyme that degrades endogenous eicosanoids. Treatment doubled the blood concentration of eicosanoids, which drastically reduced incorporation of aggregated amylin in cardiac myocytes and blood cells, without affecting pancreatic amylin secretion. Animals in the treated group showed reduced cardiac hypertrophy and left-ventricular dilation. The cardioprotective mechanisms included the mitigation of amylin-induced cardiac oxidative stress and Ca(2+) dysregulation. CONCLUSIONS: The results suggest blood amylin as a novel therapeutic target in diabetic heart disease and elevating blood levels of antiaggregation metabolites as a pharmacological strategy to reduce amylin aggregation and amylin-mediated cardiotoxicity.


Assuntos
Cardiomegalia/genética , Diabetes Mellitus Tipo 2/genética , Eicosanoides/metabolismo , Células Secretoras de Insulina/metabolismo , Polipeptídeo Amiloide das Ilhotas Pancreáticas/genética , Miócitos Cardíacos/metabolismo , Estado Pré-Diabético/genética , Adamantano/análogos & derivados , Adamantano/farmacologia , Animais , Animais Geneticamente Modificados , Cálcio/metabolismo , Cardiomegalia/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Modelos Animais de Doenças , Epóxido Hidrolases/antagonistas & inibidores , Coração/efeitos dos fármacos , Humanos , Polipeptídeo Amiloide das Ilhotas Pancreáticas/efeitos dos fármacos , Polipeptídeo Amiloide das Ilhotas Pancreáticas/ultraestrutura , Miocárdio/metabolismo , Miócitos Cardíacos/efeitos dos fármacos , Obesidade , Estresse Oxidativo/efeitos dos fármacos , Pâncreas/metabolismo , Estado Pré-Diabético/metabolismo , Ratos , Sarcolema/ultraestrutura , Ureia/análogos & derivados , Ureia/farmacologia
14.
J Struct Biol ; 188(1): 55-60, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25160725

RESUMO

The ventricular human myocyte is spatially organized for optimal ATP and Ca(2+) delivery to sarcomeric myosin and ionic pumps during every excitation-contraction cycle. Comprehension of three-dimensional geometry of the tightly packed ultrastructure has been derived from discontinuous two-dimensional images, but has never been precisely reconstructed or analyzed in human myocardium. Using a focused ion beam scanning electron microscope, we created nanoscale resolution serial images to quantify the three-dimensional ultrastructure of a human left ventricular myocyte. Transverse tubules (t-tubule), lipid droplets, A-bands, and mitochondria occupy 1.8, 1.9, 10.8, and 27.9% of the myocyte volume, respectively. The complex t-tubule system has a small tortuosity (1.04±0.01), and is composed of long transverse segments with diameters of 317±24nm and short branches. Our data indicates that lipid droplets located well beneath the sarcolemma are proximal to t-tubules, where 59% (13 of 22) of lipid droplet centroids are within 0.50µm of a t-tubule. This spatial association could have an important implication in the development and treatment of heart failure because it connects two independently known pathophysiological alterations, a substrate switch from fatty acids to glucose and t-tubular derangement.


Assuntos
Ventrículos do Coração/ultraestrutura , Células Musculares/ultraestrutura , Miocárdio/ultraestrutura , Miócitos Cardíacos/ultraestrutura , Trifosfato de Adenosina/metabolismo , Cálcio/metabolismo , Ventrículos do Coração/metabolismo , Humanos , Imageamento Tridimensional , Gotículas Lipídicas/metabolismo , Gotículas Lipídicas/ultraestrutura , Microscopia Eletrônica de Varredura , Células Musculares/metabolismo , Miócitos Cardíacos/metabolismo , Sarcolema/ultraestrutura
15.
PLoS One ; 9(6): e100513, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24949957

RESUMO

CD97 is a widely expressed adhesion class G-protein-coupled receptor (aGPCR). Here, we investigated the presence of CD97 in normal and malignant human skeletal muscle as well as the ultrastructural and functional consequences of CD97 deficiency in mice. In normal human skeletal muscle, CD97 was expressed at the peripheral sarcolemma of all myofibers, as revealed by immunostaining of tissue sections and surface labeling of single myocytes using flow cytometry. In muscle cross-sections, an intracellular polygonal, honeycomb-like CD97-staining pattern, typical for molecules located in the T-tubule or sarcoplasmatic reticulum (SR), was additionally found. CD97 co-localized with SR Ca2+-ATPase (SERCA), a constituent of the longitudinal SR, but not with the receptors for dihydropyridine (DHPR) or ryanodine (RYR), located in the T-tubule and terminal SR, respectively. Intracellular expression of CD97 was higher in slow-twitch compared to most fast-twitch myofibers. In rhabdomyosarcomas, CD97 was strongly upregulated and in part more N-glycosylated compared to normal skeletal muscle. All tumors were strongly CD97-positive, independent of the underlying histological subtype, suggesting high sensitivity of CD97 for this tumor. Ultrastructural analysis of murine skeletal myofibers confirmed the location of CD97 in the SR. CD97 knock-out mice had a dilated SR, resulting in a partial increase in triad diameter yet not affecting the T-tubule, sarcomeric, and mitochondrial structure. Despite these obvious ultrastructural changes, intracellular Ca2+ release from single myofibers, force generation and fatigability of isolated soleus muscles, and wheel-running capacity of mice were not affected by the lack of CD97. We conclude that CD97 is located in the SR and at the peripheral sarcolemma of human and murine skeletal muscle, where its absence affects the structure of the SR without impairing skeletal muscle function.


Assuntos
Antígenos CD/biossíntese , Músculo Esquelético/metabolismo , Rabdomiossarcoma/genética , Retículo Sarcoplasmático/metabolismo , Animais , Antígenos CD/genética , Canais de Cálcio Tipo L/metabolismo , Regulação Neoplásica da Expressão Gênica , Humanos , Camundongos , Camundongos Knockout , Músculo Esquelético/patologia , Músculo Esquelético/ultraestrutura , Receptores Acoplados a Proteínas G , Rabdomiossarcoma/patologia , Rianodina/metabolismo , Canal de Liberação de Cálcio do Receptor de Rianodina/genética , Canal de Liberação de Cálcio do Receptor de Rianodina/metabolismo , Sarcolema/metabolismo , Sarcolema/patologia , Sarcolema/ultraestrutura , Retículo Sarcoplasmático/patologia , Retículo Sarcoplasmático/ultraestrutura
16.
Biochem Biophys Res Commun ; 447(2): 334-40, 2014 May 02.
Artigo em Inglês | MEDLINE | ID: mdl-24721425

RESUMO

Parkinson's disease (PD), defined as a neurodegenerative disorder, is characterized by the loss of dopaminergic neurons in the substantia nigra in the midbrain. Loss-of-function mutations in the parkin gene are a major cause of autosomal recessive, early-onset PD. Parkin has been implicated in the maintenance of healthy mitochondria, although previous studies show conflicting findings regarding mitochondrial abnormalities in fibroblasts from patients harboring parkin-null mutations. The aim of the present study was to determine whether South African PD patients with parkin mutations exhibit evidence for mitochondrial dysfunction. Fibroblasts were cultured from skin biopsies obtained from three patients with homozygous parkin-null mutations, two heterozygous mutation carriers and two wild-type controls. Muscle biopsies were obtained from two of the patients. The muscle fibers showed subtle abnormalities such as slightly swollen mitochondria in focal areas of the fibers and some folding of the sarcolemma. Although no differences in the degree of mitochondrial network branching were found in the fibroblasts, ultrastructural abnormalities were observed including the presence of electron-dense vacuoles. Moreover, decreased ATP levels which are consistent with mitochondrial dysfunction were observed in the patients' fibroblasts compared to controls. Remarkably, these defects did not manifest in one patient, which may be due to possible compensatory mechanisms. These results suggest that parkin-null patients exhibit features of mitochondrial dysfunction. Involvement of mitochondria as a key role player in PD pathogenesis will have important implications for the design of new and more effective therapies.


Assuntos
Mitocôndrias/enzimologia , Mitocôndrias/ultraestrutura , Doença de Parkinson/genética , Doença de Parkinson/patologia , Ubiquitina-Proteína Ligases/genética , Trifosfato de Adenosina/metabolismo , Fibroblastos/enzimologia , Fibroblastos/ultraestrutura , Humanos , Fibras Musculares Esqueléticas/ultraestrutura , Músculo Esquelético/ultraestrutura , Mutação , Sarcolema/ultraestrutura
17.
J Mol Neurosci ; 50(2): 339-44, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23479141

RESUMO

Limb girdle muscular dystrophy type 2 G (LGMD2G) is caused by mutations in the telethonin gene. Only few families were described presenting this disease, and they are mainly Brazilians. Here, we identified one additional case carrying the same common c.157C > T mutation in the telethonin gene but with an atypical histopathological muscle pattern. In a female patient with a long duration of symptoms (46 years), muscle biopsy showed, in addition to telethonin deficiency, the presence of nemaline rods, type 1 fiber predominance, nuclear internalization, lobulated fibers, and mitochondrial paracrystalline inclusions. Her first clinical signs were identified at 8 years old, which include tiptoe walking, left lower limb deformity, and frequent falls. Ambulation loss occurred at 41 years old, and now, at 54 years old, she presented pelvic girdle atrophy, winging scapula, foot deformity with incapacity to perform ankle dorsiflexion, and absent tendon reflexes. The presence of nemaline bodies could be a secondary phenomenon, possibly associated with focal Z-line abnormalities of a long-standing disease. However, these new histopathological findings, characteristic of congenital myopathies, expand muscle phenotypic variability of telethoninopathy.


Assuntos
Fibras Musculares Esqueléticas/patologia , Distrofia Muscular do Cíngulo dos Membros/genética , Fenótipo , Criança , Conectina/genética , Feminino , Humanos , Mitocôndrias/ultraestrutura , Distrofia Muscular do Cíngulo dos Membros/diagnóstico , Polimorfismo de Nucleotídeo Único , Sarcolema/ultraestrutura
18.
Eur J Appl Physiol ; 113(5): 1189-98, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23129090

RESUMO

Decompression sickness is initiated by gas bubbles formed during decompression, and it has been generally accepted that exercise before decompression causes increased bubble formation. There are indications that exercise-induced muscle injury seems to be involved. Trauma-induced skeletal muscle injury and vigorous exercise that could theoretically injure muscle tissues before decompression have each been shown to result in profuse bubble formation. Based on these findings, we hypothesized that exercise-induced skeletal muscle injury prior to decompression from diving would cause increase of vascular bubbles and lower survival rates after decompression. In this study, we examined muscle injury caused by eccentric exercise in rats prior to simulated diving and we observed the resulting bubble formation. Female Sprague-Dawley rats (n = 42) ran downhill (-16º) for 100 min on a treadmill followed by 90 min rest before a 50-min simulated saturation dive (709 kPa) in a pressure chamber. Muscle injury was evaluated by immunohistochemistry and qPCR, and vascular bubbles after diving were detected by ultrasonic imaging. The exercise protocol resulted in increased mRNA expression of markers of muscle injury; αB-crystallin, NF-κB, and TNF-α, and myofibrillar disruption with preserved sarcolemmal integrity. Despite evident myofibrillar disruption after eccentric exercise, no differences in bubble amounts or survival rates were observed in the exercised animals as compared to non-exercised animals after diving, a novel finding that may be applicable to humans.


Assuntos
Doença da Descompressão/sangue , Mergulho/fisiologia , Miofibrilas/ultraestrutura , Esforço Físico , Sarcolema/ultraestrutura , Animais , Doença da Descompressão/metabolismo , Doença da Descompressão/patologia , Feminino , Músculo Esquelético/lesões , NF-kappa B/genética , NF-kappa B/metabolismo , Artéria Pulmonar/diagnóstico por imagem , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Transcrição Gênica , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/metabolismo , Ultrassonografia , Cadeia B de alfa-Cristalina/genética , Cadeia B de alfa-Cristalina/metabolismo
19.
FASEB J ; 26(3): 1311-22, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22131268

RESUMO

Mutation T4825I in the type 1 ryanodine receptor (RYR1(T4825I/+)) confers human malignant hyperthermia susceptibility (MHS). We report a knock-in mouse line that expresses the isogenetic mutation T4826I. Heterozygous RYR1(T4826I/+) (Het) or homozygous RYR1(T4826I/T4826I) (Hom) mice are fully viable under typical rearing conditions but exhibit genotype- and sex-dependent susceptibility to environmental conditions that trigger MH. Hom mice maintain higher core temperatures than WT in the home cage, have chronically elevated myoplasmic[Ca(2+)](rest), and present muscle damage in soleus with a strong sex bias. Mice subjected to heat stress in an enclosed 37°C chamber fail to trigger MH regardless of genotype, whereas heat stress at 41°C invariably triggers fulminant MH in Hom, but not Het, mice within 20 min. WT and Het female mice fail to maintain euthermic body temperature when placed atop a bed whose surface is 37°C during halothane anesthesia (1.75%) and have no hyperthermic response, whereas 100% Hom mice of either sex and 17% of the Het males develop fulminant MH. WT mice placed on a 41°C bed maintain body temperature while being administered halothane, and 40% of the Het females and 100% of the Het males develop fulminant MH within 40 min. Myopathic alterations in soleus were apparent by 12 mo, including abnormally distributed and enlarged mitochondria, deeply infolded sarcolemma, and frequent Z-line streaming regions, which were more severe in males. These data demonstrate that an MHS mutation within the S4-S5 cytoplasmic linker of RYR1 confers genotype- and sex-dependent susceptibility to pharmacological and environmental stressors that trigger fulminant MH and promote myopathy.


Assuntos
Predisposição Genética para Doença/genética , Hipertermia Maligna/genética , Músculo Esquelético/metabolismo , Mutação , Canal de Liberação de Cálcio do Receptor de Rianodina/genética , Substituição de Aminoácidos , Anestésicos Inalatórios/administração & dosagem , Animais , Temperatura Corporal/efeitos dos fármacos , Temperatura Corporal/genética , Temperatura Corporal/fisiologia , Feminino , Expressão Gênica , Genótipo , Halotano/administração & dosagem , Temperatura Alta , Humanos , Masculino , Potenciais da Membrana , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microscopia Eletrônica , Fibras Musculares Esqueléticas/metabolismo , Fibras Musculares Esqueléticas/fisiologia , Músculo Esquelético/patologia , Canal de Liberação de Cálcio do Receptor de Rianodina/fisiologia , Sarcolema/metabolismo , Sarcolema/ultraestrutura , Fatores Sexuais
20.
J Muscle Res Cell Motil ; 32(4-5): 271-80, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22057634

RESUMO

Ahnak1 is a giant, ubiquitously expressed, plasma membrane support protein whose function in skeletal muscle is largely unknown. Therefore, we investigated whether ahnak would be influenced by alterations of the sarcolemma exemplified by dysferlin mutations known to render the sarcolemma vulnerable or by mutations in calpain3, a protease known to cleave ahnak. Human muscle biopsy specimens obtained from patients with limb girdle muscular dystrophy (LGMD) caused by mutations in dysferlin (LGMD2B) and calpain3 (LGMD2A) were investigated for ahnak expression and localization. We found that ahnak1 has lost its sarcolemmal localization in LGMD2B but not in LGMD2A. Instead ahnak1 appeared in muscle connective tissue surrounding the extracellular site of the muscle fiber in both muscular dystrophies. The entire giant ahnak1 molecule was present outside the muscle fiber and did only partially colocalize with CD45-positive immune cell infiltration and the extracelluar matrix proteins fibronectin and collagenVI. Further, vesicles shedded in response to Ca(2+) by primary human myotubes were purified and their protein content was analysed. Ahnak1 was prominently present in these vesicles. Electron microscopy revealed a homogenous population of vesicles with a diameter of about 150 nm. This is the first study demonstrating vesicle release from human myotubes that may be one mechanism underlying abnormally localized ahnak1. Taken together, our results define ahnak1 in muscle connective tissue as a novel feature of two genetically distinct muscular dystrophies that might contribute to disease pathology.


Assuntos
Tecido Conjuntivo/ultraestrutura , Proteínas de Membrana/metabolismo , Fibras Musculares Esqueléticas/ultraestrutura , Distrofia Muscular do Cíngulo dos Membros/metabolismo , Proteínas de Neoplasias/metabolismo , Sarcolema/ultraestrutura , Vesículas Transportadoras/ultraestrutura , Calpaína/genética , Calpaína/metabolismo , Estudos de Casos e Controles , Disferlina , Homozigoto , Humanos , Imuno-Histoquímica , Proteínas de Membrana/genética , Microscopia Eletrônica , Fibras Musculares Esqueléticas/metabolismo , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Distrofia Muscular do Cíngulo dos Membros/genética , Mutação , Vesículas Transportadoras/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA