Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 79
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Biochem J ; 2024 Jan 02.
Artigo em Inglês | MEDLINE | ID: mdl-38164968

RESUMO

Mitochondrial ATP synthases form rows of dimers, which induce membrane curvature to give cristae their characteristic lamellar or tubular morphology. The angle formed between the central stalks of ATP synthase dimers varies between species. Using cryo-electron tomography and sub-tomogram averaging, we determined the structure of the ATP synthase dimer from the nematode worm C. elegans and show that the dimer angle differs from previously determined structures. The consequences of this species-specific difference at the dimer interface were investigated by comparing C. elegans and S. cerevisiae mitochondrial morphology. We reveal that C. elegans has a larger ATP synthase dimer angle with more lamellar (flatter) cristae when compared to yeast. The underlying cause of this difference was investigated by generating an atomic model of the C. elegans ATP synthase dimer by homology modelling. A comparison of our C. elegans model to an existing S. cerevisiae structure reveals the presence of extensions and rearrangements in C. elegans subunits associated with maintaining the dimer interface. We speculate that increasing dimer angles could provide an advantage for species that inhabit variable-oxygen environments by forming flatter more energetically efficient cristae.

2.
Biochem Biophys Res Commun ; 703: 149656, 2024 Apr 09.
Artigo em Inglês | MEDLINE | ID: mdl-38364681

RESUMO

Dystroglycan (DG) is a cell adhesion complex that is widely expressed in tissues. It is composed by two subunits, α-DG, a highly glycosylated protein that interacts with several extracellular matrix proteins, and transmembrane ß-DG whose, cytodomain binds to the actin cytoskeleton. Glycosylation of α-DG is crucial for functioning as a receptor for its multiple extracellular binding partners. Perturbation of α-DG glycosylation is the central event in the pathogenesis of severe pathologies such as muscular dystrophy and cancer. ß-DG acts as a scaffold for several cytoskeletal and nuclear proteins and very little is known about the fine regulation of some of these intracellular interactions and how they are perturbed in diseases. To start filling this gap by identifying uncharacterized intracellular networks preferentially associated with ß-DG, HEK-293 cells were transiently transfected with a plasmid carrying the ß-DG subunit with GFP fused at its C-terminus. With this strategy, we aimed at forcing ß-DG to occupy multiple intracellular locations instead of sitting tightly at its canonical plasma membrane milieu, where it is commonly found in association with α-DG. Immunoprecipitation by anti-GFP antibodies followed by shotgun proteomic analysis led to the identification of an interactome formed by 313 exclusive protein matches for ß-DG binding. A series of already known ß-DG interactors have been found, including ezrin and emerin, whilst significant new matches, which include potential novel ß-DG interactors and their related networks, were identified in diverse subcellular compartments, such as cytoskeleton, endoplasmic reticulum/Golgi, mitochondria, nuclear membrane and the nucleus itself. Of particular interest amongst the novel identified matches, Lamina-Associated Polypeptide-1B (LAP1B), an inner nuclear membrane protein, whose mutations are known to cause nuclear envelopathies characterized by muscular dystrophy, was found to interact with ß-DG in HEK-293 cells. This evidence was confirmed by immunoprecipitation, Western blotting and immunofluorescence experiments. We also found by immunofluorescence experiments that LAP1B looses its nuclear envelope localization in C2C12 DG-knock-out cells, suggesting that LAP1B requires ß-DG for a proper nuclear localization. These results expand the role of ß-DG as a nuclear scaffolding protein and provide novel evidence of a possible link between dystroglycanopathies and nuclear envelopathies displaying with muscular dystrophy.


Assuntos
Distroglicanas , Distrofias Musculares , Humanos , Distroglicanas/química , Células HEK293 , Proteômica , Distrofias Musculares/metabolismo , Membrana Nuclear/metabolismo
3.
Int J Mol Sci ; 24(20)2023 Oct 17.
Artigo em Inglês | MEDLINE | ID: mdl-37894956

RESUMO

Muscle weakness and muscle loss characterize many physio-pathological conditions, including sarcopenia and many forms of muscular dystrophy, which are often also associated with mitochondrial dysfunction. Verbascoside, a phenylethanoid glycoside of plant origin, also named acteoside, has shown strong antioxidant and anti-fatigue activity in different animal models, but the molecular mechanisms underlying these effects are not completely understood. This study aimed to investigate the influence of verbascoside on mitochondrial function and its protective role against H2O2-induced oxidative damage in murine C2C12 myoblasts and myotubes pre-treated with verbascoside for 24 h and exposed to H2O2. We examined the effects of verbascoside on cell viability, intracellular reactive oxygen species (ROS) production and mitochondrial function through high-resolution respirometry. Moreover, we verified whether verbascoside was able to stimulate nuclear factor erythroid 2-related factor (Nrf2) activity through Western blotting and confocal fluorescence microscopy, and to modulate the transcription of its target genes, such as heme oxygenase-1 (HO-1) and peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC-1α), by Real Time PCR. We found that verbascoside (1) improved mitochondrial function by increasing mitochondrial spare respiratory capacity; (2) mitigated the decrease in cell viability induced by H2O2 and reduced ROS levels; (3) promoted the phosphorylation of Nrf2 and its nuclear translocation; (4) increased the transcription levels of HO-1 and, in myoblasts but not in myotubes, those of PGC-1α. These findings contribute to explaining verbascoside's ability to relieve muscular fatigue and could have positive repercussions for the development of therapies aimed at counteracting muscle weakness and mitochondrial dysfunction.


Assuntos
Antioxidantes , Fator 2 Relacionado a NF-E2 , Animais , Camundongos , Antioxidantes/metabolismo , Linhagem Celular , Heme Oxigenase-1/genética , Heme Oxigenase-1/metabolismo , Peróxido de Hidrogênio/metabolismo , Mitocôndrias/metabolismo , Fibras Musculares Esqueléticas/metabolismo , Debilidade Muscular/metabolismo , Fator 2 Relacionado a NF-E2/metabolismo , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/genética , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais
4.
Behav Res Methods ; 55(7): 3929-3951, 2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-36526887

RESUMO

Procedural knowledge space theory (PKST) was recently proposed by Stefanutti (British Journal of Mathematical and Statistical Psychology, 72(2) 185-218, 2019) for the assessment of human problem-solving skills. In PKST, the problem space formally represents how a family of problems can be solved and the knowledge space represents the skills required for solving those problems. The Markov solution process model (MSPM) by Stefanutti et al. (Journal of Mathematical Psychology, 103, 102552, 2021) provides a probabilistic framework for modeling the solution process of a task, via PKST. In this article, three adaptive procedures for the assessment of problem-solving skills are proposed that are based on the MSPM. Beside execution correctness, they also consider the sequence of moves observed in the solution of a problem with the aim of increasing efficiency and accuracy of assessments. The three procedures differ from one another in the assumption underlying the solution process, named pre-planning, interim-planning, and mixed-planning. In two simulation studies, the three adaptive procedures have been compared to one another and to the continuous Markov procedure (CMP) by Doignon and Falmagne (1988a). The last one accounts for dichotomous correct/wrong answers only. Results show that all the MSP-based adaptive procedures outperform the CMP in both accuracy and efficiency. These results have been obtained in the framework of the Tower of London test but the procedures can also be applied to all psychological and neuropsychological tests that have a problem space. Thus, the adaptive procedures presented in this paper pave the way to the adaptive assessment in the area of neuropsychological tests.


Assuntos
Algoritmos , Resolução de Problemas , Humanos , Matemática , Simulação por Computador , Cadeias de Markov , Testes Neuropsicológicos
5.
J Chem Inf Model ; 60(6): 3145-3156, 2020 06 22.
Artigo em Inglês | MEDLINE | ID: mdl-32356985

RESUMO

The acetylglucosaminyltransferase-like protein LARGE1 is an enzyme that is responsible for the final steps of the post-translational modifications of dystroglycan (DG), a membrane receptor that links the cytoskeleton with the extracellular matrix in the skeletal muscle and in a variety of other tissues. LARGE1 acts by adding the repeating disaccharide unit [-3Xyl-α1,3GlcAß1-] to the extracellular portion of the DG complex (α-DG); defects in the LARGE1 gene result in an aberrant glycosylation of α-DG and consequent impairment of its binding to laminin, eventually affecting the connection between the cell and the extracellular environment. In the skeletal muscle, this leads to degeneration of the muscular tissue and muscular dystrophy. So far, a few missense mutations have been identified within the LARGE1 protein and linked to congenital muscular dystrophy, and because no structural information is available on this enzyme, our understanding of the molecular mechanisms underlying these pathologies is still very limited. Here, we generated a 3D model structure of the two catalytic domains of LARGE1, combining different molecular modeling approaches. Furthermore, by using molecular dynamics simulations, we analyzed the effect on the structure and stability of the first catalytic domain of the pathological missense mutation S331F that gives rise to a severe form of muscle-eye-brain disease.


Assuntos
Distroglicanas , Laminina , N-Acetilglucosaminiltransferases/química , Animais , Distroglicanas/química , Glicosilação , Laminina/química , Camundongos , Simulação de Dinâmica Molecular , Processamento de Proteína Pós-Traducional
6.
Int J Mol Sci ; 21(14)2020 Jul 14.
Artigo em Inglês | MEDLINE | ID: mdl-32674290

RESUMO

Nuclear ß-dystroglycan (ß-DG) is involved in the maintenance of nuclear architecture and function. Nonetheless, its relevance in defined nuclear processes remains to be determined. In this study we generated a C2C12 cell-based DG-null model using CRISPR-Cas9 technology to provide insights into the role of ß-DG on nuclear processes. Since DG-null cells exhibited decreased levels of lamin B1, we aimed to elucidate the contribution of DG to senescence, owing to the central role of lamin B1 in this pathway. Remarkably, the lack of DG enables C2C12 cells to acquire senescent features, including cell-cycle arrest, increased senescence-associated-ß-galactosidase activity, heterochromatin loss, aberrant nuclear morphology and nucleolar disruption. We demonstrated that genomic instability is one driving cause of the senescent phenotype in DG-null cells via the activation of a DNA-damage response associated with mitotic failure, as shown by the presence of multipolar mitotic spindles, which in turn induced the formation of micronuclei and γH2AX foci (DNA-damage marker), telomere shortening and p53/p21 upregulation. Altogether, these events might ultimately lead to premature senescence, impeding the replication of the damaged genome. In summary, we present evidence supporting a role for DG in protecting against senescence, through the maintenance of proper lamin B1 expression/localization and proper mitotic spindle organization.


Assuntos
Senescência Celular/genética , Distroglicanas/genética , Instabilidade Genômica/genética , Mitose/genética , Sequência de Aminoácidos , Animais , Sequência de Bases , Pontos de Checagem do Ciclo Celular/genética , Linhagem Celular , Núcleo Celular/genética , Inibidor de Quinase Dependente de Ciclina p21/genética , Dano ao DNA/genética , Histonas/genética , Lamina Tipo B/genética , Camundongos , Camundongos Knockout , Fuso Acromático/genética , Telômero/genética , Proteína Supressora de Tumor p53/genética , Regulação para Cima/genética , beta-Galactosidase/genética
7.
Int J Mol Sci ; 21(17)2020 Aug 19.
Artigo em Inglês | MEDLINE | ID: mdl-32824881

RESUMO

ß-dystroglycan (ß-DG) assembles with lamins A/C and B1 and emerin at the nuclear envelope (NE) to maintain proper nuclear architecture and function. To provide insight into the nuclear function of ß-DG, we characterized the interaction between ß-DG and emerin at the molecular level. Emerin is a major NE protein that regulates multiple nuclear processes and whose deficiency results in Emery-Dreifuss muscular dystrophy (EDMD). Using truncated variants of ß-DG and emerin, via a series of in vitro and in vivo binding experiments and a tailored computational analysis, we determined that the ß-DG-emerin interaction is mediated at least in part by their respective transmembrane domains (TM). Using surface plasmon resonance assays we showed that emerin binds to ß-DG with high affinity (KD in the nanomolar range). Remarkably, the analysis of cells in which DG was knocked out demonstrated that loss of ß-DG resulted in a decreased emerin stability and impairment of emerin-mediated processes. ß-DG and emerin are reciprocally required for their optimal targeting within the NE, as shown by immunofluorescence, western blotting and immunoprecipitation assays using emerin variants with mutations in the TM domain and B-lymphocytes of a patient with EDMD. In summary, we demonstrated that ß-DG plays a role as an emerin interacting partner modulating its stability and function.


Assuntos
Distroglicanas/metabolismo , Proteínas de Membrana/metabolismo , Distrofia Muscular de Emery-Dreifuss/metabolismo , Proteínas Nucleares/metabolismo , Transporte Ativo do Núcleo Celular , Animais , Linfócitos B/metabolismo , Sítios de Ligação , Linhagem Celular , Células Cultivadas , Distroglicanas/química , Distroglicanas/genética , Células HeLa , Humanos , Proteínas de Membrana/química , Proteínas de Membrana/genética , Camundongos , Distrofia Muscular de Emery-Dreifuss/genética , Mutação , Membrana Nuclear/metabolismo , Proteínas Nucleares/química , Proteínas Nucleares/genética , Ligação Proteica
8.
J Cell Mol Med ; 23(5): 3058-3062, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30838779

RESUMO

Dystroglycan is a major non-integrin adhesion complex that connects the cytoskeleton to the surrounding basement membranes, thus providing stability to skeletal muscle. In Vertebrates, hypoglycosylation of α-dystroglycan has been strongly linked to muscular dystrophy phenotypes, some of which also show variable degrees of cognitive impairments, collectively termed dystroglycanopathies. Only a small number of mutations in the dystroglycan gene, leading to the so called primary dystroglycanopathies, has been described so far, as opposed to the ever-growing number of identified secondary or tertiary dystroglycanopathies (caused by genetic abnormalities in glycosyltransferases or in enzymes involved in the synthesis of the carbohydrate building blocks). The few mutations found within the autonomous N-terminal domain of α-dystroglycan seem to destabilise it to different degrees, without influencing the overall folding and targeting of the dystroglycan complex. On the contrary other mutations, some located at the α/ß interface of the dystroglycan complex, seem to be able to interfere with its maturation, thus compromising its stability and eventually leading to the intracellular engulfment and/or partial or even total degradation of the dystroglycan uncleaved precursor.


Assuntos
Disfunção Cognitiva/genética , Distroglicanas/genética , Distrofias Musculares/genética , Membrana Basal/metabolismo , Disfunção Cognitiva/patologia , Citoesqueleto/genética , Glicosilação , Humanos , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Distrofias Musculares/patologia , Mutação/genética
9.
FASEB J ; 32(4): 2223-2234, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29233859

RESUMO

Class II chaperonins are essential multisubunit complexes that aid the folding of nonnative proteins in the cytosol of archaea and eukarya. They use energy derived from ATP to drive a series of structural rearrangements that enable polypeptides to fold within their central cavity. These events are regulated by an elaborate allosteric mechanism in need of elucidation. We employed mutagenesis and experimental analysis in concert with in silico molecular dynamics simulations and interface-binding energy calculations to investigate the class II chaperonin from Thermoplasma acidophilum. Here we describe the effects on the asymmetric allosteric mechanism and on hetero-oligomeric complex formation in a panel of mutants in the ATP-binding pocket of the α and ß subunits. Our observations reveal a potential model for a nonconcerted folding mechanism optimized for protecting and refolding a range of nonnative substrates under different environmental conditions, starting to unravel the role of subunit heterogeneity in this folding machine and establishing important links with the behavior of the most complex eukaryotic chaperonins.-Shoemark, D. K., Sessions, R. B., Brancaccio, A., Bigotti, M. G. Intraring allostery controls the function and assembly of a hetero-oligomeric class II chaperonin.


Assuntos
Sítio Alostérico , Proteínas Arqueais/química , Chaperoninas do Grupo II/química , Simulação de Dinâmica Molecular , Trifosfato de Adenosina/química , Trifosfato de Adenosina/metabolismo , Regulação Alostérica , Proteínas Arqueais/metabolismo , Chaperoninas do Grupo II/metabolismo , Ligação Proteica , Multimerização Proteica , Thermoplasma/química
10.
Hum Mutat ; 39(2): 266-280, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29134705

RESUMO

Dystroglycan (DG) is a cell adhesion complex composed by two subunits, the highly glycosylated α-DG and the transmembrane ß-DG. In skeletal muscle, DG is involved in dystroglycanopathies, a group of heterogeneous muscular dystrophies characterized by a reduced glycosylation of α-DG. The genes mutated in secondary dystroglycanopathies are involved in the synthesis of O-mannosyl glycans and in the O-mannosylation pathway of α-DG. Mutations in the DG gene (DAG1), causing primary dystroglycanopathies, destabilize the α-DG core protein influencing its binding to modifying enzymes. Recently, a homozygous mutation (p.Cys699Phe) hitting the ß-DG ectodomain has been identified in a patient affected by muscle-eye-brain disease with multicystic leucodystrophy, suggesting that other mechanisms than hypoglycosylation of α-DG could be implicated in dystroglycanopathies. Herein, we have characterized the DG murine mutant counterpart by transfection in cellular systems and high-resolution microscopy. We observed that the mutation alters the DG processing leading to retention of its uncleaved precursor in the endoplasmic reticulum. Accordingly, small-angle X-ray scattering data, corroborated by biochemical and biophysical experiments, revealed that the mutation provokes an alteration in the ß-DG ectodomain overall folding, resulting in disulfide-associated oligomerization. Our data provide the first evidence of a novel intracellular mechanism, featuring an anomalous endoplasmic reticulum-retention, underlying dystroglycanopathy.


Assuntos
Distroglicanas/genética , Leucoencefalopatias/genética , Proteínas Mutantes/genética , Síndrome de Walker-Warburg/genética , Linhagem Celular , Humanos
11.
Electrophoresis ; 37(2): 321-34, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26542084

RESUMO

One of the problems with 2DE is that proteins present in low amounts in a sample are usually not detected, since their signals are masked by the predominant proteins. The elimination of these abundant proteins is not a guaranteed solution to achieve the desired results. The main objective of this study was the comparison of common and simple methodologies employed for 2DE analysis followed by MS identification, focusing on a pre-purified sample using a wheat germ agglutinin (WGA) column. Adult male C57Black/Crj6 (C57BL/6) mice were chosen as the model animal in this study; the gastrocnemius muscles were collected and processed for the experiments. The initial fractionation with succinylated WGA was successful for the elimination of the most abundant proteins. Two quantification methods were employed for the purified samples, and bicinchoninic acid (BCA) was proven to be most reliable for the quantification of glycoproteins. The gel staining method, however, was found to be decisive for the detection of specific proteins, since their structures affect the interaction of the dye with the peptide backbone. The Coomassie Blue R-250 dye very weakly stained the gel with the WGA purified sample. When the same gel was stained with silver nitrate, however, MS could positively assign 12 new spots. The structure of the referred proteins was not found to be prone to interaction with Coomassie blue.


Assuntos
Eletroforese em Gel Bidimensional/métodos , Glicoproteínas/análise , Músculos/química , Animais , Cromatografia de Afinidade/métodos , Corantes/análise , Glicoproteínas/isolamento & purificação , Glicosilação , Masculino , Camundongos Endogâmicos C57BL , Proteínas Musculares/análise , Proteínas Musculares/isolamento & purificação , Corantes de Rosanilina/análise , Coloração pela Prata/métodos , Espectrometria de Massas em Tandem/métodos , Aglutininas do Germe de Trigo/química
12.
Dis Model Mech ; 17(6)2024 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-38616731

RESUMO

Dystroglycan (DG) is an extracellular matrix receptor consisting of an α- and a ß-DG subunit encoded by the DAG1 gene. The homozygous mutation (c.2006G>T, p.Cys669Phe) in ß-DG causes muscle-eye-brain disease with multicystic leukodystrophy in humans. In a mouse model of this primary dystroglycanopathy, approximately two-thirds of homozygous embryos fail to develop to term. Mutant mice that are born undergo a normal postnatal development but show a late-onset myopathy with partially penetrant histopathological changes and an impaired performance on an activity wheel. Their brains and eyes are structurally normal, but the localization of mutant ß-DG is altered in the glial perivascular end-feet, resulting in a perturbed protein composition of the blood-brain and blood-retina barrier. In addition, α- and ß-DG protein levels are significantly reduced in muscle and brain of mutant mice. Owing to the partially penetrant developmental phenotype of the C669F ß-DG mice, they represent a novel and highly valuable mouse model with which to study the molecular effects of ß-DG functional alterations both during embryogenesis and in mature muscle, brain and eye, and to gain insight into the pathogenesis of primary dystroglycanopathies.


Assuntos
Barreira Hematoencefálica , Distroglicanas , Mutação de Sentido Incorreto , Animais , Distroglicanas/metabolismo , Barreira Hematoencefálica/patologia , Barreira Hematoencefálica/metabolismo , Mutação de Sentido Incorreto/genética , Camundongos , Doenças Musculares/genética , Doenças Musculares/patologia , Perda do Embrião/patologia , Perda do Embrião/genética , Fenótipo , Embrião de Mamíferos/metabolismo , Embrião de Mamíferos/patologia , Camundongos Endogâmicos C57BL , Encéfalo/patologia , Encéfalo/metabolismo , Encéfalo/embriologia
13.
Antiviral Res ; 224: 105837, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38387750

RESUMO

The COVID-19 pandemic has shown the need to develop effective therapeutics in preparedness for further epidemics of virus infections that pose a significant threat to human health. As a natural compound antiviral candidate, we focused on α-dystroglycan, a highly glycosylated basement membrane protein that links the extracellular matrix to the intracellular cytoskeleton. Here we show that the N-terminal fragment of α-dystroglycan (α-DGN), as produced in E. coli in the absence of post-translational modifications, blocks infection of SARS-CoV-2 in cell culture, human primary gut organoids and the lungs of transgenic mice expressing the human receptor angiotensin I-converting enzyme 2 (hACE2). Prophylactic and therapeutic administration of α-DGN reduced SARS-CoV-2 lung titres and protected the mice from respiratory symptoms and death. Recombinant α-DGN also blocked infection of a wide range of enveloped viruses including the four Dengue virus serotypes, influenza A virus, respiratory syncytial virus, tick-borne encephalitis virus, but not human adenovirus, a non-enveloped virus in vitro. This study establishes soluble recombinant α-DGN as a broad-band, natural compound candidate therapeutic against enveloped viruses.


Assuntos
COVID-19 , SARS-CoV-2 , Camundongos , Animais , Humanos , Distroglicanas , Pandemias , Escherichia coli , Camundongos Transgênicos , Antivirais/farmacologia
14.
Brain Sci ; 14(2)2024 Jan 24.
Artigo em Inglês | MEDLINE | ID: mdl-38391697

RESUMO

Assessing executive functions in individuals with disorders or clinical conditions can be challenging, as they may lack the abilities needed for conventional test formats. The use of more personalized test versions, such as adaptive assessments, might be helpful in evaluating individuals with specific needs. This paper introduces PsycAssist, a web-based artificial intelligence system designed for neuropsychological adaptive assessment and training. PsycAssist is a highly flexible and scalable system based on procedural knowledge space theory and may be used potentially with many types of tests. We present the architecture and adaptive assessment engine of PsycAssist and the two currently available tests: Adap-ToL, an adaptive version of the Tower of London-like test to assess planning skills, and MatriKS, a Raven-like test to evaluate fluid intelligence. Finally, we describe the results of an investigation of the usability of Adap-ToL and MatriKS: the evaluators perceived these tools as appropriate and well-suited for their intended purposes, and the test-takers perceived the assessment as a positive experience. To sum up, PsycAssist represents an innovative and promising tool to tailor evaluation and training to the specific characteristics of the individual, useful for clinical practice.

15.
BMC Biochem ; 14: 15, 2013 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-23815856

RESUMO

BACKGROUND: α-Dystroglycan (α-DG) is heavily glycosylated within its central mucin-like domain. The glycosylation shell of α-dystroglycan is known to largely influence its functional properties toward extracellular ligands. The structural features of this α-dystroglycan domain have been poorly studied so far. For the first time, we have attempted a recombinant expression approach in E. coli cells, in order to analyze by biochemical and biophysical techniques this important domain of the α-dystroglycan core protein. RESULTS: We expressed the recombinant mucin-like domain of human α-dystroglycan in E. coli cells, and purified it as a soluble peptide of 174 aa. A cleavage event, that progressively emerges under repeated cycles of freeze/thaw, occurs at the carboxy side of Arg461, liberating a 151 aa fragment as revealed by mass spectrometry analysis. The mucin-like peptide lacks any particular fold, as confirmed by its hydrodynamic properties and its fluorescence behavior under guanidine hydrochloride denaturation. Dynamic light scattering has been used to demonstrate that this mucin-like peptide is arranged in a conformation that is prone to aggregation at room temperature, with a melting temperature of ~40°C, which indicates a pronounced instability. Such a conclusion has been corroborated by trypsin limited proteolysis, upon which the protein has been fully degraded in less than 60 min. CONCLUSIONS: Our analysis indirectly confirms the idea that the mucin-like domain of α-dystroglycan needs to be extensively glycosylated in order to reach a stable conformation. The absence/reduction of glycosylation by itself may greatly reduce the stability of the dystroglycan complex. Although an altered pattern of α-dystroglycan O-mannosylation, that is not significantly changing its overall glycosylation fraction, represents the primary molecular clue behind currently known dystroglycanopathies, it cannot be ruled out that still unidentified forms of αDG-related dystrophy might originate by a more substantial reduction of α-dystroglycan glycosylation and by its consequent destabilization.


Assuntos
Distroglicanas/metabolismo , Distroglicanas/química , Distroglicanas/genética , Glicosilação , Humanos , Luz , Espectrometria de Massas , Desnaturação Proteica , Estabilidade Proteica , Estrutura Terciária de Proteína , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/química , Proteínas Recombinantes/isolamento & purificação , Espalhamento de Radiação
16.
Exp Cell Res ; 318(19): 2460-9, 2012 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-22814252

RESUMO

Dystroglycan (DG) is an extracellular receptor composed of two subunits, α-DG and ß-DG, connected through the α-DG C-terminal domain and the ß-DG N-terminal domain. We report an alanine scanning of all DG cysteine residues performed on DG-GFP constructs overexpressed in 293-Ebna cells, demonstrating that Cys-669 and Cys-713, both located within the ß-DG N-terminal domain, are key residues for the DG precursor cleavage and trafficking, but not for the interaction between the two DG subunits. In addition, we have used immunprecipitation and confocal microscopy showing that ERp57, a member of the disulfide isomerase family involved in glycoprotein folding, is associated and colocalizes immunohistochemically with ß-DG in the ER and at the plasma membrane of 293-Ebna cells. The ß-DG-ERp57 complex also included α-DG. DG mutants, unable to undergo the precursor cleavage, were still associated to ERp57. ß-DG and ERp57 were also co-immunoprecipitated in rat heart and kidney tissues. In vitro, a mutant ERp57, mimicking the reduced form of the wild-type protein, interacts directly with the recombinant N-terminal domain of both α-DG and ß-DG with apparent dissociation constant values in the micromolar range. ERp57 is likely to be involved in the DG processing/maturation pathway, but its association to the mature DG complex might also suggest some further functional role that needs to be investigated.


Assuntos
Distroglicanas/metabolismo , Isomerases de Dissulfetos de Proteínas/metabolismo , Animais , Membrana Celular/genética , Membrana Celular/metabolismo , Células Cultivadas , Distroglicanas/genética , Glicosilação , Células HEK293 , Coração/fisiologia , Humanos , Rim/metabolismo , Rim/fisiologia , Mutação , Ligação Proteica/genética , Ligação Proteica/fisiologia , Estrutura Terciária de Proteína , Subunidades Proteicas , Transporte Proteico/fisiologia , Ratos
17.
Am J Hum Genet ; 85(2): 155-67, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19631309

RESUMO

We report the case of a congenital myasthenic syndrome due to a mutation in AGRN, the gene encoding agrin, an extracellular matrix molecule released by the nerve and critical for formation of the neuromuscular junction. Gene analysis identified a homozygous missense mutation, c.5125G>C, leading to the p.Gly1709Arg variant. The muscle-biopsy specimen showed a major disorganization of the neuromuscular junction, including changes in the nerve-terminal cytoskeleton and fragmentation of the synaptic gutters. Experiments performed in nonmuscle cells or in cultured C2C12 myotubes and using recombinant mini-agrin for the mutated and the wild-type forms showed that the mutated form did not impair the activation of MuSK or change the total number of induced acetylcholine receptor aggregates. A solid-phase assay using the dystrophin glycoprotein complex showed that the mutation did not affect the binding of agrin to alpha-dystroglycan. Injection of wild-type or mutated agrin into rat soleus muscle induced the formation of nonsynaptic acetylcholine receptor clusters, but the mutant protein specifically destabilized the endogenous neuromuscular junctions. Importantly, the changes observed in rat muscle injected with mutant agrin recapitulated the pre- and post-synaptic modifications observed in the patient. These results indicate that the mutation does not interfere with the ability of agrin to induce postsynaptic structures but that it dramatically perturbs the maintenance of the neuromuscular junction.


Assuntos
Agrina/genética , Mutação de Sentido Incorreto , Síndromes Miastênicas Congênitas/genética , Sinapses/metabolismo , Adulto , Agrina/química , Agrina/metabolismo , Animais , Biópsia , Linhagem Celular , Análise Mutacional de DNA , Distroglicanas/metabolismo , Feminino , Humanos , Masculino , Modelos Químicos , Fibras Musculares Esqueléticas/citologia , Fibras Musculares Esqueléticas/metabolismo , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Músculo Esquelético/cirurgia , Músculo Esquelético/ultraestrutura , Junção Neuromuscular/genética , Junção Neuromuscular/metabolismo , Junção Neuromuscular/fisiologia , Junção Neuromuscular/ultraestrutura , Linhagem , Estrutura Terciária de Proteína , Ratos , Receptores Colinérgicos/genética , Receptores Colinérgicos/metabolismo , Receptores Colinérgicos/fisiologia , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo
18.
IUBMB Life ; 64(12): 988-94, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23129308

RESUMO

Dystroglycan (DG) is a membrane-associated protein complex formed by two noncovalently linked subunits, α-DG, a highly glycosylated extracellular protein, and ß-DG, a transmembrane protein. The interface between the two DG subunits, which is crucial to maintain the integrity of the plasma membrane, involves the C-terminal domain of α-DG and the N-terminal extracellular domain of ß-DG. It is well known that under both, physiological and pathological conditions, gelatinases (i.e. MMP-9 and/or MMP-2) can degrade DG, disrupting the connection between the extracellular matrix and the cytoskeleton. However, the molecular mechanisms and the exact cleavage sites underlying these events are still largely unknown. In a previous study, we have characterized the enzymatic digestion of the murine ß-DG ectodomain by gelatinases, identifying a main cleavage site on the ß-DG ectodomain produced by MMP-9. In this article, we have deepened the pattern of the ß-DG ectodomain digestion by MMP-2 by using a combined approach based on SDS-PAGE, Orbitrap, and HPLC-ESI-IT mass spectrometry. Furthermore, we have characterized the kineticparameters of the digestion of some ß-DG ectodomain mutants by gelatinases.


Assuntos
Distroglicanas/metabolismo , Metaloproteinase 2 da Matriz/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Mutação , Fragmentos de Peptídeos/química , Sequência de Aminoácidos , Animais , Baculoviridae/genética , Distroglicanas/química , Distroglicanas/genética , Escherichia coli/genética , Cinética , Metaloproteinase 2 da Matriz/genética , Metaloproteinase 9 da Matriz/genética , Camundongos , Dados de Sequência Molecular , Fragmentos de Peptídeos/metabolismo , Domínios e Motivos de Interação entre Proteínas , Proteólise , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Células Sf9
19.
BMC Biochem ; 13: 14, 2012 Jul 26.
Artigo em Inglês | MEDLINE | ID: mdl-22835149

RESUMO

BACKGROUND: Epitope tags and fluorescent fusion proteins have become indispensable molecular tools for studies in the fields of biochemistry and cell biology. The knowledge collected on the subdomain organization of the two subunits of the adhesion complex dystroglycan (DG) enabled us to insert the 10 amino acids myc-tag at different locations along the α-subunit, in order to better visualize and investigate the DG complex in eukaryotic cells. RESULTS: We have generated two forms of DG polypeptides via the insertion of the myc-tag 1) within a flexible loop (between a.a. 170 and 171) that separates two autonomous subdomains, and 2) within the C-terminal domain in position 500. Their analysis showed that double-tagging (the ß-subunit is linked to GFP) does not significantly interfere with the correct processing of the DG precursor (pre-DG) and confirmed that the α-DG N-terminal domain is processed in the cell before α-DG reaches its plasma membrane localization. In addition, myc insertion in position 500, right before the second Ig-like domain of α-DG, proved to be an efficient tool for the detection and pulling-down of glycosylated α-DG molecules targeted at the membrane. CONCLUSIONS: Further characterization of these and other myc-permissive site(s) will represent a valid support for the study of the maturation process of pre-DG and could result in the creation of a new class of intrinsic doubly-fluorescent DG molecules that would allow the monitoring of the two DG subunits, or of pre-DG, in cells without the need of antibodies.


Assuntos
Distroglicanas/química , Proteínas Proto-Oncogênicas c-myc/química , Linhagem Celular , Distroglicanas/metabolismo , Transferência Ressonante de Energia de Fluorescência , Humanos , Microscopia Confocal , Modelos Moleculares
20.
Front Cardiovasc Med ; 9: 813904, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35355976

RESUMO

Background: Mature cardiomyocytes are unable to proliferate, preventing the injured adult heart from repairing itself. Studies in rodents have suggested that the extracellular matrix protein agrin promotes cardiomyocyte proliferation in the developing heart and that agrin expression is downregulated shortly after birth, resulting in the cessation of proliferation. Agrin based therapies have proven successful at inducing repair in animal models of cardiac injury, however whether similar pathways exist in the human heart is unknown. Methods: Right ventricular (RV) biopsies were collected from 40 patients undergoing surgery for congenital heart disease and the expression of agrin and associated proteins was investigated. Results: Agrin transcripts were found in all samples and their levels were significantly negatively correlated to age (p = 0.026), as were laminin transcripts (p = 0.023), whereas no such correlation was found for the other proteins analyzed. No significant correlations for any of the proteins were found when grouping patients by their gender or pathology. Immunohistochemistry and western blots to detect and localize agrin and the other proteins under analysis in RV tissue, confirmed their presence in patients of all ages. Conclusions: We show that agrin is progressively downregulated with age in human RV tissue but not as dramatically as has been demonstrated in mice; highlighting both similarities and differences to findings in rodents. Our results lay the groundwork for future studies exploring the potential of agrin-based therapies in the repair of damaged human hearts.

SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa