Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 84
Filter
Add more filters










Publication year range
1.
FEBS J ; 289(10): 2755-2770, 2022 05.
Article in English | MEDLINE | ID: mdl-33825342

ABSTRACT

Desmin is the primary intermediate filament (IF) of cardiac, skeletal, and smooth muscle. By linking the contractile myofibrils to the sarcolemma and cellular organelles, desmin IF contributes to muscle structural and cellular integrity, force transmission, and mitochondrial homeostasis. Mutations in desmin cause myofibril misalignment, mitochondrial dysfunction, and impaired mechanical integrity leading to cardiac and skeletal myopathies in humans, often characterized by the accumulation of protein aggregates. Recent evidence indicates that desmin filaments also regulate proteostasis and cell size. In skeletal muscle, changes in desmin filament dynamics can facilitate catabolic events as an adaptive response to a changing environment. In addition, post-translational modifications of desmin and its misfolding in the heart have emerged as key determinants of homeostasis and disease. In this review, we provide an overview of the structural and cellular roles of desmin and propose new models for its novel functions in preserving the homeostasis of striated muscles.


Subject(s)
Desmin , Muscle, Skeletal , Myofibrils , Desmin/genetics , Desmin/physiology , Homeostasis , Humans , Muscle, Skeletal/physiology , Muscular Diseases/genetics , Muscular Diseases/metabolism , Myofibrils/physiology
2.
Am J Physiol Cell Physiol ; 316(5): C657-C667, 2019 05 01.
Article in English | MEDLINE | ID: mdl-30811221

ABSTRACT

Mitochondria perform a central role in life and death of the eukaryotic cell. They are major players in the generation of macroergic compounds and function as integrated signaling pathways, including the regulation of Ca2+ signals and apoptosis. A growing amount of evidence is demonstrating that mitochondria of muscle cells use cytoskeletal proteins (both microtubules and intermediate filaments) not only for their movement and proper cellular positioning, but also to maintain their biogenesis, morphology, function, and regulation of energy fluxes through the outer mitochondrial membrane (MOM). Here we consider the known literature data concerning the role of tubulin, plectin, desmin and vimentin in bioenergetic function of mitochondria in striated muscle cells, as well as in controlling the permeability of MOM for adenine nucleotides (ADNs). This is of great interest since dysfunctionality of these cytoskeletal proteins has been shown to result in severe myopathy associated with pronounced mitochondrial dysfunction. Further efforts are needed to uncover the pathways by which the cytoskeleton supports the functional capacity of mitochondria and transport of ADN(s) across the MOM (through voltage-dependent anion channel).


Subject(s)
Desmin/physiology , Mitochondrial Membranes/physiology , Muscle Cells/physiology , Plectin/physiology , Tubulin/physiology , Vimentin/physiology , Animals , Humans , Mitochondria/physiology
3.
Invest Ophthalmol Vis Sci ; 59(12): 4847-4855, 2018 10 01.
Article in English | MEDLINE | ID: mdl-30347079

ABSTRACT

Purpose: To investigate the effect of absence of desmin on the extraocular muscles (EOMs) with focus on the structure and composition of the cytoskeleton. Methods: The distribution of synemin, syncoilin, plectin, nestin, and dystrophin was evaluated on cross and longitudinal sections of EOMs and limb muscles from 1-year-old desmin knockout mice (desmin-/-) by immunofluorescence. General morphology was evaluated with hematoxylin and eosin while mitochondrial content and distribution were evaluated by succinate dehydrogenase (SDH) and modified Gomori trichrome stainings. Results: The muscle fibers of the EOMs in desmin-/- mice were remarkably well preserved in contrast to those in the severely affected soleus and the slightly affected gastrocnemius muscles. There were no signs of muscular pathology in the EOMs and all cytoskeletal proteins studied showed a correct location at sarcolemma and Z-discs. However, an increase of SDH staining and mitochondrial aggregates under the sarcolemma was detected. Conclusions: The structure of the EOMs was well preserved in the absence of desmin. We suggest that desmin is not necessary for correct synemin, syncoilin, plectin, and dystrophin location on the cytoskeleton of EOMs. However, it is needed to maintain an appropriate mitochondrial distribution in both EOMs and limb muscles.


Subject(s)
Cytoskeletal Proteins/metabolism , Cytoskeleton/physiology , Desmin/physiology , Muscle Proteins/metabolism , Oculomotor Muscles/cytology , Animals , Fluorescent Antibody Technique, Indirect , Mice , Mice, Knockout , Muscle Fibers, Skeletal/cytology , Muscle Fibers, Skeletal/metabolism , Muscle, Skeletal/cytology , Muscle, Skeletal/metabolism , Oculomotor Muscles/metabolism
4.
Sci Rep ; 7(1): 1391, 2017 05 03.
Article in English | MEDLINE | ID: mdl-28469177

ABSTRACT

In striated muscle, desmin intermediate filaments interlink the contractile myofibrillar apparatus with mitochondria, nuclei, and the sarcolemma. The desmin network's pivotal role in myocytes is evident since mutations in the human desmin gene cause severe myopathies and cardiomyopathies. Here, we investigated skeletal muscle pathology in myofibers and myofibrils isolated from young hetero- and homozygous R349P desmin knock-in mice, which carry the orthologue of the most frequent human desmin missense mutation R350P. We demonstrate that mutant desmin alters myofibrillar cytoarchitecture, markedly disrupts the lateral sarcomere lattice and distorts myofibrillar angular axial orientation. Biomechanical assessment revealed a high predisposition to stretch-induced damage in fiber bundles of R349P mice. Notably, Ca2+-sensitivity and passive myofibrillar tension were decreased in heterozygous fiber bundles, but increased in homozygous fiber bundles compared to wildtype mice. In a parallel approach, we generated and subsequently subjected immortalized heterozygous R349P desmin knock-in myoblasts to magnetic tweezer experiments that revealed a significantly increased sarcolemmal lateral stiffness. Our data suggest that mutated desmin already markedly impedes myocyte structure and function at pre-symptomatic stages of myofibrillar myopathies.


Subject(s)
Desmin/physiology , Muscle, Skeletal/physiology , Myoblasts, Skeletal/physiology , Myofibrils/physiology , Animals , Biomechanical Phenomena , Calcium Signaling , Cells, Cultured , Desmin/genetics , Gene Knock-In Techniques , Mice, Transgenic , Muscle Contraction , Muscle, Skeletal/pathology , Mutation , Myofibrils/pathology
5.
Meat Sci ; 113: 51-8, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26613188

ABSTRACT

The longissimus (n=118) (LL), semimembranosus (n=104) (SM) and biceps femoris (n=134) (BF) muscles were collected from lamb and sheep carcases and aged for 5days (LL and SM) and 14days (BF) to study the impact of muscle characteristics on tenderness as assessed by shear force (SF) and sensory evaluation. The impact of gender, animal age, collagen content, sarcomere length (SL), desmin degradation, ultimate pH and intramuscular fat (IMF) on tenderness was examined. The main factors which influenced SF of the LL were IMF, SL and desmin degradation, but for sensory tenderness, IMF, ultimate pH and gender were the main factors. The SF and sensory tenderness of the SM was best predicted by the degree of desmin degradation. For the BF soluble collagen and animal age both influenced SF. Different factors affect tenderness across muscles and not one prediction model applied across all muscles equally well.


Subject(s)
Collagen/physiology , Desmin/physiology , Meat/analysis , Muscle, Skeletal/physiology , Sarcomeres/physiology , Aging , Animals , Body Fat Distribution , Collagen/chemistry , Female , Food Analysis , Hydrogen-Ion Concentration , Male , Muscle, Skeletal/chemistry , Sensation , Sheep/physiology , Time Factors
6.
Nat Med ; 21(9): 1076-84, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26280121

ABSTRACT

Tumor necrosis factor-α (TNF-α), one of the major stress-induced proinflammatory cytokines, is upregulated in the heart after tissue injury, and its sustained expression can contribute to the development of heart failure. Whether TNF-α also exerts cytoprotective effects in heart failure is not known. Here we provide evidence for a cardioprotective function of TNF-α in a genetic heart failure model, desmin-deficient mice. The cardioprotective effects of TNF-α are a consequence of nuclear factor-κB (NF-κB)-mediated ectopic expression in cardiomyocytes of keratin 8 (K8) and keratin 18 (K18), two epithelial-specific intermediate filament proteins. In cardiomyocytes, K8 and K18 (K8/K18) formed an alternative cytoskeletal network that localized mainly at intercalated discs (IDs) and conferred cardioprotection by maintaining normal ID structure and mitochondrial integrity and function. Ectopic induction of K8/K18 expression in cardiomyocytes also occurred in other genetic and experimental models of heart failure. Loss of the K8/K18 network resulted in a maladaptive cardiac phenotype following transverse aortic constriction. In human failing myocardium, where TNF-α expression is upregulated, K8/K18 were also ectopically expressed and localized primarily at IDs, which did not contain detectable amounts of desmin. Thus, TNF-α- and NF-κB-mediated formation of an alternative, stress-induced intermediate filament cytoskeleton has cardioprotective function in mice and potentially in humans.


Subject(s)
Keratin-18/physiology , Keratin-8/physiology , Tumor Necrosis Factor-alpha/physiology , Animals , Cardiomegaly/prevention & control , Desmin/physiology , Female , Humans , Male , Mice , Mice, Inbred C57BL , NF-kappa B/physiology
7.
Clin Exp Pharmacol Physiol ; 41(1): 89-97, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24117876

ABSTRACT

1. Transgenic (TG) mice overexpressing an arg120gly missense mutation in heat shock protein B5 (HSPB5; i.e. R120G TG mice) exhibit desmin-related cardiomyopathy. Recently, the cardioprotective effect of nicorandil has been shown to prolong the survival of R120G TG mice. However, whether the TG mice exhibit ventricular arrhythmias and whether nicorandil can inhibit these arrhythmias remain unknown. In the present study we examined the effects of chronic nicorandil administration on ventricular electrical remodelling and arrhythmias in R120G TG mice. 2. Mice were administered nicorandil (15 mg/kg per day) or vehicle (water) orally from 5 to 30 weeks of age. Electrocardiograms (ECG) and optical action potentials were recorded from R120G TG mouse hearts. In addition, the expression of ventricular connexin 43 and the cardiac Na(+) channel Nav1.5 was examined in TG mice. 3. All ECG parameters tested were prolonged in R120G TG compared with non-transgenic (NTG) mice. Nicorandil improved the prolonged P, PQ and QRS intervals in R120G TG mice. Interestingly, impulse conduction slowing and increases in the expression of total and phosphorylated connexin 43 and Nav1.5 were observed in ventricles from R120G TG compared with NTG mice. Nicorandil improved ventricular impulse conduction slowing and normalized the increased protein expression levels of total and phosphorylated connexin 43, but not of Nav1.5, in R120G TG mouse hearts. Electrical rapid pacing at the ventricle induced ventricular tachyarrhythmias (VT) in six of eight R120G TG mouse hearts, but not in any of the eight nicorandil-treated R120G TG mouse hearts (P < 0.05). 4. These findings demonstrate that nicorandil inhibits cardiac electrical remodelling and that the prevention of VT by nicorandil is associated with normalization of connexin 43 expression in this model.


Subject(s)
Anti-Arrhythmia Agents/pharmacology , Cardiomyopathies/physiopathology , Desmin/physiology , Nicorandil/pharmacology , Tachycardia, Ventricular/prevention & control , Anesthesia , Animals , Blotting, Western , Body Weight/drug effects , Connexin 43/biosynthesis , Echocardiography , Electric Stimulation , Electrocardiography , Electrophysiological Phenomena/drug effects , Immunohistochemistry , In Vitro Techniques , Mice , Mice, Transgenic , NAV1.5 Voltage-Gated Sodium Channel/biosynthesis , Organ Size/drug effects
8.
Cell Death Differ ; 20(8): 1116-23, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23686136

ABSTRACT

Tissues of the adult organism maintain the homeostasis and respond to injury by means of progenitor/stem cell compartments capable to give rise to appropriate progeny. In organs composed by histotypes of different embryological origins (e.g. the liver), the tissue turnover may in theory involve different stem/precursor cells able to respond coordinately to physiological or pathological stimuli. In the liver, a progenitor cell compartment, giving rise to hepatocytes and cholangiocytes, can be activated by chronic injury inhibiting hepatocyte proliferation. The precursor compartment guaranteeing turnover of hepatic stellate cells (HSCs) (perisinusoidal cells implicated with the origin of the liver fibrosis) in adult organ is yet unveiled. We show here that epithelial and mesenchymal liver cells (hepatocytes and HSCs) may arise from a common progenitor. Sca+ murine progenitor cells were found to coexpress markers of epithelial and mesenchymal lineages and to give rise, within few generations, to cells that segregate the lineage-specific markers into two distinct subpopulations. Notably, these progenitor cells, clonally derived, when transplanted in healthy livers, were found to generate epithelial and mesenchymal liver-specific derivatives (i.e. hepatocytes and HSCs) properly integrated in the liver architecture. These evidences suggest the existence of a 'bona fide' organ-specific meso-endodermal precursor cell, thus profoundly modifying current models of adult progenitor commitment believed, so far, to be lineage-restricted. Heterotopic transplantations, which confirm the dual differentiation potentiality of those cells, indicates as tissue local cues are necessary to drive a full hepatic differentiation. These data provide first evidences for an adult stem/precursor cell capable to differentiate in both parenchymal and non-parenchymal organ-specific components and candidate the liver as the instructive site for the reservoir compartment of HSC precursors as yet non-localized in the adult.


Subject(s)
Cell Differentiation/physiology , Epithelial Cells/cytology , Liver/cytology , Mesenchymal Stem Cells/cytology , Stem Cells/cytology , Animals , Cell Line , Cell Lineage , Cell Proliferation , Cells, Cultured , Desmin/physiology , Epithelial Cells/physiology , Glial Fibrillary Acidic Protein , In Vitro Techniques , Liver/physiology , Mesenchymal Stem Cells/physiology , Mice , Mice, Nude , Models, Animal , Nerve Tissue Proteins/physiology , Stem Cell Transplantation , Stem Cells/physiology
9.
Am J Physiol Cell Physiol ; 302(11): C1609-20, 2012 Jun 01.
Article in English | MEDLINE | ID: mdl-22442138

ABSTRACT

Skeletal muscle is a dynamic composite of proteins that responds to both internal and external cues to facilitate muscle adaptation. In cases of disease or altered use, these messages can be distorted resulting in myopathic conditions such as fibrosis. In this work, we describe a mild and progressive fibrotic adaptation in skeletal muscle lacking the cytoskeletal intermediate filament protein desmin. Muscles lacking desmin become progressively stiffer, accumulate increased collagen, and increase expression of genes involved in extracellular matrix turnover. Additionally, in the absence of desmin, skeletal muscle is in an increased state of inflammation and regeneration as indicated by increased centrally nucleated fibers, elevated inflammation and regeneration related gene expression, and increased numbers of inflammatory cells. These data suggest a potential link between increased cellular damage and the development of fibrosis in muscles lacking the cytoskeletal support of the desmin filament network.


Subject(s)
Desmin/deficiency , Desmin/physiology , Intermediate Filaments/pathology , Muscle Fibers, Skeletal/pathology , Muscle, Skeletal/pathology , Animals , Desmin/genetics , Extracellular Matrix/metabolism , Extracellular Matrix/pathology , Fibrosis , Intermediate Filaments/genetics , Mice , Mice, Knockout , Muscle, Skeletal/metabolism , Muscle, Skeletal/physiology , Muscular Diseases/genetics , Muscular Diseases/pathology
10.
J Biomed Biotechnol ; 2012: 704061, 2012.
Article in English | MEDLINE | ID: mdl-22287836

ABSTRACT

In skeletal muscle fibers, forces must be transmitted between the plasma membrane and the intracellular contractile lattice, and within this lattice between adjacent myofibrils. Based on their prevalence, biomechanical properties and localization, desmin and keratin intermediate filaments (IFs) are likely to participate in structural connectivity and force transmission. We examined the passive load-bearing response of single fibers from the extensor digitorum longus (EDL) muscles of young (3 months) and aged (10 months) wild-type, desmin-null, K19-null, and desmin/K19 double-null mice. Though fibers are more compliant in all mutant genotypes compared to wild-type, the structural response of each genotype is distinct, suggesting multiple mechanisms by which desmin and keratin influence the biomechanical properties of myofibers. This work provides additional insight into the influences of IFs on structure-function relationships in skeletal muscle. It may also have implications for understanding the progression of desminopathies and other IF-related myopathies.


Subject(s)
Desmin/genetics , Keratin-19/genetics , Muscle, Skeletal/chemistry , Structure-Activity Relationship , Aging , Animals , Cell Membrane/chemistry , Cell Membrane/physiology , Cytoskeleton/chemistry , Cytoskeleton/physiology , Desmin/physiology , Female , Intermediate Filaments/chemistry , Intermediate Filaments/physiology , Keratin-19/physiology , Mice , Mice, Transgenic , Muscle, Skeletal/physiology , Muscular Diseases/genetics , Muscular Diseases/pathology , Weight-Bearing
11.
FASEB J ; 26(1): 137-48, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21982947

ABSTRACT

Intermediate filaments (IFs) in cardiomyocytes consist primarily of desmin, surround myofibrils at Z disks, and transmit forces from the contracting myofilaments to the cell surface through costameres at the sarcolemma and desmosomes at intercalated disks. Synemin is a type IV IF protein that forms filaments with desmin and also binds α-actinin and vinculin. Here we examine the roles and expression of the α and ß forms of synemin in developing rat cardiomyocytes. Quantitative PCR showed low levels of expression for both synemin mRNAs, which peaked at postnatal day 7. Synemin was concentrated at sites of cell-cell adhesion and at Z disks in neonatal cardiomyocytes. Overexpression of the individual isoforms showed that α-synemin preferentially localized to cell-cell junctions, whereas ß-synemin was primarily at the level of Z disks. An siRNA targeted to both synemin isoforms reduced protein expression in cardiomyocytes by 70% and resulted in a failure of desmin to align with Z disks and disrupted cell-cell junctions, with no effect on sarcomeric organization. Solubility assays showed that ß-synemin was soluble and interacted with sarcomeric α-actinin by coimmunoprecipitation, while α-synemin and desmin were insoluble. We conclude that ß-synemin mediates the association of desmin IFs with Z disks, whereas α-synemin stabilizes junctional complexes between cardiomyocytes.


Subject(s)
Desmin/physiology , Intercellular Junctions/physiology , Intermediate Filament Proteins/physiology , Myocytes, Cardiac/cytology , Myocytes, Cardiac/physiology , Actinin/metabolism , Animals , Animals, Newborn , Cell Adhesion/physiology , Intermediate Filament Proteins/chemistry , Intermediate Filament Proteins/genetics , Intermediate Filaments/physiology , Isomerism , Primary Cell Culture , RNA, Messenger/metabolism , RNA, Small Interfering/pharmacology , Rats , Sarcomeres/physiology , Solubility , Vinculin/metabolism
12.
Biol Aujourdhui ; 205(3): 163-77, 2011.
Article in French | MEDLINE | ID: mdl-21982405

ABSTRACT

Desmin, the muscle-specific intermediate filament protein, is one of the earliest markers expressed in all muscle tissues during development. It forms a three-dimensional scaffold around the myofibril Z-disc and connects the entire contractile apparatus to the subsarcolemmal cytoskeleton, the nuclei and other cytoplasmic organelles. Desmin is essential for tensile strength and muscle integrity. In humans, disorganization of the desmin network is associated with cardiac and/or skeletal myopathies characterized by accumulation of desmin-containing aggregates in the cells. Currently, 49 mutations have been identified in desmin gene. The majority of these mutations alter desmin filament assembly process through different molecular mechanisms and also its interaction with its protein partners. Here, we will give an overview of desmin network organization as well as the impact of desmin mutations on this process. Furthermore, we will discuss the different molecular mechanisms implicated in perturbation of the desmin filament assembly process.


Subject(s)
Desmin/physiology , Muscular Dystrophies/genetics , Myofibrils/pathology , Myopathies, Structural, Congenital/genetics , Amino Acid Sequence , Cardiomyopathies/genetics , Cardiomyopathies/pathology , Desmin/chemistry , Desmin/genetics , Humans , Models, Molecular , Molecular Sequence Data , Morphogenesis , Muscle Cells/ultrastructure , Muscle Contraction , Muscular Dystrophies/pathology , Mutation , Myocytes, Cardiac/ultrastructure , Myofibrils/chemistry , Myopathies, Structural, Congenital/pathology , Protein Conformation , Protein Structure, Tertiary , Structure-Activity Relationship , Tensile Strength
13.
Am J Physiol Heart Circ Physiol ; 301(4): H1220-8, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21784990

ABSTRACT

The intermediate filament protein desmin is an integral component of the cardiomyocyte and serves to maintain the overall structure and cytoskeletal organization within striated muscle cells. Desmin-related myopathy can be caused by mutations in desmin or associated proteins, which leads to intracellular accumulation of misfolded protein and production of soluble pre-amyloid oligomers, which leads to weakened skeletal and cardiac muscle. In this review, we examine the cellular phenotypes in relevant animal models of desmin-related cardiomyopathy. These models display characteristic sarcoplasmic protein aggregates. Aberrant protein aggregation leads to mitochondrial dysfunction, abnormal metabolism, and altered cardiomyocyte structure. These deficits to cardiomyocyte function may stem from impaired cellular proteolytic mechanisms. The data obtained from these models allow a more complete picture of the pathology in desmin-related cardiomyopathy to be described. Moreover, these studies highlight the importance of desmin in maintaining cardiomyocyte structure and illustrate how disrupting this network can be deleterious to the heart. We emphasize the similarities observed between desmin-related cardiomyopathy and other protein conformational disorders and speculate that therapies to treat this disease may be broadly applicable to diverse protein aggregation-based disorders.


Subject(s)
Cardiomyopathies/pathology , Desmin/physiology , Animals , Cardiomyopathies/metabolism , Cell Death/physiology , Desmin/metabolism , Humans , Inclusion Bodies/physiology , Mitochondria, Heart/physiology , Oxidative Stress/physiology , Protein Unfolding
14.
Circ Res ; 109(3): 296-308, 2011 Jul 22.
Article in English | MEDLINE | ID: mdl-21659648

ABSTRACT

RATIONALE: Recent studies suggest an important role of autophagy in protection against αB-crystallin-based (CryAB(R120G)) desmin-related cardiomyopathies (DRC), but this has not been demonstrated in a different model of cardiac proteinopathy. Mechanisms underlying the response of cardiomyocytes to proteotoxic stress remain incompletely understood. OBJECTIVE: Our first objective was to determine whether and how the autophagic activity is changed in a mouse model of desminopathy. We also investigated the role of p62 in the protein quality control of cardiomyocytes. METHODS AND RESULTS: Using an autophagosome reporter and determining changes in LC3-II protein levels in response to lysosomal inhibition, we found significantly increased autophagic flux in mouse hearts with transgenic overexpression of a DRC-linked mutant desmin. Similarly, autophagic flux was increased in cultured neonatal rat ventricular myocytes (NRVMs) expressing a mutant desmin. Suppression of autophagy by 3-methyladenine increased, whereas enhancement of autophagy by rapamycin reduced the ability of a comparable level of mutant desmin overexpression to accumulate ubiquitinated proteins in NRVMs. Furthermore, p62 mRNA and protein expression was significantly up-regulated in cardiomyocytes by transgenic overexpression of the mutant desmin or CryAB(R120G) both in intact mice and in vitro. The p62 depletion impaired aggresome and autophagosome formation, exacerbated cell injury, and decreased cell viability in cultured NRVMs expressing the misfolded proteins. CONCLUSIONS: Autophagic flux is increased in desminopathic hearts, and as previously suggested in CryAB(R120G)-based DRC, this increased autophagic flux serves as an adaptive response to overexpression of misfolded proteins. The p62 is up-regulated in mouse proteinopathic hearts. The p62 promotes aggresome formation and autophagy activation and protects cardiomyocytes against proteotoxic stress.


Subject(s)
Autophagy/physiology , Cardiomyopathies/pathology , Desmin/genetics , Myocytes, Cardiac/pathology , Myocytes, Cardiac/physiology , Proteostasis Deficiencies/pathology , Transcription Factors/physiology , Adaptation, Physiological/physiology , Animals , Cardiomyopathies/genetics , Cardiomyopathies/physiopathology , Cells, Cultured , Desmin/physiology , Genes, Reporter , Mice , Mice, Transgenic , Microscopy, Electron , Mutagenesis/physiology , Myocytes, Cardiac/ultrastructure , Proteostasis Deficiencies/genetics , Proteostasis Deficiencies/physiopathology , Rats , Stress, Physiological/physiology , Transcription Factor TFIIH , Transcription Factors/genetics , Ubiquitination/physiology , alpha-Crystallin B Chain/genetics , alpha-Crystallin B Chain/physiology
16.
J Clin Invest ; 121(1): 70-85, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21135508

ABSTRACT

Muscle contraction relies on a highly organized intracellular network of membrane organelles and cytoskeleton proteins. Among the latter are the intermediate filaments (IFs), a large family of proteins mutated in more than 30 human diseases. For example, mutations in the DES gene, which encodes the IF desmin, lead to desmin-related myopathy and cardiomyopathy. Here, we demonstrate that myotubularin (MTM1), which is mutated in individuals with X-linked centronuclear myopathy (XLCNM; also known as myotubular myopathy), is a desmin-binding protein and provide evidence for direct regulation of desmin by MTM1 in vitro and in vivo. XLCNM-causing mutations in MTM1 disrupted the MTM1-desmin complex, resulting in abnormal IF assembly and architecture in muscle cells and both mouse and human skeletal muscles. Adeno-associated virus-mediated ectopic expression of WT MTM1 in Mtm1-KO muscle reestablished normal desmin expression and localization. In addition, decreased MTM1 expression and XLCNM-causing mutations induced abnormal mitochondrial positioning, shape, dynamics, and function. We therefore conclude that MTM1 is a major regulator of both the desmin cytoskeleton and mitochondria homeostasis, specifically in skeletal muscle. Defects in IF stabilization and mitochondrial dynamics appear as common physiopathological features of centronuclear myopathies and desmin-related myopathies.


Subject(s)
Desmin/physiology , Muscle, Skeletal/physiology , Protein Tyrosine Phosphatases, Non-Receptor/physiology , Animals , Cell Line , Desmin/genetics , Humans , In Vitro Techniques , Intermediate Filaments/physiology , Intermediate Filaments/ultrastructure , Mice , Mice, Knockout , Microscopy, Electron, Transmission , Mitochondria, Muscle/physiology , Mitochondria, Muscle/ultrastructure , Models, Molecular , Muscle, Skeletal/ultrastructure , Mutation , Myopathies, Structural, Congenital/genetics , Myopathies, Structural, Congenital/physiopathology , Protein Interaction Domains and Motifs , Protein Tyrosine Phosphatases, Non-Receptor/deficiency , Protein Tyrosine Phosphatases, Non-Receptor/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism
17.
Cardiovasc Res ; 85(2): 253-62, 2010 Jan 15.
Article in English | MEDLINE | ID: mdl-19696071

ABSTRACT

Protein quality control (PQC) depends on elegant collaboration between molecular chaperones and targeted proteolysis in the cell. The latter is primarily carried out by the ubiquitin-proteasome system, but recent advances in this area of research suggest a supplementary role for the autophagy-lysosomal pathway in PQC-related proteolysis. The (patho)physiological significance of PQC in the heart is best illustrated in cardiac proteinopathy, which belongs to a family of cardiac diseases caused by expression of aggregation-prone proteins in cardiomyocytes. Cardiac proteasome functional insufficiency (PFI) is best studied in desmin-related cardiomyopathy, a bona fide cardiac proteinopathy. Emerging evidence suggests that many common forms of cardiomyopathy may belong to proteinopathy. This review focuses on examining current evidence, as it relates to the hypothesis that PFI impairs PQC in cardiomyocytes and contributes to the progression of cardiac proteinopathies to heart failure.


Subject(s)
Autophagy , Cardiomyopathies/etiology , Proteasome Endopeptidase Complex/physiology , Proteins/metabolism , Ubiquitin/metabolism , Animals , Desmin/physiology , Heart Failure/etiology , Humans , Molecular Chaperones/physiology , Myocytes, Cardiac/metabolism , Proteasome Inhibitors , Proteins/chemistry
18.
J Clin Invest ; 119(7): 1806-13, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19587455

ABSTRACT

Muscle fiber deterioration resulting in progressive skeletal muscle weakness, heart failure, and respiratory distress occurs in more than 20 inherited myopathies. As discussed in this Review, one of the newly identified myopathies is desminopathy, a disease caused by dysfunctional mutations in desmin, a type III intermediate filament protein, or alphaB-crystallin, a chaperone for desmin. The range of clinical manifestations in patients with desminopathy is wide and may overlap with those observed in individuals with other myopathies. Awareness of this disease needs to be heightened, diagnostic criteria reliably outlined, and molecular testing readily available; this would ensure prevention of sudden death from cardiac arrhythmias and other complications.


Subject(s)
Cardiomyopathies/etiology , Desmin/physiology , Muscular Diseases/etiology , Animals , Cardiomyopathies/diagnosis , Cardiomyopathies/therapy , Desmin/chemistry , Desmin/genetics , Disease Models, Animal , Humans , Muscular Diseases/diagnosis , Muscular Diseases/therapy , Mutation , Phenotype , Protein Structure, Secondary , alpha-Crystallin B Chain/genetics
19.
J Cell Biol ; 181(5): 761-75, 2008 Jun 02.
Article in English | MEDLINE | ID: mdl-18519735

ABSTRACT

We explored the involvement of the muscle-specific intermediate filament protein desmin in the model of tumor necrosis factor alpha (TNF-alpha)-induced cardiomyopathy. We demonstrate that in mice overexpressing TNF-alpha in the heart (alpha-myosin heavy chain promoter-driven secretable TNF-alpha [MHCsTNF]), desmin is modified, loses its intercalated disk (ID) localization, and forms aggregates that colocalize with heat shock protein 25 and ubiquitin. Additionally, other ID proteins such as desmoplakin and beta-catenin show similar localization changes in a desmin-dependent fashion. To address underlying mechanisms, we examined whether desmin is a substrate for caspase-6 in vivo as well as the implications of desmin cleavage in MHCsTNF mice. We generated transgenic mice with cardiac-restricted expression of a desmin mutant (D263E) and proved that it is resistant to caspase cleavage in the MHCsTNF myocardium. The aggregates are diminished in these mice, and D263E desmin, desmoplakin, and beta-catenin largely retain their proper ID localization. Importantly, D263E desmin expression attenuated cardiomyocyte apoptosis, prevented left ventricular wall thinning, and improved the function of MHCsTNF hearts.


Subject(s)
Desmin/physiology , Heart Failure/metabolism , Tumor Necrosis Factor-alpha/metabolism , Animals , Apoptosis , Cardiomyopathies/pathology , Caspase 6/metabolism , Desmin/metabolism , Desmoplakins/metabolism , Mice , Mice, Transgenic , Models, Biological , Mutation , Myocardium/metabolism , Ubiquitin/chemistry , beta Catenin/metabolism
20.
Am J Physiol Cell Physiol ; 295(2): C324-31, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18562479

ABSTRACT

Role of the intermediate filament protein desmin in hypertrophy of smooth muscle was examined in desmin-deficient mice (Des(-/-)). A partial obstruction of the urethra was created, and after 9-19 days bladder weight increased approximately threefold in both Des(-/-) and wild type (Des(+/+)) animals. Bladder growth was associated with the synthesis of actin and myosin. In the hypertrophic Des(+/+) bladder, the relative content of desmin increased. In Des(-/-)mice, desmin was absent. No alterations in the amount of vimentin were observed. Although Des(-/-) obstructed bladders were capable of growth, they had structural changes with a partial disruption of the wall. Des(-/-)bladders had slightly lower passive stress and significantly lower active stress compared with Des(+/+). Des(-/-)preparations had lower shortening velocity. During hypertrophy, these structural and mechanical alterations in the Des(-/-)urinary bladder became more pronounced. In conclusion, desmin in the bladder smooth muscle is not needed for growth but has a role in active force transmission and maintenance of wall structure.


Subject(s)
Desmin/physiology , Muscle, Smooth/physiopathology , Urinary Bladder/physiopathology , Actins/analysis , Animals , Biomechanical Phenomena , Desmin/analysis , Desmin/genetics , Disease Models, Animal , Female , Hypertrophy , Intermediate Filament Proteins/analysis , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Electron, Transmission , Muscle Contraction/physiology , Muscle, Smooth/ultrastructure , Myocardium/pathology , Myosins/analysis , Organ Size , Urethral Obstruction/physiopathology , Urinary Bladder/chemistry , Urinary Bladder/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...