Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
BioDrugs ; 38(1): 95-119, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37917377

ABSTRACT

Duchenne muscular dystrophy is a devastating disease that leads to progressive muscle loss and premature death. While medical management focuses mostly on symptomatic treatment, decades of research have resulted in first therapeutics able to restore the affected reading frame of dystrophin transcripts or induce synthesis of a truncated dystrophin protein from a vector, with other strategies based on gene therapy and cell signaling in preclinical or clinical development. Nevertheless, recent reports show that potentially therapeutic dystrophins can be immunogenic in patients. This raises the question of whether a dystrophin paralog, utrophin, could be a more suitable therapeutic protein. Here, we compare dystrophin and utrophin amino acid sequences and structures, combining published data with our extended in silico analyses. We then discuss these results in the context of therapeutic approaches for Duchenne muscular dystrophy. Specifically, we focus on strategies based on delivery of micro-dystrophin and micro-utrophin genes with recombinant adeno-associated viral vectors, exon skipping of the mutated dystrophin pre-mRNAs, reading through termination codons with small molecules that mask premature stop codons, dystrophin gene repair by clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated protein 9 (CRISPR/Cas9)-mediated genetic engineering, and increasing utrophin levels. Our analyses highlight the importance of various dystrophin and utrophin domains in Duchenne muscular dystrophy treatment, providing insights into designing novel therapeutic compounds with improved efficacy and decreased immunoreactivity. While the necessary actin and ß-dystroglycan binding sites are present in both proteins, important functional distinctions can be identified in these domains and some other parts of truncated dystrophins might need redesigning due to their potentially immunogenic qualities. Alternatively, therapies based on utrophins might provide a safer and more effective approach.


Subject(s)
Dystrophin , Muscular Dystrophy, Duchenne , Humans , Dystrophin/genetics , Dystrophin/metabolism , Genetic Therapy/methods , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/therapy , Muscular Dystrophy, Duchenne/metabolism , Utrophin/genetics , Utrophin/metabolism
2.
Psychiatr Pol ; 56(3): 551-570, 2022 Jun 30.
Article in English, Polish | MEDLINE | ID: mdl-36342985

ABSTRACT

OBJECTIVES: One of the most important questions in personality psychology and psychopathology is whether working models of attachment change during lifetime. It is assumed that early childhood experiences influence the formation of secure or insecure internal working models of attachment The belief that attachment representations formed in childhood are relatively stable is no longer so obvious: new reports have appeared, according to which important life experiences may lead to a change in attachment style from insecure to secure or the other way around. The main aim of the present project was to investigate whether and in what manner positive and negative life experiences lead to changes in internal working models of attachment. METHODS: The specific style of attachment to mother, father, partner, and friend was measured with the ECR-RS, while global attachment was assessed based on the SAAM and ECR-RS G. The number and intensity of positive and negative life experiences was assessed by means of the LES-M. We analyzed the results for a sample of 156 adults. RESULTS: The study revealed significant relations between the level of positive life experiences and global and specific styles of attachment to a friend and partner. However, no significant relationships were observed between the intensity of negative life events and attachment style. Between-group comparisons showed that in the group in which a change of attachment style from insecure to secure had taken place the experience of positive events was significantly stronger than in the group in which there had been no such change. The compared groups did not differ in the level of negative life experiences. CONCLUSIONS: The study supports the hypothesis about a change of global attachment style and selected specific aspects of insecure attachment to a secure style as a result of experiencing positive life events.


Subject(s)
Life Change Events , Object Attachment , Child, Preschool , Adult , Humans
3.
Nucleic Acids Res ; 49(16): 9479-9495, 2021 09 20.
Article in English | MEDLINE | ID: mdl-34358321

ABSTRACT

Fragile X-associated tremor/ataxia syndrome (FXTAS) is a late-onset neurodegenerative disorder caused by a limited expansion of CGG repeats in the FMR1 gene. Degeneration of neurons in FXTAS cell models can be triggered by accumulation of polyglycine protein (FMRpolyG), a by-product of translation initiated upstream to the repeats. Specific aims of our work included testing if naphthyridine-based molecules could (i) block FMRpolyG synthesis by binding to CGG repeats in RNA, (ii) reverse pathological alterations in affected cells and (iii) preserve the content of FMRP, translated from the same FMR1 mRNA. We demonstrate that cyclic mismatch binding ligand CMBL4c binds to RNA structure formed by CGG repeats and attenuates translation of FMRpolyG and formation of nuclear inclusions in cells transfected with vectors expressing RNA with expanded CGG repeats. Moreover, our results indicate that CMBL4c delivery can reduce FMRpolyG-mediated cytotoxicity and apoptosis. Importantly, its therapeutic potential is also observed once the inclusions are already formed. We also show that CMBL4c-driven FMRpolyG loss is accompanied by partial FMRP reduction. As complete loss of FMRP induces FXS in children, future experiments should aim at evaluation of CMBL4c therapeutic intervention in differentiated tissues, in which FMRpolyG translation inhibition might outweigh adverse effects related to FMRP depletion.


Subject(s)
Ataxia/genetics , Fragile X Mental Retardation Protein/genetics , Fragile X Syndrome/genetics , Naphthyridines/pharmacology , Tremor/genetics , Trinucleotide Repeat Expansion/drug effects , Apoptosis/drug effects , Ataxia/drug therapy , Ataxia/pathology , Cell Proliferation/drug effects , Fragile X Mental Retardation Protein/antagonists & inhibitors , Fragile X Syndrome/drug therapy , Fragile X Syndrome/pathology , HeLa Cells , Humans , Ligands , Neurons/drug effects , Neurons/pathology , Peptides/genetics , Protein Biosynthesis/drug effects , Surface Plasmon Resonance , Tremor/drug therapy , Tremor/pathology , Trinucleotide Repeat Expansion/genetics , Trinucleotide Repeats/drug effects , Trinucleotide Repeats/genetics
4.
Cell Mol Life Sci ; 78(11): 4867-4891, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33825942

ABSTRACT

Duchenne muscular dystrophy (DMD) is a devastating chromosome X-linked disease that manifests predominantly in progressive skeletal muscle wasting and dysfunctions in the heart and diaphragm. Approximately 1/5000 boys and 1/50,000,000 girls suffer from DMD, and to date, the disease is incurable and leads to premature death. This phenotypic severity is due to mutations in the DMD gene, which result in the absence of functional dystrophin protein. Initially, dystrophin was thought to be a force transducer; however, it is now considered an essential component of the dystrophin-associated protein complex (DAPC), viewed as a multicomponent mechanical scaffold and a signal transduction hub. Modulating signal pathway activation or gene expression through epigenetic modifications has emerged at the forefront of therapeutic approaches as either an adjunct or stand-alone strategy. In this review, we propose a broader perspective by considering DMD to be a disease that affects myofibers and muscle stem (satellite) cells, as well as a disorder in which abrogated communication between different cell types occurs. We believe that by taking this systemic view, we can achieve safe and holistic treatments that can restore correct signal transmission and gene expression in diseased DMD tissues.


Subject(s)
Cell Communication , Dystrophin/metabolism , Muscular Dystrophy, Duchenne/pathology , Signal Transduction , Animals , Bone and Bones/metabolism , Dystrophin/chemistry , Dystrophin/genetics , Humans , Microvessels/metabolism , Muscle, Skeletal/metabolism , Muscular Dystrophy, Duchenne/metabolism , Muscular Dystrophy, Duchenne/therapy , Neurons/metabolism , Signal Transduction/genetics
5.
Clin Epigenetics ; 13(1): 13, 2021 01 19.
Article in English | MEDLINE | ID: mdl-33468200

ABSTRACT

Duchenne muscular dystrophy (DMD) is a multisystemic disorder that affects 1:5000 boys. The severity of the phenotype varies dependent on the mutation site in the DMD gene and the resultant dystrophin expression profile. In skeletal muscle, dystrophin loss is associated with the disintegration of myofibers and their ineffective regeneration due to defective expansion and differentiation of the muscle stem cell pool. Some of these phenotypic alterations stem from the dystrophin absence-mediated serine-threonine protein kinase 2 (MARK2) misplacement/downregulation in activated muscle stem (satellite) cells and neuronal nitric oxide synthase loss in cells committed to myogenesis. Here, we trace changes in DNA methylation, histone modifications, and expression of regulatory noncoding RNAs during muscle regeneration, from the stage of satellite cells to myofibers. Furthermore, we describe the abrogation of these epigenetic regulatory processes due to changes in signal transduction in DMD and point to therapeutic treatments increasing the regenerative potential of diseased muscles based on this acquired knowledge.


Subject(s)
DNA Methylation , Dystrophin/genetics , Epigenesis, Genetic , Genetic Therapy , Muscle, Skeletal/growth & development , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/physiopathology , Regeneration/genetics , Adolescent , Adult , Child , Child, Preschool , Disease Progression , Gene Expression Regulation , Humans , Infant , Infant, Newborn , Male , Muscular Dystrophy, Duchenne/therapy , Mutation , Phenotype , Young Adult
6.
Nucleic Acids Res ; 48(5): 2531-2543, 2020 03 18.
Article in English | MEDLINE | ID: mdl-31965181

ABSTRACT

Expansion of an unstable CTG repeat in the 3'UTR of the DMPK gene causes Myotonic Dystrophy type 1 (DM1). CUG-expanded DMPK transcripts (CUGexp) sequester Muscleblind-like (MBNL) alternative splicing regulators in ribonuclear inclusions (foci), leading to abnormalities in RNA processing and splicing. To alleviate the burden of CUGexp, we tested therapeutic approach utilizing antisense oligonucleotides (AONs)-mediated DMPK splice-switching and degradation of mutated pre-mRNA. Experimental design involved: (i) skipping of selected constitutive exons to induce frameshifting and decay of toxic mRNAs by an RNA surveillance mechanism, and (ii) exclusion of the alternative exon 15 (e15) carrying CUGexp from DMPK mRNA. While first strategy failed to stimulate DMPK mRNA decay, exclusion of e15 enhanced DMPK nuclear export but triggered accumulation of potentially harmful spliced out pre-mRNA fragment containing CUGexp. Neutralization of this fragment with antisense gapmers complementary to intronic sequences preceding e15 failed to diminish DM1-specific spliceopathy due to AONs' chemistry-related toxicity. However, intronic gapmers alone reduced the level of DMPK mRNA and mitigated DM1-related cellular phenotypes including spliceopathy and nuclear foci. Thus, a combination of the correct chemistry and experimental approach should be carefully considered to design a safe AON-based therapeutic strategy for DM1.


Subject(s)
Alternative Splicing/genetics , Myotonic Dystrophy/genetics , Myotonic Dystrophy/therapy , Myotonin-Protein Kinase/genetics , Oligonucleotides, Antisense/therapeutic use , RNA Precursors/genetics , RNA Stability/genetics , Active Transport, Cell Nucleus , Cell Nucleus/metabolism , Exons/genetics , Humans , Myotonin-Protein Kinase/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Trinucleotide Repeat Expansion/genetics
7.
Front Genet ; 9: 216, 2018.
Article in English | MEDLINE | ID: mdl-29971092

ABSTRACT

Fragile X-associated tremor/ataxia syndrome (FXTAS) is a late-onset neurodegenerative disorder caused by expanded CGG (CGGexp) trinucleotides in the 5'UTR of the FMR1 gene encoding fragile X mental retardation protein (FMRP). The patients, with the number of the repeats ranging from 55 to 200, show specific manifestation of clinical symptoms that include intention tremor, gait ataxia, cognitive deficits, and brain atrophy. Accumulation of toxic polyglycine (FMRpolyG), a by-product of the CGGexp repeat-associated non-ATG (RAN) translation, is considered to be one of the main factors triggering neurodegenerative processes in FXTAS patients. Nevertheless, the nature of the FMRpolyG-induced cell damage, especially in the context of its soluble and inclusion-associated forms, is still elusive. Targeting either biosynthesis, cellular stability or aggregation capacity of toxic FMRpolyG could be considered as a potential therapeutic strategy for FXTAS. Therefore, we tested a variety of quantitative methods based on forced expression of genetic constructs carrying CGGexp repeats in the context of the FMR1 5'UTR fused to GFP, mCherry or Firefly luciferase gene in or out of frame to the polyglycine encoding sequence. We show that FMRpolyG translation either from native or an AUG-induced start codon as well as the translation yield of the FMRP open reading frame equivalent located downstream of the CGGexp element can be effectively estimated using fluorescence microscopy, flow cytometry or luciferase assay. We also quantitatively estimated soluble fraction and insoluble form of FMRpolyG aggregated in foci using an electrophoretic separation of cell lysates and fluorescence microscopy, respectively. Importantly, we show that dependent on a fusion tag, FMRpolyG has a different potential for aggregate formation. Our established protocols enable sensitive tracking of FMRP and FMRpolyG quantitative and qualitative changes after treatment with potential therapeutic agents for FXTAS. Furthermore, they can be modified for application to other RAN translation- and aggregation-related diseases.

8.
RNA Biol ; 15(1): 1-8, 2018 01 02.
Article in English | MEDLINE | ID: mdl-28949831

ABSTRACT

Muscleblind-like (MBNL) proteins bind to hundreds of pre- and mature mRNAs to regulate their alternative splicing, alternative polyadenylation, stability and subcellular localization. Once MBNLs are withheld from transcript regulation, cellular machineries generate products inapt for precise embryonal/adult developmental tasks and myotonic dystrophy, a devastating multi-systemic genetic disorder, develops. We have recently demonstrated that all three MBNL paralogs are capable of fine-tuning cellular content of one of the three MBNL paralogs, MBNL1, by binding to the first coding exon (e1) of its pre-mRNA. Intriguingly, this autoregulatory feedback loop grounded on alternative splicing of e1 appears to play a crucial role in delaying the onset of myotonic dystrophy. Here, we describe this process in the context of other autoregulatory and regulatory loops that maintain the content and diverse functions of MBNL proteins at optimal level in health and disease, thus supporting the overall cellular homeostasis.


Subject(s)
Alternative Splicing/genetics , Myotonic Dystrophy/genetics , RNA-Binding Proteins/genetics , Exons/genetics , Feedback, Physiological , Gene Expression Regulation, Developmental , Humans , Myotonic Dystrophy/pathology , Polyadenylation/genetics , Protein Binding , RNA Precursors/genetics , RNA Splicing Factors/genetics
9.
Nucleic Acids Res ; 45(4): 1760-1775, 2017 02 28.
Article in English | MEDLINE | ID: mdl-27903900

ABSTRACT

Muscleblind-like proteins (MBNLs) are regulators of RNA metabolism. During tissue differentiation the level of MBNLs increases, while their functional insufficiency plays a crucial role in myotonic dystrophy (DM). Deep sequencing of RNA molecules cross-linked to immunoprecipitated protein particles (CLIP-seq) revealed that MBNL1 binds to MBNL1 exon 1 (e1) encoding both the major part of 5΄UTR and an amino-terminal region of MBNL1 protein. We tested several hypotheses regarding the possible autoregulatory function of MBNL1 binding to its own transcript. Our data indicate that MBNLs induce skipping of e1 from precursor MBNL1 mRNA and that e1 exclusion may impact transcript association with polysomes and translation. Furthermore, e1-deficient protein isoform lacking the first two zinc fingers is highly unstable and its EGFP fusion protein has severely compromised splicing activity. We also show that MBNL1 can be transcribed from three different promoters and that the transcription initiation site determines the mode of e1 regulation. Taken together, we demonstrate that MBNL proteins control steady-state levels of MBNL1 through an interaction with e1 in its precursor mRNA. Insights from our study open a new avenue in therapies against DM based on manipulation of the transcription initiation site and e1 splicing of MBNL1 mRNA.


Subject(s)
Alternative Splicing , Exons , Gene Expression Regulation , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Animals , Cell Line , Gene Expression , Gene Knockdown Techniques , Genes, Reporter , Humans , Mice , Nucleic Acid Conformation , Protein Binding , Protein Stability , RNA Isoforms , RNA Precursors/chemistry , RNA Precursors/genetics , RNA Precursors/metabolism
10.
Nucleic Acids Res ; 44(21): 10326-10342, 2016 Dec 01.
Article in English | MEDLINE | ID: mdl-27733504

ABSTRACT

Muscleblind-like (MBNL) proteins are critical RNA processing factors in development. MBNL activity is disrupted in the neuromuscular disease myotonic dystrophy type 1 (DM1), due to the instability of a non-coding microsatellite in the DMPK gene and the expression of CUG expansion (CUGexp) RNAs. Pathogenic interactions between MBNL and CUGexp RNA lead to the formation of nuclear complexes termed foci and prevent MBNL function in pre-mRNA processing. The existence of multiple MBNL genes, as well as multiple protein isoforms, raises the question of whether different MBNL proteins possess unique or redundant functions. To address this question, we coexpressed three MBNL paralogs in cells at equivalent levels and characterized both specific and redundant roles of these proteins in alternative splicing and RNA foci dynamics. When coexpressed in the same cells, MBNL1, MBNL2 and MBNL3 bind the same RNA motifs with different affinities. While MBNL1 demonstrated the highest splicing activity, MBNL3 showed the lowest. When forming RNA foci, MBNL1 is the most mobile paralog, while MBNL3 is rather static and the most densely packed on CUGexp RNA. Therefore, our results demonstrate that MBNL paralogs and gene-specific isoforms possess inherent functional differences, an outcome that could be enlisted to improve therapeutic strategies for DM1.


Subject(s)
RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Alternative Splicing , Binding Sites , Cell Line , Exons , Humans , Nucleotide Motifs , Position-Specific Scoring Matrices , Protein Binding , Protein Transport , RNA/chemistry , RNA/metabolism , RNA Isoforms , RNA-Binding Proteins/chemistry
11.
Nucleic Acids Res ; 42(17): 10873-87, 2014.
Article in English | MEDLINE | ID: mdl-25183524

ABSTRACT

Muscleblind-like (MBNL) proteins are key regulators of precursor and mature mRNA metabolism in mammals. Based on published and novel data, we explore models of tissue-specific MBNL interaction with RNA. We portray MBNL domains critical for RNA binding and splicing regulation, and the structure of MBNL's normal and pathogenic RNA targets, particularly in the context of myotonic dystrophy (DM), in which expanded CUG or CCUG repeat transcripts sequester several nuclear proteins including MBNLs. We also review the properties of MBNL/RNA complex, including recent data obtained from UV cross-linking and immunoprecipitation (CLIP-Seq), and discuss how this interaction shapes normal MBNL-dependent alternative splicing regulation. Finally, we review how this acquired knowledge about the pathogenic RNA structure and nature of MBNL sequestration can be translated into the design of therapeutic strategies against DM.


Subject(s)
Alternative Splicing , RNA, Messenger/metabolism , RNA-Binding Proteins/metabolism , Animals , Humans , Mice , Protein Binding , Protein Structure, Tertiary , RNA Precursors/chemistry , RNA Precursors/metabolism , RNA, Messenger/chemistry , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/genetics
12.
Article in English | MEDLINE | ID: mdl-25580445

ABSTRACT

Adeno-associated viral (AAV) vectors are becoming an important tool for gene therapy of numerous genetic and other disorders. Several recombinant AAV vectors (rAAV) have the ability to transduce striated muscles in a variety of animals following intramuscular and intravascular administration, and have attracted widespread interest for therapy of muscle disorders such as the muscular dystrophies. However, most studies have focused on the ability to transduce mature muscle cells, and have not examined the ability to target myogenic stem cells such as skeletal muscle satellite cells. Here we examined the relative ability of rAAV vectors derived from AAV6 to target myoblasts, myocytes and myotubes in culture and satellite cells and myofibers in vivo. AAV vectors are able to transduce proliferating myoblasts in culture, albeit with reduced efficiency relative to post-mitotic myocytes and myotubes. In contrast, quiescent satellite cells are refractory to transduction in adult mice. These results suggest that while muscle disorders characterized by myofiber regeneration can be slowed or halted by AAV transduction, little if any vector transduction can be obtained in myogenic stems cells that might other wise support ongoing muscle regeneration.

13.
Muscle Nerve ; 47(5): 649-63, 2013 May.
Article in English | MEDLINE | ID: mdl-23553671

ABSTRACT

Duchenne muscular dystrophy (DMD) is a devastating muscle disorder that affects 1 in 3,500 boys. Despite years of research and considerable progress in understanding the molecular mechanism of the disease and advancement of therapeutic approaches, there is no cure for DMD. The current treatment options are limited to physiotherapy and corticosteroids, and although they provide a substantial improvement in affected children, they only slow the course of the disorder. On a more optimistic note, more recent approaches either significantly alleviate or eliminate muscular dystrophy in murine and canine models of DMD and importantly, many of them are being tested in early phase human clinical trials. This review summarizes advancements that have been made in viral and nonviral gene therapy as well as stem cell therapy for DMD with a focus on the replacement and repair of the affected dystrophin gene.


Subject(s)
Genetic Therapy , Muscular Dystrophies/therapy , Stem Cell Transplantation , Humans , Muscular Dystrophies/genetics
14.
Prog Mol Biol Transl Sci ; 105: 83-111, 2012.
Article in English | MEDLINE | ID: mdl-22137430

ABSTRACT

The muscular dystrophies (MDs) represent a diverse collection of inherited human disorders, which affect to varying degrees skeletal, cardiac, and sometimes smooth muscle (Emery, 2002). To date, more than 50 different genes have been implicated as causing one or more types of MD (Bansal et al., 2003). In many cases, invaluable insights into disease mechanisms, structure and function of gene products, and approaches for therapeutic interventions have benefited from the study of animal models of the different MDs (Arnett et al., 2009). The large number of genes that are associated with MD and the tremendous number of animal models that have been developed preclude a complete discussion of each in the context of this review. However, we summarize here a number of the more commonly used models together with a mixture of different types of gene and MD, which serves to give a general overview of the value of animal models of MD for research and therapeutic development.


Subject(s)
Disease Models, Animal , Muscular Dystrophies/pathology , Animals , Calcium/metabolism , Humans , Muscle Proteins/chemistry , Muscle Proteins/metabolism , Muscular Dystrophies/metabolism
15.
Exp Cell Res ; 316(3): 297-313, 2010 Feb 01.
Article in English | MEDLINE | ID: mdl-19932097

ABSTRACT

BPAG1-b is the major muscle-specific isoform encoded by the dystonin gene, which expresses various protein isoforms belonging to the plakin protein family with complex, tissue-specific expression profiles. Recent observations in mice with either engineered or spontaneous mutations in the dystonin gene indicate that BPAG1-b serves as a cytolinker important for the establishment and maintenance of the cytoarchitecture and integrity of striated muscle. Here, we studied in detail its distribution in skeletal and cardiac muscles and assessed potential binding partners. BPAG1-b was detectable in vitro and in vivo as a high molecular mass protein in striated and heart muscle cells, co-localizing with the sarcomeric Z-disc protein alpha-actinin-2 and partially with the cytolinker plectin as well as with the intermediate filament protein desmin. Ultrastructurally, like alpha-actinin-2, BPAG1-b was predominantly localized at the Z-discs, adjacent to desmin-containing structures. BPAG1-b was able to form complexes with both plectin and alpha-actinin-2, and its NH(2)-terminus, which contains an actin-binding domain, directly interacted with that of plectin and alpha-actinin. Moreover, the protein level of BPAG1-b was reduced in muscle tissues from plectin-null mutant mice versus wild-type mice. These studies provide new insights into the role of BPAG1-b in the cytoskeletal organization of striated muscle.


Subject(s)
Actinin/metabolism , Carrier Proteins/metabolism , Cytoskeletal Proteins/metabolism , Myocardium/metabolism , Nerve Tissue Proteins/metabolism , Plectin/metabolism , Animals , Carrier Proteins/chemistry , Cell Extracts , Cells, Cultured , Cytoskeletal Proteins/chemistry , Dystonin , Humans , Immune Sera , Intermediate Filament Proteins/chemistry , Intermediate Filament Proteins/metabolism , Mice , Mice, Knockout , Muscle, Skeletal/metabolism , Muscle, Skeletal/ultrastructure , Myocardium/cytology , Myocardium/ultrastructure , Nerve Tissue Proteins/chemistry , Plectin/deficiency , Protein Binding , Protein Isoforms/chemistry , Protein Isoforms/metabolism , Protein Transport , Rats , Repetitive Sequences, Amino Acid
16.
J Cell Biol ; 181(4): 667-81, 2008 May 19.
Article in English | MEDLINE | ID: mdl-18490514

ABSTRACT

Dysfunction of plectin, a 500-kD cytolinker protein, leads to skin blistering and muscular dystrophy. Using conditional gene targeting in mice, we show that plectin deficiency results in progressive degenerative alterations in striated muscle, including aggregation and partial loss of intermediate filament (IF) networks, detachment of the contractile apparatus from the sarcolemma, profound changes in myofiber costameric cytoarchitecture, and decreased mitochondrial number and function. Analysis of newly generated plectin isoform-specific knockout mouse models revealed that IF aggregates accumulate in distinct cytoplasmic compartments, depending on which isoform is missing. Our data show that two major plectin isoforms expressed in muscle, plectin 1d and 1f, integrate fibers by specifically targeting and linking desmin IFs to Z-disks and costameres, whereas plectin 1b establishes a linkage to mitochondria. Furthermore, disruption of Z-disk and costamere linkages leads to the pathological condition of epidermolysis bullosa with muscular dystrophy. Our findings establish plectin as the major organizer of desmin IFs in myofibers and provide new insights into plectin- and desmin-related muscular dystrophies.


Subject(s)
Desmin/metabolism , Intermediate Filaments/metabolism , Muscle Fibers, Skeletal/metabolism , Plectin/metabolism , Sarcomeres/metabolism , Animals , Disease Progression , Intermediate Filaments/ultrastructure , Mice , Mice, Knockout , Mitochondria/pathology , Mitochondria/ultrastructure , Models, Biological , Muscle Contraction , Muscle Fibers, Skeletal/pathology , Muscle, Striated/pathology , Muscle, Striated/ultrastructure , Physical Endurance , Plectin/deficiency , Protein Isoforms/metabolism , Sarcolemma/pathology , Sarcolemma/ultrastructure , Survival Analysis
17.
Adv Exp Med Biol ; 642: 165-75, 2008.
Article in English | MEDLINE | ID: mdl-19181100

ABSTRACT

Myocytes are characterized by the presence of highly specialized cytoskeletal structures that are part of regularly spaced functional units distributed over long distances. In this chapter we discuss previously published evidence as well as novel data showing that the proper positioning and architecture of Z-disks and of sarcolemma-associated costameric structures are largely dependent on the cytolinker protein plectin and its associated intermediate filament (desmin) cytoskeleton. Deficiency in either plectin or desmin lead to muscular dystrophies of similar pathology. However, while in the absence of plectin, desmin networks collapse and form aggregates, when desmin is missing, plectin retains its typical localization. This suggests that plectin recruits and anchors desmin filaments to both Z-disks and costameres and thus is a key element for maintaining and reinforcing myocyte cytoarchitecture. We hypothesize that as an essential link of the Z-disk-costamere axis, plectin is likely to play also a crucial role in myofiber signaling.


Subject(s)
Muscle, Skeletal/metabolism , Plectin/metabolism , Animals , Epidermolysis Bullosa/genetics , Epidermolysis Bullosa/metabolism , Humans , Muscular Diseases/genetics , Muscular Diseases/metabolism , Muscular Diseases/pathology , Mutation/genetics , Plectin/deficiency , Plectin/genetics , Protein Binding
18.
J Cell Biol ; 176(7): 965-77, 2007 Mar 26.
Article in English | MEDLINE | ID: mdl-17389230

ABSTRACT

In skeletal muscle, the cytolinker plectin is prominently expressed at Z-disks and the sarcolemma. Alternative splicing of plectin transcripts gives rise to more than eight protein isoforms differing only in small N-terminal sequences (5-180 residues), four of which (plectins 1, 1b, 1d, and 1f) are found at substantial levels in muscle tissue. Using plectin isoform-specific antibodies and isoform expression constructs, we show the differential regulation of plectin isoforms during myotube differentiation and their localization to different compartments of muscle fibers, identifying plectins 1 and 1f as sarcolemma-associated isoforms, whereas plectin 1d localizes exclusively to Z-disks. Coimmunoprecipitation and in vitro binding assays using recombinant protein fragments revealed the direct binding of plectin to dystrophin (utrophin) and beta-dystroglycan, the key components of the dystrophin-glycoprotein complex. We propose a model in which plectin acts as a universal mediator of desmin intermediate filament anchorage at the sarcolemma and Z-disks. It also explains the plectin phenotype observed in dystrophic skeletal muscle of mdx mice and Duchenne muscular dystrophy patients.


Subject(s)
Dystroglycans/metabolism , Muscle Fibers, Skeletal/metabolism , Muscle, Skeletal/metabolism , Muscular Dystrophy, Animal/metabolism , Plectin/metabolism , Sarcolemma/metabolism , Animals , Cell Compartmentation/physiology , Cell Differentiation/physiology , Cells, Cultured , Cytoskeleton/metabolism , Cytoskeleton/ultrastructure , Desmin/metabolism , Humans , Immunohistochemistry , Intermediate Filaments/metabolism , Intermediate Filaments/ultrastructure , Mice , Mice, Inbred C57BL , Mice, Inbred mdx , Models, Biological , Muscle Fibers, Skeletal/pathology , Muscle Fibers, Skeletal/ultrastructure , Muscle, Skeletal/pathology , Muscle, Skeletal/physiopathology , Muscular Dystrophy, Animal/genetics , Muscular Dystrophy, Animal/physiopathology , Plectin/immunology , Protein Isoforms/immunology , Protein Isoforms/metabolism , Rats , Sarcolemma/pathology , Sarcolemma/ultrastructure , Utrophin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...