Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 615
Filter
1.
Hum Mol Genet ; 33(13): 1107-1119, 2024 Jun 21.
Article in English | MEDLINE | ID: mdl-38507070

ABSTRACT

The dystrophin-glycoprotein complex (DGC) plays a crucial role in maintaining the structural integrity of the plasma membrane and the neuromuscular junction. In this study, we investigated the impact of the deficiency of α-dystrobrevin (αdbn), a component of the DGC, on the homeostasis of intracellular organelles, specifically mitochondria and the sarcoplasmic reticulum (SR). In αdbn deficient muscles, we observed a significant increase in the membrane-bound ATP synthase complex levels, a marker for mitochondria in oxidative muscle fiber types compared to wild-type. Furthermore, examination of muscle fibers deficient in αdbn using electron microscopy revealed profound alterations in the organization of mitochondria and the SR within certain myofibrils of muscle fibers. This included the formation of hyper-branched intermyofibrillar mitochondria with extended connections, an extensive network spanning several myofibrils, and a substantial increase in the number/density of subsarcolemmal mitochondria. Concurrently, in some cases, we observed significant structural alterations in mitochondria, such as cristae loss, fragmentation, swelling, and the formation of vacuoles and inclusions within the mitochondrial matrix cristae. Muscles deficient in αdbn also displayed notable alterations in the morphology of the SR, along with the formation of distinct anomalous concentric SR structures known as whorls. These whorls were prevalent in αdbn-deficient mice but were absent in wild-type muscles. These results suggest a crucial role of the DGC αdbn in regulating intracellular organelles, particularly mitochondria and the SR, within muscle cells. The remodeling of the SR and the formation of whorls may represent a novel mechanism of the unfolded protein response (UPR) in muscle cells.


Subject(s)
Dystrophin-Associated Proteins , Dystrophin , Mitochondria , Sarcoplasmic Reticulum , Animals , Mice , Dystrophin/genetics , Dystrophin/metabolism , Dystrophin/deficiency , Dystrophin-Associated Proteins/genetics , Dystrophin-Associated Proteins/metabolism , Glycoproteins/metabolism , Glycoproteins/genetics , Glycoproteins/deficiency , Mice, Knockout , Mitochondria/metabolism , Mitochondria/ultrastructure , Mitochondria/genetics , Muscle Fibers, Skeletal/metabolism , Muscle Fibers, Skeletal/ultrastructure , Muscle, Skeletal/metabolism , Muscle, Skeletal/ultrastructure , Myofibrils/metabolism , Myofibrils/ultrastructure , Sarcoplasmic Reticulum/metabolism , Sarcoplasmic Reticulum/ultrastructure
2.
Hum Mol Genet ; 31(14): 2370-2385, 2022 07 21.
Article in English | MEDLINE | ID: mdl-35157076

ABSTRACT

α-syntrophin (α-syn) and α-dystrobrevin (α-dbn), two components of the dystrophin-glycoprotein complex, are essential for the maturation and maintenance of the neuromuscular junction (NMJ) and mice deficient in either α-syn or α-dbn exhibit similar synaptic defects. However, the functional link between these two proteins and whether they exert distinct or redundant functions in the postsynaptic organization of the NMJ remain largely unknown. We generated and analyzed the synaptic phenotype of double heterozygote (α-dbn+/-, α-syn+/-), and double homozygote knockout (α-dbn-/-; α-syn-/-) mice and examined the ability of individual molecules to restore their defects in the synaptic phenotype. We showed that in double heterozygote mice, NMJs have normal synaptic phenotypes and no signs of muscular dystrophy. However, in double knockout mice (α-dbn-/-; α-syn-/-), the synaptic phenotype (the density, the turnover and the distribution of AChRs within synaptic branches) is more severely impaired than in single α-dbn-/- or α-syn-/- mutants. Furthermore, double mutant and single α-dbn-/- mutant mice showed more severe exercise-induced fatigue and more significant reductions in grip strength than single α-syn-/- mutant and wild-type. Finally, we showed that the overexpression of the transgene α-syn-GFP in muscles of double mutant restores primarily the abnormal extensions of membrane containing AChRs that extend beyond synaptic gutters and lack synaptic folds, whereas the overexpression of α-dbn essentially restores the abnormal dispersion of patchy AChR aggregates in the crests of synaptic folds. Altogether, these data suggest that α-syn and α-dbn act in parallel pathways and exert distinct functions on the postsynaptic structural organization of NMJs.


Subject(s)
Dystrophin , Receptors, Cholinergic , Animals , Calcium-Binding Proteins , Dystrophin/genetics , Dystrophin/metabolism , Dystrophin-Associated Proteins/genetics , Dystrophin-Associated Proteins/metabolism , Glycoproteins/metabolism , Membrane Proteins , Mice , Muscle Proteins/genetics , Muscle Proteins/metabolism , Muscle, Skeletal/metabolism , Receptors, Cholinergic/genetics , Receptors, Cholinergic/metabolism , Synapses/genetics , Synapses/metabolism
3.
RNA ; 27(10): 1173-1185, 2021 10.
Article in English | MEDLINE | ID: mdl-34215685

ABSTRACT

RNA binding proteins (RBPs) take part in all steps of the RNA life cycle and are often essential for cell viability. Most RBPs have a modular organization and comprise a set of canonical RNA binding domains. However, in recent years a number of high-throughput mRNA interactome studies on yeast, mammalian cell lines, and whole organisms have uncovered a multitude of novel mRNA interacting proteins that lack classical RNA binding domains. Whereas a few have been confirmed to be direct and functionally relevant RNA binders, biochemical and functional validation of RNA binding of most others is lacking. In this study, we used a combination of NMR spectroscopy and biochemical studies to test the RNA binding properties of six putative RBPs. Half of the analyzed proteins showed no interaction, whereas the other half displayed weak chemical shift perturbations upon titration with RNA. One of the candidates we found to interact weakly with RNA in vitro is Drosophila melanogaster end binding protein 1 (EB1), a master regulator of microtubule plus-end dynamics. Further analysis showed that EB1's RNA binding occurs on the same surface as that with which EB1 interacts with microtubules. RNA immunoprecipitation and colocalization experiments suggest that EB1 is a rather nonspecific, opportunistic RNA binder. Our data suggest that care should be taken when embarking on an RNA binding study involving these unconventional, novel RBPs, and we recommend initial and simple in vitro RNA binding experiments.


Subject(s)
Drosophila Proteins/metabolism , Dystrophin-Associated Proteins/metabolism , Microtubule-Associated Proteins/metabolism , RNA-Binding Proteins/metabolism , RNA/metabolism , Thioredoxins/metabolism , Transcription Factors/metabolism , Tripartite Motif Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , Binding Sites , Cloning, Molecular , Drosophila Proteins/chemistry , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Dystrophin-Associated Proteins/chemistry , Dystrophin-Associated Proteins/genetics , Electrophoretic Mobility Shift Assay , Escherichia coli/genetics , Escherichia coli/metabolism , Female , Gene Expression , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Humans , Microtubule-Associated Proteins/chemistry , Microtubule-Associated Proteins/genetics , Microtubules/metabolism , Microtubules/ultrastructure , Models, Molecular , Ovary/cytology , Ovary/metabolism , Poly U/chemistry , Poly U/genetics , Poly U/metabolism , Protein Binding , RNA/chemistry , RNA/genetics , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Thioredoxins/chemistry , Thioredoxins/genetics , Transcription Factors/chemistry , Transcription Factors/genetics , Tripartite Motif Proteins/chemistry , Tripartite Motif Proteins/genetics , Ubiquitin-Protein Ligases/chemistry , Ubiquitin-Protein Ligases/genetics
4.
Acta Neuropathol ; 145(4): 479-496, 2023 04.
Article in English | MEDLINE | ID: mdl-36799992

ABSTRACT

DTNA encodes α-dystrobrevin, a component of the macromolecular dystrophin-glycoprotein complex (DGC) that binds to dystrophin/utrophin and α-syntrophin. Mice lacking α-dystrobrevin have a muscular dystrophy phenotype, but variants in DTNA have not previously been associated with human skeletal muscle disease. We present 12 individuals from four unrelated families with two different monoallelic DTNA variants affecting the coiled-coil domain of α-dystrobrevin. The five affected individuals from family A harbor a c.1585G > A; p.Glu529Lys variant, while the recurrent c.1567_1587del; p.Gln523_Glu529del DTNA variant was identified in the other three families (family B: four affected individuals, family C: one affected individual, and family D: two affected individuals). Myalgia and exercise intolerance, with variable ages of onset, were reported in 10 of 12 affected individuals. Proximal lower limb weakness with onset in the first decade of life was noted in three individuals. Persistent elevations of serum creatine kinase (CK) levels were detected in 11 of 12 affected individuals, 1 of whom had an episode of rhabdomyolysis at 20 years of age. Autism spectrum disorder or learning disabilities were reported in four individuals with the c.1567_1587 deletion. Muscle biopsies in eight affected individuals showed mixed myopathic and dystrophic findings, characterized by fiber size variability, internalized nuclei, and slightly increased extracellular connective tissue and inflammation. Immunofluorescence analysis of biopsies from five affected individuals showed reduced α-dystrobrevin immunoreactivity and variably reduced immunoreactivity of other DGC proteins: dystrophin, α, ß, δ and γ-sarcoglycans, and α and ß-dystroglycans. The DTNA deletion disrupted an interaction between α-dystrobrevin and syntrophin. Specific variants in the coiled-coil domain of DTNA cause skeletal muscle disease with variable penetrance. Affected individuals show a spectrum of clinical manifestations, with severity ranging from hyperCKemia, myalgias, and exercise intolerance to childhood-onset proximal muscle weakness. Our findings expand the molecular etiologies of both muscular dystrophy and paucisymptomatic hyperCKemia, to now include monoallelic DTNA variants as a novel cause of skeletal muscle disease in humans.


Subject(s)
Autism Spectrum Disorder , Muscular Dystrophies , Neuropeptides , Mice , Humans , Animals , Child , Dystrophin/genetics , Dystrophin/metabolism , Autism Spectrum Disorder/metabolism , Muscular Dystrophies/metabolism , Dystroglycans/metabolism , Alternative Splicing , Muscle, Skeletal/pathology , Neuropeptides/genetics , Neuropeptides/metabolism , Dystrophin-Associated Proteins/genetics , Dystrophin-Associated Proteins/metabolism
5.
Mol Psychiatry ; 27(4): 1963-1969, 2022 04.
Article in English | MEDLINE | ID: mdl-35246634

ABSTRACT

Alzheimer's disease (AD) is a genetically complex disease for which nearly 40 loci have now been identified via genome-wide association studies (GWAS). We attempted to identify groups of rare variants (alternate allele frequency <0.01) associated with AD in a region-based, whole-genome sequencing (WGS) association study (rvGWAS) of two independent AD family datasets (NIMH/NIA; 2247 individuals; 605 families). Employing a sliding window approach across the genome, we identified several regions that achieved association p values <10-6, using the burden test or the SKAT statistic. The genomic region around the dystobrevin beta (DTNB) gene was identified with the burden and SKAT test and replicated in case/control samples from the ADSP study reaching genome-wide significance after meta-analysis (pmeta = 4.74 × 10-8). SKAT analysis also revealed region-based association around the Discs large homolog 2 (DLG2) gene and replicated in case/control samples from the ADSP study (pmeta = 1 × 10-6). In conclusion, in a region-based rvGWAS of AD we identified two novel AD genes, DLG2 and DTNB, based on association with rare variants.


Subject(s)
Alzheimer Disease , Dystrophin-Associated Proteins/genetics , Neuropeptides/genetics , Alzheimer Disease/genetics , Dithionitrobenzoic Acid , Genetic Predisposition to Disease/genetics , Genome-Wide Association Study , Genomics , Guanylate Kinases/genetics , Humans , Polymorphism, Single Nucleotide/genetics , Tumor Suppressor Proteins/genetics , Whole Genome Sequencing
6.
Cell Mol Life Sci ; 79(2): 109, 2022 Jan 30.
Article in English | MEDLINE | ID: mdl-35098363

ABSTRACT

Duchenne muscular dystrophy (DMD), the most severe form of dystrophinopathies, is a fatal X-linked recessive neuromuscular disorder characterized by progressive muscle degeneration and various extents of intellectual disabilities. Physiological and pathological roles of the responsible gene, dystrophin, in the brain remain elusive due to the presence of multiple dystrophin products, mainly full-length dystrophin, Dp427, and the short product, Dp71. In this study, we generated a Dp71-specific hemagglutinin (HA) peptide tag-insertion mice to enable specific detection of intrinsic Dp71 expression by anti-HA-tag antibodies. Immunohistochemical detections in the transgenic mice demonstrated Dp71 expression not only at the blood-brain barrier, where astrocytic endfeet surround the microvessels, but also at the inhibitory postsynapse of hippocampal dentate granule neurons. Interestingly, hippocampal cornu ammonis (CA)1 pyramidal neurons were negative for Dp71, although Dp427 detected by anti-dystrophin antibody was clearly present at the inhibitory postsynapse, suggesting cell-type dependent dystrophin expressions. Precise examination using the primary hippocampal culture validated exclusive localization of Dp71 at the inhibitory postsynaptic compartment but not at the excitatory synapse in neurons. We further performed interactome analysis and found that Dp71 formed distinct molecular complexes, i.e. synapse-associated Dp71 interacted with dystroglycan (Dg) and dystrobrevinß (Dtnb), whereas glia-associated Dp71 did with Dg and dystrobrevinα (Dtna). Thus, our data indicate that Dp71 and its binding partners are relevant to the inhibitory postsynaptic function of hippocampal granule neurons and the novel Dp71-transgenic mouse provides a valuable tool to understand precise physiological expressions and functions of Dp71 and its interaction proteins in vivo and in vitro.


Subject(s)
Dystroglycans/metabolism , Dystrophin-Associated Proteins/metabolism , Dystrophin/metabolism , Neuroglia/metabolism , Neuropeptides/metabolism , Synapses/metabolism , Animals , Blood-Brain Barrier/metabolism , Cells, Cultured , Dystroglycans/genetics , Dystrophin/genetics , Dystrophin-Associated Proteins/genetics , HEK293 Cells , Hippocampus/cytology , Hippocampus/metabolism , Humans , Mice, Transgenic , Microscopy, Confocal , Neurons/metabolism , Neuropeptides/genetics , Protein Binding
7.
J Biol Chem ; 296: 100516, 2021.
Article in English | MEDLINE | ID: mdl-33676892

ABSTRACT

Cells can switch between Rac1 (lamellipodia-based) and RhoA (blebbing-based) migration modes, but the molecular mechanisms regulating this shift are not fully understood. Diacylglycerol kinase ζ (DGKζ), which phosphorylates diacylglycerol to yield phosphatidic acid, forms independent complexes with Rac1 and RhoA, selectively dissociating each from their common inhibitor RhoGDI. DGKζ catalytic activity is required for Rac1 dissociation but is dispensable for RhoA dissociation; instead, DGKζ stimulates RhoA release via a kinase-independent scaffolding mechanism. The molecular determinants that mediate the selective targeting of DGKζ to Rac1 or RhoA signaling complexes are unknown. Here, we show that protein kinase Cα (PKCα)-mediated phosphorylation of the DGKζ MARCKS domain increased DGKζ association with RhoA and decreased its interaction with Rac1. The same modification also enhanced DGKζ interaction with the scaffold protein syntrophin. Expression of a phosphomimetic DGKζ mutant stimulated membrane blebbing in mouse embryonic fibroblasts and C2C12 myoblasts, which was augmented by inhibition of endogenous Rac1. DGKζ expression in differentiated C2 myotubes, which have low endogenous Rac1 levels, also induced substantial membrane blebbing via the RhoA-ROCK pathway. These events were independent of DGKζ catalytic activity, but dependent upon a functional C-terminal PDZ-binding motif. Rescue of RhoA activity in DGKζ-null cells also required the PDZ-binding motif, suggesting that syntrophin interaction is necessary for optimal RhoA activation. Collectively, our results define a switch-like mechanism whereby DGKζ phosphorylation by PKCα plays a role in the interconversion between Rac1 and RhoA signaling pathways that underlie different cellular migration modes.


Subject(s)
Cell Movement , Diacylglycerol Kinase/physiology , Dystrophin-Associated Proteins/metabolism , Myristoylated Alanine-Rich C Kinase Substrate/metabolism , Neuropeptides/metabolism , Protein Kinase C-alpha/pharmacology , rac1 GTP-Binding Protein/metabolism , rhoA GTP-Binding Protein/metabolism , Animals , Diglycerides/metabolism , Dystrophin-Associated Proteins/genetics , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/metabolism , Mice , Mice, Knockout , Myristoylated Alanine-Rich C Kinase Substrate/genetics , Neuropeptides/genetics , Protein Domains , rac1 GTP-Binding Protein/genetics , rhoA GTP-Binding Protein/genetics
8.
BMC Cardiovasc Disord ; 22(1): 37, 2022 02 11.
Article in English | MEDLINE | ID: mdl-35148685

ABSTRACT

Atrial fibrillation (AF) is a morbid and heritable irregular cardiac rhythm that affects about 2%-3% of the population. Patients with early-onset AF have a strong genetic association with the disease; nonetheless, the exact underlying mechanisms need clarification. We herein present our evaluation of a 2-generation Iranian pedigree with early-onset AF. Whole-exome sequencing was applied to elucidate the genetic predisposition. Direct DNA sequencing was utilized to confirm and screen the variants in the proband and his available family members. The pathogenicity of the identified nucleotide variations was scrutinized via either segregation analysis in the family or in silico predictive software. The comprehensive variant analysis revealed a missense variant (c.G681C, p.E227D, rs1477078144) in the human α-dystrobrevin gene (DTNA), which is rare in genetic databases. Most in silico analyses have predicted this variant as a disease-causing variant, and the variant is co-segregated with the disease phenotype in the family. Previous studies have demonstrated the association between the DTNA gene and left ventricular noncompaction cardiomyopathy. Taken together, we provide the first evidence of an association between a nucleotide variation in the DTNA gene and early-onset AF in an Iranian family. However, the genetic testing of AF in the Iranian population is still limited. This finding not only further confirms the significant role of genetics in the incidence of early-onset AF but also expands the spectrum of the gene variations that lead to AF. Additionally, it may have further implications for the treatment and prevention of AF.


Subject(s)
Atrial Fibrillation/genetics , DNA Mutational Analysis , Dystrophin-Associated Proteins/genetics , Exome Sequencing , Heart Rate/genetics , Mutation, Missense , Neuropeptides/genetics , Adolescent , Atrial Fibrillation/diagnosis , Atrial Fibrillation/physiopathology , Electrocardiography , Female , Genetic Predisposition to Disease , Heredity , Humans , Iran , Male , Middle Aged , Pedigree , Phenotype , Predictive Value of Tests , Young Adult
9.
J Biol Chem ; 295(31): 10677-10688, 2020 07 31.
Article in English | MEDLINE | ID: mdl-32532815

ABSTRACT

The maintenance of a high density of the acetylcholine receptor (AChR) is the hallmark of the neuromuscular junction. Muscle-specific anchoring protein (αkap) encoded within the calcium/calmodulin-dependent protein kinase IIα (CAMK2A) gene is essential for the maintenance of AChR clusters both in vivo and in cultured muscle cells. The underlying mechanism by which αkap is maintained and regulated remains unknown. Here, using human cell lines, fluorescence microscopy, and pulldown and immunoblotting assays, we show that α-dystrobrevin (α-dbn), an intracellular component of the dystrophin glycoprotein complex, directly and robustly promotes the stability of αkap in a concentration-dependent manner. Mechanistically, we found that the phosphorylatable tyrosine residues of α-dbn are essential for the stability of α-dbn itself and its interaction with αkap, with substitution of three tyrosine residues in the α-dbn C terminus with phenylalanine compromising the αkap-α-dbn interaction and significantly reducing both αkap and α-dbn accumulation. Moreover, the αkap-α-dbn interaction was critical for αkap accumulation and stability. We also found that the absence of either αkap or α-dbn markedly reduces AChRα accumulation and that overexpression of α-dbn or αkap in cultured muscle cells promotes the formation of large agrin-induced AChR clusters. Collectively, these results indicate that the stability of αkap and α-dbn complex plays an important role in the maintenance of high-level expression of AChRs.


Subject(s)
A Kinase Anchor Proteins/metabolism , Dystrophin-Associated Proteins/metabolism , Multiprotein Complexes/metabolism , Neuropeptides/metabolism , Receptors, Cholinergic/biosynthesis , A Kinase Anchor Proteins/genetics , Animals , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Dystrophin-Associated Proteins/genetics , HEK293 Cells , HeLa Cells , Humans , Mice , Multiprotein Complexes/genetics , Neuropeptides/genetics , Protein Domains , Protein Stability , Receptors, Cholinergic/genetics
10.
Hum Mol Genet ; 28(3): 386-395, 2019 02 01.
Article in English | MEDLINE | ID: mdl-30256963

ABSTRACT

Syntrophins are a family of modular adaptor proteins that are part of the dystrophin protein complex, where they recruit and anchor a variety of signaling proteins. Previously we generated mice lacking α- and/or ß2-syntrophin but showed that in the absence of one isoform, other syntrophin isoforms can partially compensate. Therefore, in the current study, we generated mice that lacked α, ß1 and ß2-syntrophins [triple syntrophin knockout (tKO) mice] and assessed skeletal and cardiac muscle function. The tKO mice showed a profound reduction in voluntary wheel running activity at both 6 and 12 months of age. Function of the tibialis anterior was assessed in situ and we found that the specific force of tKO muscle was decreased by 20-25% compared with wild-type mice. This decrease was accompanied by a shift in fiber-type composition from fast 2B to more oxidative fast 2A fibers. Using echocardiography to measure cardiac function, it was revealed that tKO hearts had left ventricular cardiac dysfunction and were hypertrophic, with a thicker left ventricular posterior wall. Interestingly, we also found that membrane-localized dystrophin expression was lower in both skeletal and cardiac muscles of tKO mice. Since dystrophin mRNA levels were not different in tKO, this finding suggests that syntrophins may regulate dystrophin trafficking to, or stabilization at, the sarcolemma. These results show that the loss of all three major muscle syntrophins has a profound effect on exercise performance, and skeletal and cardiac muscle dysfunction contributes to this deficiency.


Subject(s)
Calcium-Binding Proteins/physiology , Dystrophin-Associated Proteins/physiology , Membrane Proteins/physiology , Muscle Proteins/physiology , Animals , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Dystrophin/genetics , Dystrophin/physiology , Dystrophin-Associated Proteins/genetics , Heart/physiology , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Motor Activity , Muscle Proteins/genetics , Muscle, Skeletal/metabolism , Muscle, Skeletal/physiology , Myocardium/metabolism , Protein Isoforms/genetics , Protein Isoforms/physiology
11.
Int J Legal Med ; 135(4): 1341-1349, 2021 Jul.
Article in English | MEDLINE | ID: mdl-33895855

ABSTRACT

Sudden unexplained death (SUD) takes up a considerable part in overall sudden death cases, especially in adolescents and young adults. During the past decade, many channelopathy- and cardiomyopathy-associated single nucleotide variants (SNVs) have been identified in SUD studies by means of postmortem molecular autopsy, yet the number of cases that remain inconclusive is still high. Recent studies had suggested that structural variants (SVs) might play an important role in SUD, but there is no consensus on the impact of SVs on inherited cardiac diseases. In this study, we searched for potentially pathogenic SVs in 244 genes associated with cardiac diseases. Whole-exome sequencing and appropriate data analysis were performed in 45 SUD cases. Re-analysis of the exome data according to the current ACMG guidelines identified 14 pathogenic or likely pathogenic variants in 10 (22.2%) out of the 45 SUD cases, whereof 2 (4.4%) individuals had variants with likely functional effects in the channelopathy-associated genes SCN5A and TRDN and 1 (2.2%) individual in the cardiomyopathy-associated gene DTNA. In addition, 18 structural variants (SVs) were identified in 15 out of the 45 individuals. Two SVs with likely functional impairment were found in the coding regions of PDSS2 and TRPM4 in 2 SUD cases (4.4%). Both were identified as heterozygous deletions, which were confirmed by multiplex ligation-dependent probe amplification. In conclusion, our findings support that SVs could contribute to the pathology of the sudden death event in some of the cases and therefore should be investigated on a routine basis in suspected SUD cases.


Subject(s)
Death, Sudden/pathology , Genomic Structural Variation/genetics , Heart Diseases/genetics , Polymorphism, Single Nucleotide , Adolescent , Adult , Alkyl and Aryl Transferases , Carrier Proteins/genetics , Child , Child, Preschool , Cohort Studies , Dystrophin-Associated Proteins/genetics , Female , Humans , Infant , Male , Middle Aged , Muscle Proteins/genetics , NAV1.5 Voltage-Gated Sodium Channel/genetics , Neuropeptides/genetics , Open Reading Frames , Switzerland/epidemiology , TRPM Cation Channels , Exome Sequencing
12.
Int J Mol Sci ; 22(23)2021 Dec 02.
Article in English | MEDLINE | ID: mdl-34884867

ABSTRACT

Duchenne muscular dystrophy (DMD) is a lethal X-linked recessive disorder caused by mutations in the DMD gene and the subsequent lack of dystrophin protein. Recently, phosphorodiamidate morpholino oligomer (PMO)-antisense oligonucleotides (ASOs) targeting exon 51 or 53 to reestablish the DMD reading frame have received regulatory approval as commercially available drugs. However, their applicability and efficacy remain limited to particular patients. Large animal models and exon skipping evaluation are essential to facilitate ASO development together with a deeper understanding of dystrophinopathies. Using recombinant adeno-associated virus-mediated gene targeting and somatic cell nuclear transfer, we generated a Yucatan miniature pig model of DMD with an exon 52 deletion mutation equivalent to one of the most common mutations seen in patients. Exon 52-deleted mRNA expression and dystrophin deficiency were confirmed in the skeletal and cardiac muscles of DMD pigs. Accordingly, dystrophin-associated proteins failed to be recruited to the sarcolemma. The DMD pigs manifested early disease onset with severe bodywide skeletal muscle degeneration and with poor growth accompanied by a physical abnormality, but with no obvious cardiac phenotype. We also demonstrated that in primary DMD pig skeletal muscle cells, the genetically engineered exon-52 deleted pig DMD gene enables the evaluation of exon 51 or 53 skipping with PMO and its advanced technology, peptide-conjugated PMO. The results show that the DMD pigs developed here can be an appropriate large animal model for evaluating in vivo exon skipping efficacy.


Subject(s)
Dystrophin/genetics , Exons , Muscle, Skeletal/pathology , Muscular Dystrophy, Duchenne/genetics , Animals , Animals, Genetically Modified , Dependovirus/genetics , Disease Models, Animal , Dystrophin-Associated Proteins/genetics , Dystrophin-Associated Proteins/metabolism , Female , Gene Deletion , Male , Muscle Fibers, Skeletal/pathology , Nuclear Transfer Techniques , Oligonucleotides, Antisense/genetics , Sarcolemma/metabolism , Swine , Swine, Miniature
13.
Esophagus ; 18(2): 315-325, 2021 04.
Article in English | MEDLINE | ID: mdl-32737801

ABSTRACT

BACKGROUND: Esophageal cancer (EC) ranks the eighth in morbidity and the sixth in mortality around the whole world, which is an aggressive malignancy. To authenticate potential therapeutic targets for EC is therefore imperative. Although miR-301b might display changed expression in esophageal adenocarcinoma by utilizing Taqman miRNA profiling analysis, much less is known about the impact of miR-301b in EC. METHODS AND RESULTS: By analyzing the data of 187 cancer tissues and 13 normal samples from TCGA database, we discovered that miR-301b was highly expressed in EC tissues. Then, RT-qPCR determined that miR-301b was up-regulated in EC cell lines (ECA109, JAR, TE-1 and OE33). Besides, miR-301b expression level was higher in ESCC cell line-TE-1 cells and lower in ESCC cell line-ECA109 cells compared to other EC cell lines. Hence, ECA109 cell line was used to up-regulate miR-301b expression while TE-1 cell line was applied to down-regulate miR-301b expression in the subsequent experiments. Additionally, OE33, as an ECA cell line, was applied to upregulate miR-301b expression to reflect the influence of miR-301b overexpression on EC progression. More interestingly, miR-301b appeared to act as a promoting effect on the proliferation of EC cells, which was tested by CCK8. Dystrobrevin alpha (DTNA) was a targeting gene of miR-301b, which was predicted by the websites of miRanda, miRWalk and TargetScan. Additionally, DTNA was low expressed in EC tissues and was an independent predictor of EC. Meanwhile, the low expression of DTNA was related to worse overall survival in EC patients. The Pearson correlation coefficient analyzed that DTNA expression was negatively correlated with miR-301b. Furthermore, RT-qPCR and western blotting assays ulteriorly indicated that DTNA was negatively modulated by miR-301b. The facilitating impact of miR-301b re-expression on ECA109 and OE33 cell growth, invasion and migration was receded by DTNA over-expression, whilst the repressive effect of miR-301b ablation on TE-1 cell growth, invasion and migration was inversed by DTNA silencing. Overexpression of miR-301b accelerated EC cell growth, migration and invasion through targeting DTNA. CONCLUSIONS: Above all, we concluded that miR-301b was concerned with the progression of EC via regulating DTNA, suggesting that miR-301b and its target gene, DTNA, might serve as predictive biomarkers for EC therapy.


Subject(s)
Dystrophin-Associated Proteins , Esophageal Neoplasms , MicroRNAs , Neuropeptides , Cell Line, Tumor , Cell Movement , Cell Proliferation/genetics , Dystrophin-Associated Proteins/genetics , Dystrophin-Associated Proteins/metabolism , Esophageal Neoplasms/genetics , Esophageal Neoplasms/metabolism , Esophageal Neoplasms/pathology , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , Neoplasm Invasiveness , Neuropeptides/genetics , Neuropeptides/metabolism
14.
Hum Mol Genet ; 27(17): 2978-2985, 2018 09 01.
Article in English | MEDLINE | ID: mdl-29790927

ABSTRACT

Mutation of the gene encoding dystrophin leads to Duchenne and Becker muscular dystrophy (DMD and BMD). Currently, dystrophin is thought to function primarily as a structural protein, connecting the muscle cell actin cytoskeleton to the extra-cellular matrix. In addition to this structural role, dystrophin also plays an important role as a scaffold that organizes an array of signaling proteins including sodium, potassium, and calcium channels, kinases, and nitric oxide synthase (nNOS). Many of these signaling proteins are linked to dystrophin via syntrophin, an adapter protein that is known to bind directly to two sites in the carboxyl terminal region of dystrophin. A search of the dystrophin sequence revealed three additional potential syntrophin binding sites (SBSs) within the spectrin-like repeat (SLR) region of dystrophin. Binding assays revealed that the site at SLR 17 bound specifically to the α isoform of syntrophin while the site at SLR 22 bound specifically to the ß-syntrophins. The SLR 17 α-SBS contained the core sequence known to be required for nNOS-dystrophin interaction. In vitro and in vivo assays indicate that α-syntrophin facilitates the nNOS-dystrophin interaction at this site rather than nNOS binding directly to dystrophin as previously reported. The identification of multiple SBSs within the SLR region of dystrophin demonstrates that this region functions as a signaling scaffold. The signaling role of the SLR region of dystrophin will need to be considered for effective gene replacement or exon skipping based DMD/BMD therapies.


Subject(s)
Dystrophin-Associated Proteins/metabolism , Dystrophin/metabolism , Nitric Oxide Synthase Type I/physiology , Repetitive Sequences, Amino Acid , Spectrin/metabolism , Amino Acid Sequence , Animals , Binding Sites , Dystrophin-Associated Proteins/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Sequence Homology , Spectrin/chemistry
15.
Am J Pathol ; 189(4): 813-825, 2019 04.
Article in English | MEDLINE | ID: mdl-30653956

ABSTRACT

Syntrophins are a family of proteins forming membrane-anchored scaffolds and serving as adaptors for various transmembrane and intracellular signaling molecules. To understand the physiological roles of ß1 syntrophin, one of the least characterized members, we generated mouse models to eliminate ß1 syntrophin specifically in the endocrine or exocrine pancreas. ß1 syntrophin is dispensable for the morphology and function of insulin-producing ß cells. However, mice with ß1 syntrophin deletion in exocrine acinar cells exhibit increased severity of cerulein-induced acute pancreatitis. Reduced expression of cystic fibrosis transmembrane conductance regulator and dilation of acinar lumen are potential predisposition factors. During the disease progression, a relative lack of autophagy is associated with deficiencies in both actin assembly and endoplasmic reticulum nucleation. Our findings reveal, for the first time, that ß1 syntrophin is a critical regulator of actin cytoskeleton and autophagy in pancreatic acinar cells and is potently protective against cerulein-induced acute pancreatitis.


Subject(s)
Autophagy , Ceruletide/toxicity , Dystrophin-Associated Proteins/physiology , Pancreatitis/prevention & control , Protective Agents , Acinar Cells/drug effects , Acinar Cells/metabolism , Animals , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Pancreas/drug effects , Pancreas/metabolism , Pancreatitis/chemically induced , Pancreatitis/metabolism , Pancreatitis/pathology
16.
J Cell Physiol ; 234(7): 10726-10740, 2019 07.
Article in English | MEDLINE | ID: mdl-30565678

ABSTRACT

Ischemia-reperfusion (I/R) injury often leads to myocardial apoptosis and necrosis. Studies have demonstrated the role microRNAs (miRs) played in myocardial I/R injury. Thus, we established a myocardial I/R injury model and a thoracic epidural anesthesia (TEA) model in mice to explore whether microRNA-374 (miR-374) affects myocardial I/R injury. We collected myocardial tissues to evaluate whether TEA exerts a protection effect on myocardial tissues. In addition, the levels of miR-374, dystrobrevin alpha (DTNA), and the statue of the Notch1 axis were detected. Subsequently, cardiomyocytes extracted from TEA mice were treated to regulate their levels of miR-374 and DTNA. After that, cell viability, cell cycle distribution, and apoptosis of cardiomyocytes were assessed. This was followed by the detection of the myocardial infarction area. The mice models of myocardial I/R injury were associated with poorly expressed miR-374 and highly expressed DTNA. TEA was found to protect myocardial tissues against myocardial I/R injury by elevating miR-374 and reducing DTNA. Dual-luciferase reporter assay validated that DTNA was the target gene of miR-374. Cardiomyocytes with overexpressed miR-374 were shown to have downregulated DTNA levels and blocked Notch1 axis. Overexpressed miR-374 was also found to promote the viability and inhibit the apoptosis of cardiomyocytes, as well as to increase the number of cells arrested in the S phase. In accordance with this, the myocardial infarction area was decreased with the upregulated miR-347 and downregulated DTNA. Collectively, these results demonstrated that, by inhibiting the activity of DTNA-mediated Notch1 axis, miR-374 could protect against myocardial I/R injury in mice after TEA.


Subject(s)
Dystrophin-Associated Proteins/genetics , MicroRNAs/genetics , Myocardial Ischemia/genetics , Neuropeptides/genetics , Receptor, Notch1/genetics , Reperfusion Injury/genetics , Anesthesia, Epidural/adverse effects , Animals , Apoptosis/genetics , Cell Survival/genetics , Disease Models, Animal , Humans , Mice , Myocardial Ischemia/pathology , Myocytes, Cardiac/metabolism , Reperfusion Injury/pathology , Signal Transduction/genetics
17.
Glia ; 67(6): 1138-1149, 2019 06.
Article in English | MEDLINE | ID: mdl-30803043

ABSTRACT

Proper function of the retina depends heavily on a specialized form of retinal glia called Müller cells. These cells carry out important homeostatic functions that are contingent on their polarized nature. Specifically, the Müller cell endfeet that contact retinal microvessels and the corpus vitreum show a tenfold higher concentration of the inwardly rectifying potassium channel Kir 4.1 than other Müller cell plasma membrane domains. This highly selective enrichment of Kir 4.1 allows K+ to be siphoned through endfoot membranes in a special form of spatial buffering. Here, we show that Kir 4.1 is enriched in endfoot membranes through an interaction with ß1-syntrophin. Targeted disruption of this syntrophin caused a loss of Kir 4.1 from Müller cell endfeet without affecting the total level of Kir 4.1 expression in the retina. Targeted disruption of α1-syntrophin had no effect on Kir 4.1 localization. Our findings show that the Kir 4.1 aggregation that forms the basis for K+ siphoning depends on a specific syntrophin isoform that colocalizes with Kir 4.1 in Müller endfoot membranes.


Subject(s)
Dystrophin-Associated Proteins/deficiency , Ependymoglial Cells/metabolism , Gene Deletion , Potassium Channels, Inwardly Rectifying/deficiency , Retina/metabolism , Animals , Dystrophin-Associated Proteins/genetics , Ependymoglial Cells/pathology , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Potassium Channels, Inwardly Rectifying/genetics , Protein Aggregates/physiology , Retina/pathology
18.
EMBO J ; 34(15): 2059-77, 2015 Aug 04.
Article in English | MEDLINE | ID: mdl-26108535

ABSTRACT

Recycling synaptic vesicles (SVs) transit through early endosomal sorting stations, which raises a fundamental question: are SVs sorted toward endolysosomal pathways? Here, we used snapin mutants as tools to assess how endolysosomal sorting and trafficking impact presynaptic activity in wild-type and snapin(-/-) neurons. Snapin acts as a dynein adaptor that mediates the retrograde transport of late endosomes (LEs) and interacts with dysbindin, a subunit of the endosomal sorting complex BLOC-1. Expressing dynein-binding defective snapin mutants induced SV accumulation at presynaptic terminals, mimicking the snapin(-/-) phenotype. Conversely, over-expressing snapin reduced SV pool size by enhancing SV trafficking to the endolysosomal pathway. Using a SV-targeted Ca(2+) sensor, we demonstrate that snapin-dysbindin interaction regulates SV positional priming through BLOC-1/AP-3-dependent sorting. Our study reveals a bipartite regulation of presynaptic activity by endolysosomal trafficking and sorting: LE transport regulates SV pool size, and BLOC-1/AP-3-dependent sorting fine-tunes the Ca(2+) sensitivity of SV release. Therefore, our study provides new mechanistic insights into the maintenance and regulation of SV pool size and synchronized SV fusion through snapin-mediated LE trafficking and endosomal sorting.


Subject(s)
Lysosomes/metabolism , Models, Neurological , Neurons/physiology , Synaptic Transmission/physiology , Synaptic Vesicles/metabolism , Vesicular Transport Proteins/metabolism , Animals , Biological Transport/physiology , Blotting, Western , Calcium/metabolism , Cell Fractionation , Cells, Cultured , Circular Dichroism , Dysbindin , Dystrophin-Associated Proteins , Immunohistochemistry , Mice , Microscopy, Electron , Time-Lapse Imaging , Vesicular Transport Proteins/genetics
19.
J Cell Sci ; 130(10): 1752-1759, 2017 05 15.
Article in English | MEDLINE | ID: mdl-28364093

ABSTRACT

A bimolecular fluorescence complementation (BiFC) approach was used to study the molecular interactions between different components of the postsynaptic protein complex at the neuromuscular junction of living mice. We show that rapsyn forms complex with both α-dystrobrevin and α-syntrophin at the crests of junctional folds. The linkage of rapsyn to α-syntrophin and/or α-dystrobrevin is mediated by utrophin, a protein localized at acetylcholine receptor (AChR)-rich domains. In mice deficient in α-syntrophin, in which utrophin is no longer present at the synapse, rapsyn interaction with α-dystrobrevin was completely abolished. This interaction was completely restored when either utrophin or α-syntrophin was introduced into muscles deficient in α-syntrophin. However, in neuromuscular junctions deficient in α-dystrobrevin, in which utrophin is retained, complex formation between rapsyn and α-syntrophin was unaffected. Using fluorescence recovery after photobleaching, we found that α-syntrophin turnover is 5-7 times faster than that of AChRs, and loss of α-dystrobrevin has no effect on rapsyn and α-syntrophin half-life, whereas the half-life of AChR was significantly altered. Altogether, these results provide new insights into the spatial distribution of dystrophin glycoprotein components and their dynamics in living mice.


Subject(s)
Dystrophin/chemistry , Dystrophin/metabolism , Molecular Dynamics Simulation , Neuromuscular Junction/metabolism , Animals , Calcium-Binding Proteins/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Dystrophin-Associated Proteins/metabolism , Female , Fluorescence , Membrane Proteins/metabolism , Mice , Muscle Proteins/metabolism , Protein Binding , Synapses/metabolism , Utrophin/metabolism
20.
Cell Physiol Biochem ; 52(5): 1151-1165, 2019.
Article in English | MEDLINE | ID: mdl-30990585

ABSTRACT

BACKGROUND/AIMS: Adipocyte hypertrophy in obesity is associated with inflammation and adipose tissue fibrosis which both contribute to metabolic diseases. Mechanisms regulating lipid droplet expansion are poorly understood. Knock down of the scaffold protein beta 2 syntrophin (SNTB2) increases lipid droplet size of 3T3-L1 adipocytes and the physiological relevance of SNTB2 in adipose tissue morphology and metabolic health was analyzed herein. METHODS: Wild type and SNTB2-/- mice were challenged with 24 weeks high fat diet. Adipose tissue morphology and expression of various genes / proteins including collagens and caveolin-1 was examined. Glucose, insulin, fasting and fed free fatty acids were measured in serum. SNTB2 expression was determined in adipose tissues of patients. RESULTS: Upon high fat diet SNTB2-/- mice displayed reduced adiposity and adipocyte hypertrophy. Expression of various proteins was normal in the different white fat depots of SNTB2-/- mice while caveolin-1 protein and collagen mRNA levels were diminished. Null mice had reduced systemic glucose while fasting and postprandial insulin and insulin response were normal. Fatty acid clearance in the fed state and after insulin injection was enhanced. SNTB2 and caveolin-1 were increased in fat of ob/ob mice. However, no correlation between body mass index and SNTB2 protein in adipose tissues of seven patients was found. In subcutaneous but not in visceral fat the ratio of SNTB2 to alpha syntrophin protein, which affects lipid droplet size in the opposite manner, was associated with BMI. In subcutaneous fat of extremely obese patients SNTB2 mRNA levels were not correlated with weight loss after bariatric surgery. CONCLUSION: Current study shows that high SNTB2 in obese adipose tissues restricts adipocyte growth and thereby may contribute to metabolic diseases.


Subject(s)
Adipocytes/metabolism , Dietary Fats/pharmacology , Dystrophin-Associated Proteins , Lipid Metabolism , Obesity/metabolism , Postprandial Period , Adipocytes/pathology , Adult , Aged , Animals , Caveolin 1/genetics , Caveolin 1/metabolism , Dystrophin-Associated Proteins/genetics , Dystrophin-Associated Proteins/metabolism , Female , Humans , Male , Mice , Mice, Knockout , Middle Aged , Obesity/genetics , Obesity/pathology
SELECTION OF CITATIONS
SEARCH DETAIL