Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 2.105
1.
J Physiol ; 602(9): 1893-1910, 2024 May.
Article En | MEDLINE | ID: mdl-38615232

Dysferlin is a 237 kDa membrane-associated protein characterised by multiple C2 domains with a diverse role in skeletal and cardiac muscle physiology. Mutations in DYSF are known to cause various types of human muscular dystrophies, known collectively as dysferlinopathies, with some patients developing cardiomyopathy. A myriad of in vitro membrane repair studies suggest that dysferlin plays an integral role in the membrane repair complex in skeletal muscle. In comparison, less is known about dysferlin in the heart, but mounting evidence suggests that dysferlin's role is similar in both muscle types. Recent findings have shown that dysferlin regulates Ca2+ handling in striated muscle via multiple mechanisms and that this becomes more important in conditions of stress. Maintenance of the transverse (t)-tubule network and the tight coordination of excitation-contraction coupling are essential for muscle contractility. Dysferlin regulates the maintenance and repair of t-tubules, and it is suspected that dysferlin regulates t-tubules and sarcolemmal repair through a similar mechanism. This review focuses on the emerging complexity of dysferlin's activity in striated muscle. Such insights will progress our understanding of the proteins and pathways that regulate basic heart and skeletal muscle function and help guide research into striated muscle pathology, especially that which arises due to dysferlin dysfunction.


Calcium , Dysferlin , Humans , Calcium/metabolism , Dysferlin/metabolism , Membrane Proteins/metabolism , Membrane Proteins/genetics , Membrane Proteins/physiology , Muscle Proteins/metabolism , Muscle Proteins/genetics , Muscle Proteins/physiology , Muscle, Skeletal/metabolism , Muscle, Skeletal/physiology , Muscle, Striated/metabolism , Muscle, Striated/physiology
2.
Adv Protein Chem Struct Biol ; 137: 205-223, 2023.
Article En | MEDLINE | ID: mdl-37709377

We know that numerous proteins expressed in the heart are influenced by environmental signals (such as light and diet), which cause either an increase or decrease in their expression. Cardiovascular health is sensitive to diet composition (macronutrient content), as well as the percentage of energy, frequency and regularity of meal intake during the 24-hour cycle, and the fasting period. Furthermore, light is an important synchronizer of the circadian clock and, in turn, of several physiological processes, among them cardiovascular physiology. In this chapter, we address the effects of these environmental cues and the known mechanisms that lead to this variation in protein expression in the heart, as well as cardiac function.


Circadian Clocks , Heart , Muscle Proteins , Fasting , Muscle Proteins/physiology , Humans , Light
3.
J Phys Chem Lett ; 13(40): 9473-9479, 2022 Oct 13.
Article En | MEDLINE | ID: mdl-36198174

The nanomechanical response of a folded single protein, the natural nanomachine responsible for myriad biological processes, provides insight into its function. The conformational flexibility of a folded state, characterized by its viscoelasticity, allows proteins to adopt different shapes to perform their function. Despite efforts, its direct measurement has not been possible so far. We present a direct and simultaneous measurement of the stiffness and internal friction of the folded domains of the protein titin using a special interferometer based atomic force microscope. We analyzed the data by carefully separating different contributions affecting the response of the experimental probe to obtain the folded state's viscoelasticity. Above ∼95 pN of force, the individual immunoglobulins of titin transition from an elastic solid-like native state to a soft viscoelastic intermediate.


Immunoglobulins , Muscle Proteins , Connectin , Elasticity , Friction , Muscle Proteins/physiology
4.
J Exp Biol ; 225(16)2022 08 15.
Article En | MEDLINE | ID: mdl-35875854

Evidence suggests that the giant muscle protein titin functions as a tunable spring in active muscle. However, the mechanisms for increasing titin stiffness with activation are not well understood. Previous studies have suggested that during muscle activation, titin binds to actin, which engages the PEVK region of titin, thereby increasing titin stiffness. In this study, we investigated the role of PEVK titin in active muscle stiffness during rapid unloading. We measured elastic recoil of active and passive soleus muscles from TtnΔ112-158 mice characterized by a 75% deletion of PEVK titin and increased passive stiffness. We hypothesized that activated TtnΔ112-158 muscles are stiffer than wild-type muscles as a result of the increased stiffness of PEVK titin. Using a servomotor force lever, we compared the stress-strain relationships of elastic elements in active and passive muscles during rapid unloading and quantified the change in stiffness upon activation. The results show that the elastic modulus of TtnΔ112-158 muscles increased with activation. However, elastic elements developed force at 7% longer lengths and exhibited 50% lower active stiffness in TtnΔ112-158 soleus muscles than in wild-type muscles. Thus, despite having a shorter, stiffer PEVK segment, during rapid unloading, TtnΔ112-158 soleus muscles exhibited reduced active stiffness compared with wild-type soleus muscles. These results are consistent with the idea that PEVK titin contributes to active muscle stiffness; however, the reduction in active stiffness of TtnΔ112-158 muscles suggests that other mechanisms compensate for the increased PEVK stiffness.


Muscle Proteins , Muscle, Skeletal , Protein Kinases/metabolism , Animals , Connectin/genetics , Elastic Modulus , Mice , Muscle Proteins/physiology , Muscle, Skeletal/physiology , Protein Kinases/genetics
5.
J Gene Med ; 24(1): e3390, 2022 01.
Article En | MEDLINE | ID: mdl-34558151

BACKGROUND: Hypertrophic cardiomyopathy (HCM) is a hereditary disease manifested by a thickened ventricular wall. Cysteine and glycine-rich protein 3 (CSRP3), the gene encoding muscle LIM protein, is important for initiating hypertrophic gene expression. The mutation of CSRP3 causes dilated cardiomyopathy or HCM. METHODS: In the present study, we enrolled a Chinese family with HCM across three generations. Whole-exome sequencing (WES) was performed in the proband to detect the candidate genes of the family. Sanger sequencing was performed for mutational analysis and confirmation of cosegregation. RESULTS: Through histopathological and imaging examinations, an obvious left ventricular hypertrophy was found in the proband. After WES data filtering, bioinformatic prediction and co-segregation analysis, a nonsense mutation (NM_003476.5:c.364C>T; NP_003467.1:p.Arg122*) of CSRP3 was identified in this family. This variant was predicted to be disease-causing and resulted in a truncated protein. CONCLUSIONS: This is the first HCM family case of CSRP3 (p.Arg122*) variation in Asia. The finding here not only contributes to the genetic diagnosis and counseling of the family, but also provides a new case with detailed phenotypes that may be caused by the CSRP3 variant.


Cardiomyopathy, Hypertrophic/genetics , LIM Domain Proteins/genetics , LIM Domain Proteins/physiology , Muscle Proteins/genetics , Muscle Proteins/physiology , Adult , Biopsy , China/epidemiology , Computational Biology , Family Health , Female , Genetic Predisposition to Disease , Genotype , Heterozygote , Humans , Male , Middle Aged , Mutation , Myocardium/pathology , Pedigree , Phenotype , Polymorphism, Single Nucleotide , Exome Sequencing
6.
J Endocrinol ; 251(2): 125-135, 2021 09 20.
Article En | MEDLINE | ID: mdl-34382577

Apoptosis repressor with caspase recruitment domain (ARC) is an endogenous inhibitor of cell death signaling that is expressed in insulin-producing ß cells. ARC has been shown to reduce ß-cell death in response to diabetogenic stimuli in vitro, but its role in maintaining glucose homeostasis in vivo has not been fully established. Here we examined whether loss of ARC in FVB background mice exacerbates high fat diet (HFD)-induced hyperglycemia in vivo over 24 weeks. Prior to commencing 24-week HFD, ARC-/- mice had lower body weight than wild type (WT) mice. This body weight difference was maintained until the end of the study and was associated with decreased epididymal and inguinal adipose tissue mass in ARC-/- mice. Non-fasting plasma glucose was not different between ARC-/- and WT mice prior to HFD feeding, and ARC-/- mice displayed a greater increase in plasma glucose over the first 4 weeks of HFD. Plasma glucose remained elevated in ARC-/- mice after 16 weeks of HFD feeding, at which time it had returned to baseline in WT mice. Following 24 weeks of HFD, non-fasting plasma glucose in ARC-/- mice returned to baseline and was not different from WT mice. At this final time point, no differences were observed between genotypes in plasma glucose or insulin under fasted conditions or following intravenous glucose administration. However, HFD-fed ARC-/- mice exhibited significantly decreased ß-cell area compared to WT mice. Thus, ARC deficiency delays, but does not prevent, metabolic adaptation to HFD feeding in mice, worsening transient HFD-induced hyperglycemia.


Apoptosis Regulatory Proteins/physiology , Diet, High-Fat/adverse effects , Hyperglycemia/etiology , Insulin-Secreting Cells/physiology , Muscle Proteins/physiology , Animals , Blood Glucose , Insulin Secretion , Mice
7.
PLoS Genet ; 17(7): e1009690, 2021 07.
Article En | MEDLINE | ID: mdl-34319989

Recent studies have focused on capillary pruning in various organs and species. However, the way in which large-diameter vessels are pruned remains unclear. Here we show that pruning of the zebrafish caudal vein (CV) from ventral capillaries of the CV plexus in different transgenic embryos is driven by endothelial cell (EC) rearrangement, which involves EC nucleus migration, junction remodeling, and actin cytoskeleton remodeling. Further observation reveals a growing difference in blood flow velocity between the two vessels in CV pruning in zebrafish embryos. With this model, we identify the critical role of Kruppel-like factor 6a (klf6a) in CV pruning. Disruption of klf6a functioning impairs CV pruning in zebrafish. klf6a is required for EC nucleus migration, junction remodeling, and actin cytoskeleton dynamics in zebrafish embryos. Moreover, actin-related protein transgelin 2 (tagln2) is a direct downstream target of klf6a in CV pruning in zebrafish embryos. Together these results demonstrate that the klf6a-tagln2 axis regulates CV pruning by promoting EC rearrangement.


Blood Circulation/physiology , Microfilament Proteins/physiology , Muscle Proteins/physiology , Nerve Tissue Proteins/physiology , Zebrafish Proteins/physiology , Actin Cytoskeleton/metabolism , Actin Cytoskeleton/physiology , Animals , Animals, Genetically Modified , Capillaries/metabolism , Cell Movement , Endothelial Cells/metabolism , Endothelial Cells/physiology , Kruppel-Like Transcription Factors/genetics , Microfilament Proteins/genetics , Microfilament Proteins/metabolism , Morphogenesis , Muscle Proteins/genetics , Muscle Proteins/metabolism , Nerve Tissue Proteins/metabolism , Zebrafish/metabolism , Zebrafish/physiology , Zebrafish Proteins/metabolism
8.
Int J Mol Sci ; 22(14)2021 Jul 08.
Article En | MEDLINE | ID: mdl-34298968

Mitochondrial dysfunction is considered the major contributor to skeletal muscle wasting in different conditions. Genetically determined neuromuscular disorders occur as a result of mutations in the structural proteins of striated muscle cells and therefore are often combined with cardiac phenotype, which most often manifests as a cardiomyopathy. The specific roles played by mitochondria and mitochondrial energetic metabolism in skeletal muscle under muscle-wasting conditions in cardiomyopathies have not yet been investigated in detail, and this aspect of genetic muscle diseases remains poorly characterized. This review will highlight dysregulation of mitochondrial representation and bioenergetics in specific skeletal muscle disorders caused by mutations that disrupt the structural and functional integrity of muscle cells.


Cardiomyopathies/genetics , Heart/physiopathology , Mitochondria, Muscle/metabolism , Muscle, Skeletal/metabolism , Neuromuscular Diseases/genetics , Animals , Cardiomyopathies/metabolism , Cardiomyopathies/pathology , Disease Models, Animal , Energy Metabolism , Humans , Mice , Mitochondria, Heart/metabolism , Muscle Proteins/deficiency , Muscle Proteins/genetics , Muscle Proteins/physiology , Muscle, Skeletal/ultrastructure , Muscular Atrophy/metabolism , Muscular Dystrophies/genetics , Muscular Dystrophies/metabolism , Muscular Dystrophies/pathology , Muscular Dystrophy, Animal/genetics , Muscular Dystrophy, Animal/metabolism , Muscular Dystrophy, Animal/pathology , Neuromuscular Diseases/metabolism , Neuromuscular Diseases/pathology , Phenotype
9.
Physiol Rep ; 9(13): e14927, 2021 07.
Article En | MEDLINE | ID: mdl-34197700

Cachexia, a condition prevalent in many chronically ill patients, is characterized by weight loss, fatigue, and decreases in muscle mass and function. Cachexia is associated with tumor burden and disease-related malnutrition, but other studies implicate chemotherapy as being causative. We investigated the effects of a chemotherapy drug cocktail on myofibrillar protein abundance and synthesis, anabolic signaling mechanisms, and substrate availability. On day 4 of differentiation, L6 myotubes were treated with vehicle (1.4 µl/ml DMSO) or a chemotherapy drug cocktail (a mixture of cisplatin [20 µg/ml], leucovorin [10 µg/ml], and 5-fluorouracil [5-FLU; 50 µg/ml]) for 24-72 h. Compared to myotubes treated with vehicle, those treated with the drug cocktail showed 50%-80% reductions in the abundance of myofibrillar proteins, including myosin heavy chain-1, troponin, and tropomyosin (p < 0.05). Cells treated with only a mixture of cisplatin and 5-FLU had identical reductions in myofibrillar protein abundance. Myotubes treated with the drug cocktail also showed >50% reductions in the phosphorylation of AKTSer473 and of mTORC1 substrates ribosomal protein S6Ser235/236 , its kinase S6K1Thr389 and eukaryotic translation initiation factor 4E-binding protein 1 (all p < 0.05). Drug treatment impaired peptide chain initiation in myofibrillar protein fractions and insulin-stimulated glucose uptake (p = 0.06) but increased the expression of autophagy markers beclin-1 and microtubule-associated proteins 1A/1B light chain 3B (p < 0.05), and of apoptotic marker, cleaved caspase 3 (p < 0.05). Drug treatment reduced the expression of mitochondrial markers cytochrome oxidase and succinate dehydrogenase (p < 0.05). The observed profound negative effects of this chemotherapy drug cocktail on myotubes underlie a need for approaches that can reduce the negative effects of these drugs on muscle metabolism.


Muscle Fibers, Skeletal/drug effects , Muscle Proteins/drug effects , Animals , Blotting, Western , Cachexia/chemically induced , Cells, Cultured , Cisplatin/administration & dosage , Cisplatin/pharmacology , Drug Therapy, Combination , Fluorouracil/administration & dosage , Fluorouracil/pharmacology , Leucovorin/administration & dosage , Leucovorin/pharmacology , Muscle Fibers, Skeletal/chemistry , Muscle Fibers, Skeletal/ultrastructure , Muscle Proteins/analysis , Muscle Proteins/physiology , Myosin Heavy Chains/analysis , Rats , Tropomyosin/analysis , Troponin/analysis
10.
Cells ; 10(5)2021 05 06.
Article En | MEDLINE | ID: mdl-34066362

Selenoprotein N (SEPN1) is a type II glycoprotein of the endoplasmic reticulum (ER) that senses calcium levels to tune the activity of the sarcoplasmic reticulum calcium pump (SERCA pump) through a redox-mediated mechanism, modulating ER calcium homeostasis. In SEPN1-depleted muscles, altered ER calcium homeostasis triggers ER stress, which induces CHOP-mediated malfunction, altering excitation-contraction coupling. SEPN1 is localized in a region of the ER where the latter is in close contact with mitochondria, i.e., the mitochondria-associated membranes (MAM), which are important for calcium mobilization from the ER to mitochondria. Accordingly, SEPN1-depleted models have impairment of both ER and mitochondria calcium regulation and ATP production. SEPN1-related myopathy (SEPN1-RM) is an inherited congenital muscle disease due to SEPN1 loss of function, whose main histopathological features are minicores, i.e., areas of mitochondria depletion and sarcomere disorganization in muscle fibers. SEPN1-RM presents with weakness involving predominantly axial and diaphragmatic muscles. Since there is currently no disease-modifying drug to treat this myopathy, analysis of SEPN1 function in parallel with that of the muscle phenotype in SEPN1 loss of function models should help in understanding the pathogenic basis of the disease and possibly point to novel drugs for therapy. The present essay recapitulates the novel biological findings on SEPN1 and how these reconcile with the muscle and bioenergetics phenotype of SEPN1-related myopathy.


Calcium/metabolism , Endoplasmic Reticulum/metabolism , Muscle Proteins/physiology , Muscle, Skeletal/metabolism , Muscular Diseases/metabolism , Selenoproteins/physiology , Animals , Endoplasmic Reticulum Stress , Humans , Mitochondria/metabolism , Muscle, Skeletal/cytology , Muscle, Skeletal/pathology , Oxidation-Reduction
11.
Cell Death Dis ; 12(6): 604, 2021 06 11.
Article En | MEDLINE | ID: mdl-34117213

Kindlin-2 is known to play important roles in the development of mesoderm-derived tissues including myocardium, smooth muscle, cartilage and blood vessels. However, nothing is known for the role of Kindlin-2 in mesoderm-derived reproductive organs. Here, we report that loss of Kindlin-2 in Sertoli cells caused severe testis hypoplasia, abnormal germ cell development and complete infertility in male mice. Functionally, loss of Kindlin-2 inhibits proliferation, increases apoptosis, impairs phagocytosis in Sertoli cells and destroyed the integration of blood-testis barrier structure in testes. Mechanistically, Kindlin-2 interacts with LATS1 and YAP, the key components of Hippo pathway. Kindlin-2 impedes LATS1 interaction with YAP, and depletion of Kindlin-2 enhances LATS1 interaction with YAP, increases YAP phosphorylation and decreases its nuclear translocation. For clinical relevance, lower Kindlin-2 expression and decreased nucleus localization of YAP was found in SCOS patients. Collectively, we demonstrated that Kindlin-2 in Sertoli cells is essential for sperm development and male reproduction.


Cytoskeletal Proteins/physiology , Fertility/genetics , Muscle Proteins/physiology , Testis/growth & development , Animals , Cells, Cultured , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism , HEK293 Cells , Humans , Infertility, Male/genetics , Infertility, Male/metabolism , Infertility, Male/pathology , Male , Mice , Mice, Transgenic , Muscle Proteins/genetics , Muscle Proteins/metabolism , Sertoli Cells/metabolism , Sertoli Cells/physiology , Testis/metabolism
12.
Commun Biol ; 4(1): 611, 2021 05 21.
Article En | MEDLINE | ID: mdl-34021256

Accumulation of vascular smooth muscle cells (VSMCs) is a hallmark of multiple vascular pathologies, including following neointimal formation after injury and atherosclerosis. However, human VSMCs in advanced atherosclerotic lesions show reduced cell proliferation, extensive and persistent DNA damage, and features of premature cell senescence. Here, we report that stress-induced premature senescence (SIPS) and stable expression of a telomeric repeat-binding factor 2 protein mutant (TRF2T188A) induce senescence of human VSMCs, associated with persistent telomeric DNA damage. VSMC senescence is associated with formation of micronuclei, activation of cGAS-STING cytoplasmic sensing, and induction of multiple pro-inflammatory cytokines. VSMC-specific TRF2T188A expression in a multicolor clonal VSMC-tracking mouse model shows no change in VSMC clonal patches after injury, but an increase in neointima formation, outward remodeling, senescence and immune/inflammatory cell infiltration or retention. We suggest that persistent telomere damage in VSMCs inducing cell senescence has a major role in driving persistent inflammation in vascular disease.


Atherosclerosis/pathology , Cellular Senescence , Inflammation/pathology , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/pathology , Neointima/pathology , Telomere/pathology , Animals , Atherosclerosis/etiology , Atherosclerosis/metabolism , Cell Proliferation , Cells, Cultured , DNA Damage , Disease Models, Animal , Humans , Inflammation/etiology , Inflammation/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microfilament Proteins/physiology , Muscle Proteins/physiology , Muscle, Smooth, Vascular/immunology , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/immunology , Myocytes, Smooth Muscle/metabolism , Neointima/etiology , Neointima/metabolism , Telomere/genetics , Telomeric Repeat Binding Protein 2/metabolism
13.
DNA Cell Biol ; 40(6): 798-810, 2021 Jun.
Article En | MEDLINE | ID: mdl-34030484

Recent studies have revealed the significant role of TEA domain family member 4 (TEAD4) in the development and progression of cancer. However, the potential role of TEAD4 in the progression of bladder cancer (BC) remains to be explored. The aim of this study was to determine whether TEAD4 could serve as a pan-cancer predictor of the prognosis for BC. Based on data mined from public databases, expression levels and clinical value of TEAD4 were identified in BC and human pan-cancers. Reverse transcription-quantitative polymerase chain reaction (RT-qPCR) analysis was performed to detect the TEAD4 expression levels in BC cell lines. Gene Set Enrichment Analysis (GSEA) was carried out for functional analysis in BC, and the relationship between infiltrating immune cells and TEAD4 expression was evaluated by the CIBERSORT algorithm in BC and pan-cancer data. TEAD4 was overexpressed and associated with poor prognosis in BC and several types of cancers. GSEA and CIBERSORT algorithm suggested that various pathways including immune-related pathways were enriched in TEAD4 high expression group and several immunocytes infiltrated were correlated with the expression of TEAD4. This study revealed TEAD4 is an immune regulating-related predictor of prognosis for BC and has generalization value in pan-cancer.


DNA-Binding Proteins/physiology , Muscle Proteins/physiology , Transcription Factors/physiology , Urinary Bladder Neoplasms/metabolism , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor/physiology , Disease Progression , Female , Gene Expression Regulation, Neoplastic , Humans , Male , Middle Aged , TEA Domain Transcription Factors
14.
Pharmacol Res ; 168: 105592, 2021 06.
Article En | MEDLINE | ID: mdl-33813027

Abnormal glycolytic metabolism contributes to angiogenic sprouting involved in atherogenesis. We investigated the potential anti-angiogenic properties of specific 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase-3 (PFKFB3) inhibitors in endothelial cells (ECs). ECs were treated with PFKFB3 inhibitors (named PA-1 and PA-2) and their effects on metabolic and functional characteristics of ECs were investigated. The anti-glycolytic compound 3-(pyridinyl)- 1-(4-pyridinyl)- 2-propen-1-one (3PO) was used as reference compound. PFKFB3 expression and activity (IC50 about 3-21 nM) was inhibited upon treatment with both compounds. Glucose uptake and lactate export were measured using commercial assays and showed a partial reduction up to 40%. PFKFB3 inhibition increased intracellular lactate accumulation, and reduced expression of monocarboxylate transporters-1 (MCT1) and MCT4. Furthermore, endothelial cell migration and proliferation assays demonstrated significant reduction upon treatment with both compounds. Matrix- metalloproteinase (MMP) activity, measured by gelatin zymography, and expression was significantly reduced (up to 25%). In addition, PA compounds downregulated the expression of VCAM-1, VE-cadherin, VEGFa, VEGFR2, TGF-ß, and IL-1ß, in inflamed ECs. Finally, PA-1 and PA-2 treatment impaired the formation of angiogenic sprouts measured by both morphogenesis and spheroid-based angiogenesis assays. Our data demonstrate that the anti-glycolytic PA compounds may affect several steps involved in angiogenesis. Targeting the key glycolytic enzyme PFKFB3 might represent an attractive therapeutic strategy to improve the efficacy of cancer treatments, or to be applied in other pathologies where angiogenesis is a detrimental factor.


Angiogenesis Inhibitors/pharmacology , Phosphofructokinase-2/antagonists & inhibitors , Cells, Cultured , Humans , Lactic Acid/metabolism , Monocarboxylic Acid Transporters/physiology , Muscle Proteins/physiology , NAD/metabolism , Neovascularization, Pathologic/drug therapy , Symporters/physiology
15.
Dev Cell ; 56(6): 747-760.e6, 2021 03 22.
Article En | MEDLINE | ID: mdl-33667344

Loss of insulin-secreting pancreatic ß cells through apoptosis contributes to the progression of type 2 diabetes, but underlying mechanisms remain elusive. Here, we identify a pathway in which the cell death inhibitor ARC paradoxically becomes a killer during diabetes. While cytoplasmic ARC maintains ß cell viability and pancreatic architecture, a pool of ARC relocates to the nucleus to induce ß cell apoptosis in humans with diabetes and several pathophysiologically distinct mouse models. ß cell death results through the coordinate downregulation of serpins (serine protease inhibitors) not previously known to be synthesized and secreted by ß cells. Loss of the serpin α1-antitrypsin from the extracellular space unleashes elastase, triggering the disruption of ß cell anchorage and subsequent cell death. Administration of α1-antitrypsin to mice with diabetes prevents ß cell death and metabolic abnormalities. These data uncover a pathway for ß cell loss in type 2 diabetes and identify an FDA-approved drug that may impede progression of this syndrome.


Apoptosis , Cell Nucleus/metabolism , Cytoskeletal Proteins/metabolism , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Type 2/pathology , Insulin-Secreting Cells/pathology , Nerve Tissue Proteins/metabolism , alpha 1-Antitrypsin/chemistry , Animals , Apoptosis Regulatory Proteins/physiology , Cytoplasm/metabolism , Cytoskeletal Proteins/genetics , Diabetes Mellitus, Experimental/etiology , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 2/etiology , Diabetes Mellitus, Type 2/metabolism , Female , Humans , Insulin-Secreting Cells/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle Proteins/physiology , Nerve Tissue Proteins/genetics , alpha 1-Antitrypsin/genetics , alpha 1-Antitrypsin/metabolism
16.
Hum Mol Genet ; 29(24): 3882-3891, 2021 02 25.
Article En | MEDLINE | ID: mdl-33355670

Striated preferentially expressed gene (SPEG), a member of the myosin light chain kinase family, is localized at the level of triad surrounding myofibrils in skeletal muscles. In humans, SPEG mutations are associated with centronuclear myopathy and cardiomyopathy. Using a striated muscle-specific Speg-knockout (KO) mouse model, we have previously shown that SPEG is critical for triad maintenance and calcium handling. Here, we further examined the molecular function of SPEG and characterized the effects of SPEG deficiency on triad and focal adhesion proteins. We used yeast two-hybrid assay, and identified desmin, an intermediate filament protein, to interact with SPEG and confirmed this interaction by co-immunoprecipitation. Using domain-mapping assay, we defined that Ig-like and fibronectin III domains of SPEG interact with rod domain of desmin. In skeletal muscles, SPEG depletion leads to desmin aggregates in vivo and a shift in desmin equilibrium from soluble to insoluble fraction. We also profiled the expression and localization of triadic proteins in Speg-KO mice using western blot and immunofluorescence. The amount of RyR1 and triadin were markedly reduced, whereas DHPRα1, SERCA1 and triadin were abnormally accumulated in discrete areas of Speg-KO myofibers. In addition, Speg-KO muscles exhibited internalized vinculin and ß1 integrin, both of which are critical components of the focal adhesion complex. Further, ß1 integrin was abnormally accumulated in early endosomes of Speg-KO myofibers. These results demonstrate that SPEG-deficient skeletal muscles exhibit several pathological features similar to those seen in MTM1 deficiency. Defects of shared cellular pathways may underlie these structural and functional abnormalities in both types of diseases.


Cell Adhesion Molecules/metabolism , Desmin/metabolism , Focal Adhesions/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Muscle Proteins/metabolism , Muscle Proteins/physiology , Muscle, Skeletal/pathology , Myopathies, Structural, Congenital/pathology , Myosin-Light-Chain Kinase/physiology , Animals , Calcium/metabolism , Cell Adhesion Molecules/genetics , Desmin/genetics , Intracellular Signaling Peptides and Proteins/genetics , Male , Mice , Mice, Knockout , Mice, Transgenic , Muscle Proteins/genetics , Muscle, Skeletal/metabolism , Mutation , Myopathies, Structural, Congenital/etiology , Myopathies, Structural, Congenital/metabolism
17.
Proteins ; 89(4): 427-435, 2021 04.
Article En | MEDLINE | ID: mdl-33244801

Sarcolipin (SLN) is an important transmembrane (TM) protein encoded by long noncoding RNA. SLN is expressed in the sarcoplasmic reticulum and regulates cardiac and skeletal muscle contractions. SLN forms a pentameric hydrophobic ligand-gated ion channel. The protonation of Glu7 (protonated SLN, pSLN) and mutation of Thr18 to Ala18 (T18A) have been reported to exert a significant influence on the permeability of the channel. In this study, the altered permeability of both the pSLN and T18A pentameric channels was simulated. Combined with molecular dynamics simulation, the free-energy landscape for single ions, computational electrophysiology, diffusion coefficient, and pore geometrical characteristic analyses were performed to further understand the properties of amino acid modifications in the SLN pentameric channel. The results suggest that both the pSLN and T18A pentameric channels form stable hydrophobic ligand-gated channels. The TM voltage has a positive effect on the permeability of water molecules and ions. By using pSLN and T18A, our study provides helpful information on the pore-forming mechanism of SLN and furthers our understanding of the regulatory mechanisms underlying the permeation of ions and water molecules in the pentameric SLN channel.


Muscle Proteins , Proteolipids , Amino Acids/chemistry , Amino Acids/metabolism , Cell Membrane Permeability , Humans , Hydrophobic and Hydrophilic Interactions , Ions/metabolism , Molecular Dynamics Simulation , Muscle Proteins/chemistry , Muscle Proteins/metabolism , Muscle Proteins/physiology , Proteolipids/chemistry , Proteolipids/metabolism , Proteolipids/physiology , Water/metabolism
18.
FEBS Lett ; 595(1): 85-98, 2021 01.
Article En | MEDLINE | ID: mdl-33053208

The four-and-a-half LIM domain protein 1 (FHL1) plays a key role in multiple cancers. Here, we characterized its role in glioblastoma (GBM), the most common and incurable form of brain cancer. Overexpression of FHL1 promotes growth, migration, and invasion of GBM cells in vivo and in vitro. In contrast, FHL1 silencing by RNAi exhibits the opposite effects. FHL1 interacts with the transcription factor SP1 to upregulate epidermal growth factor receptor (EGFR) expression and activate the downstream signaling cascades, including Src, Akt, Erk1/2, and Stat3, leading to GBM malignancy. FHL1 is highly expressed and positively correlated with EGFR levels in human GBM, particularly those of the classical subtype. Our results suggest that the FHL1-SP1-EGFR axis plays a tumor-promoting role, and highlight the translational potential of inhibiting FHL1 for GBM treatment.


Brain Neoplasms/pathology , Glioblastoma/pathology , Intracellular Signaling Peptides and Proteins/physiology , LIM Domain Proteins/physiology , Muscle Proteins/physiology , Cell Proliferation , Disease Progression , ErbB Receptors/physiology , Gene Silencing , Humans , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , LIM Domain Proteins/genetics , LIM Domain Proteins/metabolism , Muscle Proteins/genetics , Muscle Proteins/metabolism , Neoplasm Invasiveness , Neoplasm Metastasis , Protein Kinases/metabolism , STAT2 Transcription Factor/metabolism , Sp1 Transcription Factor/metabolism
19.
J Chin Med Assoc ; 84(1): 79-86, 2021 Jan 01.
Article En | MEDLINE | ID: mdl-32796321

BACKGROUND: Premature infants often require oxygen (O2) therapy for respiratory distress syndrome; however, excessive use of O2 can cause clinical conditions such as bronchopulmonary dysplasia. Although many treatment methods are currently available, they are not effective in preventing bronchopulmonary dysplasia. Herein, we explored the role of tripartite motif protein 72 (TRIM72), a factor involved in repairing alveolar epithelial wounds, in regulating alveolar cells upon hyperoxia exposure. METHODS: In this in vivo study, we used Sprague-Dawley rat pups that were reared in room air or 85% O2 for 2 weeks after birth. The lungs were excised for histological analyses, and TRIM72 expression was assessed on postnatal days 7 and 14. For in vitro experiments, RLE-6TN cells (i.e., rat alveolar type II epithelial cells) and A549 cells (i.e., human lung carcinoma epithelial cells) were exposed to 85% O2 for 5 days. The cells were then analyzed for cell viability, and TRIM72 expression was determined. RESULTS: Exposure to hyperoxia reduced body and lung weight, increased mean linear intercept values, and upregulated TRIM72 expression. In vitro study results revealed increased or decreased lung cell viability upon hyperoxia exposure depending on the suppression or overexpression of TRIM72, respectively. CONCLUSION: Hyperoxia upregulates TRIM72 expression in neonatal rat lung tissue; moreover, it initiates TRIM72-dependent alveolar epithelial cell death, leading to hyperoxia-induced lung injury.


Hyperoxia/pathology , Lung/pathology , Muscle Proteins/physiology , Vesicular Transport Proteins/physiology , Animals , Cell Survival , Cells, Cultured , Epithelial Cells/pathology , Female , Muscle Proteins/analysis , Rats , Rats, Sprague-Dawley , Vesicular Transport Proteins/analysis
20.
Elife ; 92020 12 24.
Article En | MEDLINE | ID: mdl-33357376

The giant muscle protein titin is a major contributor to passive force; however, its role in active force generation is unresolved. Here, we use a novel titin-cleavage (TC) mouse model that allows specific and rapid cutting of elastic titin to quantify how titin-based forces define myocyte ultrastructure and mechanics. We show that under mechanical strain, as TC doubles from heterozygous to homozygous TC muscles, Z-disks become increasingly out of register while passive and active forces are reduced. Interactions of elastic titin with sarcomeric actin filaments are revealed. Strikingly, when titin-cleaved muscles contract, myosin-containing A-bands become split and adjacent myosin filaments move in opposite directions while also shedding myosins. This establishes intact titin filaments as critical force-transmission networks, buffering the forces observed by myosin filaments during contraction. To perform this function, elastic titin must change stiffness or extensible length, unveiling its fundamental role as an activation-dependent spring in contracting muscle.


Muscle Contraction , Muscle Proteins/physiology , Muscle, Skeletal/physiology , Protein Kinases/physiology , Animals , Female , Male , Mice , Muscle Proteins/metabolism , Muscle, Skeletal/metabolism , Protein Kinases/metabolism , Tensile Strength
...