Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 698
Filter
1.
Physiol Rep ; 9(17): e15003, 2021 09.
Article in English | MEDLINE | ID: mdl-34435451

ABSTRACT

Myosin, the most abundant myofibrillar protein in skeletal muscle, functions as a motor protein in muscle contraction. Myosin polymerizes into the thick filaments in the sarcomere where approximately 50% of embryonic myosin (Myh3) are replaced within 3 h (Ojima K, Ichimura E, Yasukawa Y, Wakamatsu J, Nishimura T, Am J Physiol Cell Physiol 309: C669-C679, 2015). The sarcomere structure including the thick filament is maintained by a balance between protein biosynthesis and degradation. However, the involvement of a protein degradation system in the myosin replacement process remains unclear. Here, we show that the muscle-specific ubiquitin ligase Ozz regulates replacement rate of Myh3. To examine the direct effect of Ozz on myosin replacement, eGFP-Myh3 replacement rate was measured in myotubes overexpressing Ozz by fluorescence recovery after photobleaching. Ozz overexpression significantly decreased the replacement rate of eGFP-Myh3 in the myofibrils, whereas it had no effect on other myosin isoforms. It is likely that ectopic Ozz promoted myosin degradation through increment of ubiquitinated myosin, and decreased myosin supply for replacement, thereby reducing myosin replacement rate. Intriguingly, treatment with a proteasome inhibitor MG132 also decreased myosin replacement rate, although MG132 enhanced the accumulation of ubiquitinated myosin in the cytosol where replaceable myosin is pooled, suggesting that ubiquitinated myosin is not replaced by myosin in the myofibril. Collectively, our findings showed that Myh3 replacement rate was reduced in the presence of overexpressed Ozz probably through enhanced ubiquitination and degradation of Myh3 by Ozz.


Subject(s)
Embryo, Nonmammalian/enzymology , Muscle Proteins/biosynthesis , Myofibrils/enzymology , Myosins/biosynthesis , Ubiquitin-Protein Ligase Complexes/biosynthesis , Animals , Cells, Cultured , Chick Embryo , Cytosol/enzymology , Myosins/antagonists & inhibitors
2.
Sci Rep ; 11(1): 9794, 2021 05 07.
Article in English | MEDLINE | ID: mdl-33963248

ABSTRACT

The profound energy-expending nature of brown adipose tissue (BAT) thermogenesis makes it an attractive target tissue to combat obesity-associated metabolic disorders. While cold exposure is the strongest inducer of BAT activity, the temporal mechanisms tuning BAT adaptation during this activation process are incompletely understood. Here we show that the scaffold protein Afadin is dynamically regulated by cold in BAT, and participates in cold acclimation. Cold exposure acutely increases Afadin protein levels and its phosphorylation in BAT. Knockdown of Afadin in brown pre-adipocytes does not alter adipogenesis but restricts ß3-adrenegic induction of thermogenic genes expression and HSL phosphorylation in mature brown adipocytes. Consistent with a defect in thermogenesis, an impaired cold tolerance was observed in fat-specific Afadin knockout mice. However, while Afadin depletion led to reduced Ucp1 mRNA induction by cold, stimulation of Ucp1 protein was conserved. Transcriptomic analysis revealed that fat-specific ablation of Afadin led to decreased functional enrichment of gene sets controlling essential metabolic functions at thermoneutrality in BAT, whereas it led to an altered reprogramming in response to cold, with enhanced enrichment of different pathways related to metabolism and remodeling. Collectively, we demonstrate a role for Afadin in supporting the adrenergic response in brown adipocytes and BAT function.


Subject(s)
Adipocytes, Brown/metabolism , Adipose Tissue, Brown/metabolism , Cold Temperature , Gene Expression Regulation , Kinesins/biosynthesis , Myosins/biosynthesis , Thermogenesis , Animals , Kinesins/genetics , Mice , Mice, Knockout , Myosins/genetics
3.
Sci Rep ; 10(1): 12371, 2020 07 23.
Article in English | MEDLINE | ID: mdl-32704163

ABSTRACT

MYO18B has been proposed to contribute to the progression of hepatocellular carcinoma (HCC). However, the signals that govern MYO18B transcription are not known. Here we show that, a network of C19MC miRNA-520G, IFN-γ, CEBPB and p53 transcriptional-defects promote MYO18B mRNA expression in HCCs. IFN-γ by itself suppresses MYO18B transcription, but promotes it when miRNA-520G is stably overexpressed. Similarly, CEBPB-liver-enriched activator protein (LAP) isoform overexpression suppresses MYO18B transcription but promotes transcription when the cells are treated with IFN-γ. Furthermore, miR-520G together with mutant-p53 promotes MYO18B transcription. Conversely, bFGF suppresses MYO18B mRNA irrespective of CEBPB, miR-520G overexpression or IFN-γ treatment. Finally high MYO18B expression reflects poor prognosis while high MYL5 or MYO1B expression reflects better survival of HCC patients. Thus, we identified a network of positive and negative regulators of MYO18B mRNA expression which reflects the survival of HCC patients.


Subject(s)
CCAAT-Enhancer-Binding Protein-beta/biosynthesis , Carcinoma, Hepatocellular/metabolism , Fibroblast Growth Factor 2/biosynthesis , Gene Expression Regulation, Neoplastic , Interferon-gamma/biosynthesis , Liver Neoplasms/metabolism , MicroRNAs/biosynthesis , Myosins/biosynthesis , RNA, Messenger/biosynthesis , RNA, Neoplasm/biosynthesis , Tumor Suppressor Protein p53/biosynthesis , Tumor Suppressor Proteins/biosynthesis , CCAAT-Enhancer-Binding Protein-beta/genetics , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Cell Line , Female , Fibroblast Growth Factor 2/genetics , Humans , Interferon-gamma/genetics , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Male , MicroRNAs/genetics , Myosins/genetics , RNA, Messenger/genetics , RNA, Neoplasm/genetics , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Proteins/genetics
4.
Urol Int ; 104(11-12): 891-901, 2020.
Article in English | MEDLINE | ID: mdl-32674099

ABSTRACT

AIM: To describe architecture and expression of myosin isoforms of the human cremaster muscle (CM) and to individuate changes in clinically differentiated abnormalities of testicular descent: cryptorchidism or undescended testis (UDT) and retractile testis (RT). BACKGROUND: The CM is a nonsomitic striated muscle differentiating from mesenchyme of the gubernaculum testis. Morphofunctional and molecular peculiarities linked to its unique embryological origin are not yet completely defined. Its role in abnormalities of testicular descent is being investigated. SUBJECTS AND METHODS: Biopsy samples were obtained from corrective surgery in cases of cryptorchidism, retractile testis, inguinal hernia, or hydrocele. Muscle specimens were processed for morphology, histochemistry, and immunohistology. RESULTS AND CONCLUSIONS: The CM differs from the skeletal muscles both for morphological and molecular characteristics. The presence of fascicles with different characterization and its myosinic pattern suggested that the CM could be included in the specialized muscle groups, such as the extrinsic ocular muscles (EOMs) and laryngeal and masticatory muscles. The embryological origin from the nonsomitic mesoderm is, also for the CM, the basis of distinct molecular pathways. In UDT, the histological alterations of CM are suggestive of denervation; the genitofemoral nerve and its molecular messengers directed to this muscle are likely defective. Compared with the other samples, RT has a distinct myosinic pattern; therefore, it has been considered a well-defined entity with respect to the other testicular descent abnormalities.


Subject(s)
Abdominal Muscles/metabolism , Cryptorchidism/metabolism , Myosins/biosynthesis , Testicular Diseases/metabolism , Abdominal Muscles/anatomy & histology , Child , Child, Preschool , Humans , Infant , Male , Prospective Studies , Protein Isoforms/biosynthesis
5.
J Appl Physiol (1985) ; 123(3): 513-525, 2017 Sep 01.
Article in English | MEDLINE | ID: mdl-28522766

ABSTRACT

Terrestrial opossums use their semiprehensile tail for grasping nesting materials as opposed to arboreal maneuvering. We relate the development of this adaptive behavior with ontogenetic changes in myosin heavy chain (MHC) isoform expression from 21 days to adulthood. Monodelphis domestica is expected to demonstrate a progressive ability to flex the distal tail up to age 7 mo, when it should exhibit routine nest construction. We hypothesize that juvenile stages (3-7 mo) will be characterized by retention of the neonatal isoform (MHC-Neo), along with predominant expression of fast MHC-2X and -2B, which will transition into greater MHC-1ß and -2A isoform content as development progresses. This hypothesis was tested using Q-PCR to quantify and compare gene expression of each isoform with its protein content determined by gel electrophoresis and densitometry. These data were correlated with nesting activity in an age-matched sample of each age group studied. Shifts in regulation of MHC gene transcripts matched well with isoform expression. Notably, mRNA for MHC-Neo and -2B decrease, resulting in little-to-no isoform translation after age 7 mo, whereas mRNA for MHC-1ß and -2A increase, and this corresponds with subtle increases in content for these isoforms into late adulthood. Despite the tail remaining intrinsically fast-contracting, a critical growth period for isoform transition is observed between 7 and 13 mo, correlating primarily with use of the tail during nesting activities. Functional transitions in MHC isoforms and fiber type properties may be associated with muscle "tuning" repetitive nest remodeling tasks requiring sustained contractions of the caudal flexors.NEW & NOTEWORTHY Little is understood about skeletal muscle development as it pertains to tail prehensility in mammals. This study uses an integrative approach of relating both MHC gene and protein expression with behavioral and morphometric changes to reveal a predominant fast MHC expression with subtle isoform transitions in caudal muscle across ontogeny. The functional shifts observed are most notably correlated with increased tail grasping for nesting activities.


Subject(s)
Hand Strength/physiology , Monodelphis/physiology , Myosin Heavy Chains/biosynthesis , Myosin Heavy Chains/genetics , Tail/physiology , Animals , Female , Gene Expression , Male , Myosins/biosynthesis , Myosins/genetics , Protein Isoforms/biosynthesis , Protein Isoforms/genetics
6.
Hum Mol Genet ; 26(6): 1146-1156, 2017 03 15.
Article in English | MEDLINE | ID: mdl-28104788

ABSTRACT

Congenital myopathies are muscle degenerative disorders with a broad clinical spectrum. A number of myopathies have been associated with molecular defects within sarcomeres, the force-generating component of the muscle cell. Whereas the highly regular organization of the myofibril has been studied in detail, in vivo assembly of sarcomeres remains a poorly understood process. Therefore, a more detailed knowledge of sarcomere assembly is crucial to better understand the pathogenic mechanisms within myopathies. Recently, mutations in myosin XVIIIB (MYO18B) have been associated with cases of myopathies, although the underlying mechanism for the resulting pathology remains to be defined. To analyze the role of myosin XVIIIB in skeletal muscle disease, zebrafish mutants for myo18b were generated. Full loss of myo18b function results in a complete lack of sarcomeric structure, revealing a highly surprising and essential role for myo18b in sarcomere assembly. Importantly, scattered thin and thick filaments assemble throughout the sarcoplasm; but fail to organize into recognizable sarcomeric structures in myo18b null mutants. In myo18b partial loss-of-function mutants sarcomeric structures are assembled, but thin and thick filaments remain misaligned within these structures. These observations suggest a novel model of sarcomere assembly where Myo18b coordinates the integration of preformed thick and thin filaments into the sarcomere. Disruption of this highly coordinated process results in a block in sarcomere biogenesis and the onset of myopathic pathology.


Subject(s)
Muscle, Skeletal/metabolism , Myopathies, Structural, Congenital/genetics , Myosins/genetics , Sarcomeres/genetics , Tumor Suppressor Proteins/genetics , Zebrafish/genetics , Actin Cytoskeleton/genetics , Actin Cytoskeleton/metabolism , Animals , Humans , Muscle, Skeletal/pathology , Mutant Proteins/genetics , Myopathies, Structural, Congenital/pathology , Myosins/biosynthesis , Sarcomeres/metabolism , Sarcomeres/pathology , Tumor Suppressor Proteins/biosynthesis , Zebrafish/physiology
7.
PLoS One ; 12(1): e0170613, 2017.
Article in English | MEDLINE | ID: mdl-28125727

ABSTRACT

Myosin storage myopathy is a protein aggregate myopathy associated with the characteristic subsarcolemmal accumulation of myosin heavy chain in muscle fibers. Despite similar histological findings, the clinical severity and age of onset are highly variable, ranging from no weakness to severe impairment of ambulation, and usually childhood-onset to onset later in life. Mutations located in the distal end of the tail of slow/ß-cardiac myosin heavy chain are associated with myosin storage myopathy. Four missense mutations (L1793P, R1845W, E1883K and H1901L), two of which have been reported in several unrelated families, are located within or closed to the assembly competence domain. This location is critical for the proper assembly of sarcomeric myosin rod filaments. To assess the mechanisms leading to protein aggregation in myosin storage myopathy and to evaluate the impact of these mutations on myosin assembly and muscle function, we expressed mutated myosin proteins in cultured human muscle cells and in the nematode Caenorhabditis elegans. While L1793P mutant myosin protein efficiently incorporated into the sarcomeric thick filaments, R1845W and H1901L mutants were prone to formation of myosin aggregates without assembly into striated sarcomeric thick filaments in cultured muscle cells. In C. elegans, mutant alleles of the myosin heavy chain gene unc-54 corresponding to R1845W, E1883K and H1901L, were as effective as the wild-type myosin gene in rescuing the null mutant worms, indicating that they retain functionality. Taken together, our results suggest that the basis for the pathogenic effect of the R1845W and H1901L mutations are primarily structural rather than functional. Further analyses are needed to identify the primary trigger for the histological changes seen in muscle biopsies of patients with L1793P and E1883K mutations.


Subject(s)
Caenorhabditis elegans Proteins/genetics , Muscular Diseases/congenital , Myosin Heavy Chains/genetics , Myosins/genetics , Protein Aggregation, Pathological/genetics , Actin Cytoskeleton/genetics , Actin Cytoskeleton/metabolism , Animals , Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/biosynthesis , Humans , Muscle Cells/metabolism , Muscle Cells/pathology , Muscle Fibers, Skeletal/metabolism , Muscle Fibers, Skeletal/pathology , Muscular Diseases/genetics , Muscular Diseases/metabolism , Muscular Diseases/pathology , Mutation , Myosin Heavy Chains/biosynthesis , Myosins/biosynthesis , Sarcomeres/genetics , Sarcomeres/metabolism
8.
Circ Res ; 120(5): 799-815, 2017 Mar 03.
Article in English | MEDLINE | ID: mdl-28115390

ABSTRACT

RATIONALE: Abdominal aortic aneurysms (AAAs) are characterized by pathological remodeling of the aortic wall. Although both increased Krüppel-like factor 5 (KLF5) expression and macrophage infiltration have been implicated in vascular remodeling, the role of KLF5 in macrophage infiltration and AAA formation remains unclear. OBJECTIVE: To determine the role of KLF5 in AAA formation and macrophage infiltration into AAAs. METHODS AND RESULTS: KLF5 expression was significantly increased in human AAA tissues and in 2 mouse models of experimental AAA. Moreover, in myeloid-specific Klf5 knockout mice (myeKlf5-/- mice), macrophage infiltration, medial smooth muscle cell loss, elastin degradation, and AAA formation were markedly decreased. In cell migration and time-lapse imaging analyses, the migration of murine myeKlf5-/- macrophages was impaired, and in luciferase reporter assays, KLF5 activated Myo9b (myosin IXB) transcription by direct binding to the Myo9b promoter. In subsequent coimmunostaining studies, Myo9b was colocalized with filamentous actin, cortactin, vinculin, and Tks5 in the podosomes of phorbol 12,13-dibutyrate-treated macrophages, indicating that Myo9b participates in podosome formation. Gain- and loss-of-function experiments showed that KLF5 promoted podosome formation in macrophages by upregulating Myo9b expression. Furthermore, RhoA-GTP levels increased after KLF5 knockdown in macrophages, suggesting that KLF5 lies upstream of RhoA signaling. Finally, Myo9b expression was increased in human AAA tissues, located in macrophages, and positively correlated with AAA size. CONCLUSIONS: These data are the first to indicate that KLF5-dependent regulation of Myo9b/RhoA is required for podosome formation and macrophage migration during AAA formation, warranting consideration of the KLF5-Myo9b-RhoA pathway as a therapeutic target for AAA treatment.


Subject(s)
Aortic Aneurysm, Abdominal/metabolism , Aortic Aneurysm, Abdominal/prevention & control , Kruppel-Like Transcription Factors/biosynthesis , Macrophages/metabolism , Myosins/biosynthesis , Podosomes/metabolism , rhoA GTP-Binding Protein/biosynthesis , Animals , Cell Line , HEK293 Cells , Humans , Kruppel-Like Transcription Factors/deficiency , Male , Mice , Mice, Knockout , Myosins/deficiency , Signal Transduction/physiology , rhoA GTP-Binding Protein/deficiency
9.
Pharmacol Ther ; 166: 136-49, 2016 10.
Article in English | MEDLINE | ID: mdl-27456554

ABSTRACT

Chronic heart failure is one of the leading causes for hospitalization in the United States and Europe, and is accompanied by high mortality. Current pharmacological therapy of chronic heart failure with reduced ejection fraction is largely based on compounds that inhibit the detrimental action of the adrenergic and the renin-angiotensin-aldosterone systems on the heart. More than one decade after spironolactone, two novel therapeutic principles have been added to the very recently released guidelines on heart failure therapy: the HCN-channel inhibitor ivabradine and the combined angiotensin and neprilysin inhibitor valsartan/sacubitril. New compounds that are in phase II or III clinical evaluation include novel non-steroidal mineralocorticoid receptor antagonists, guanylate cyclase activators or myosine activators. A variety of novel candidate targets have been identified and the availability of gene transfer has just begun to accelerate translation from basic science to clinical application. This review provides an overview of current pharmacology and pharmacotherapy in chronic heart failure at three stages: the updated clinical guidelines of the American Heart Association and the European Society of Cardiology, new drugs which are in clinical development, and finally innovative drug targets and their mechanisms in heart failure which are emerging from preclinical studies will be discussed.


Subject(s)
Cardiovascular Agents/pharmacology , Cardiovascular Agents/therapeutic use , Heart Failure/drug therapy , Heart Failure/physiopathology , Aminobutyrates/pharmacology , Angiotensin Receptor Antagonists/pharmacology , Benzazepines/pharmacology , Biphenyl Compounds , Cardiovascular Agents/administration & dosage , Cardiovascular Agents/adverse effects , Chronic Disease , Clinical Trials as Topic , Drug Combinations , Guanylate Cyclase/biosynthesis , Humans , Ivabradine , Mineralocorticoid Receptor Antagonists/pharmacology , Myosins/biosynthesis , Neprilysin/antagonists & inhibitors , Practice Guidelines as Topic , Renin-Angiotensin System , Tetrazoles/pharmacology , Valsartan
10.
Am J Med Genet A ; 170A(5): 1155-64, 2016 May.
Article in English | MEDLINE | ID: mdl-26866830

ABSTRACT

Shwachman-Diamond-Bodian syndrome (SDS) is a pleiotropic disorder in which the main features are bone marrow dysfunction and pancreatic insufficiency. Skeletal changes can occur, and in rare cases manifest as severe congenital thoracic dystrophy. We report a newborn boy with asphyxia, narrow thorax, and severe hypotonia initially suggesting a neuromuscular disease. The muscle biopsy showed myopathic changes with prominent variability in muscle fiber size and abnormal expression of developmental isoforms of myosin. The myofibrils showed focal loss and disorganization of myofilaments, and thickening of the Z-discs including some abortive nemaline rods. The boy became permanently dependent on assisted ventilation. Pancreatic insufficiency was subsequently diagnosed, explaining the malabsorption and failure to thrive. Except transitory thrombocytopenia and leukopenia, no major hematological abnormalities were noted. He had bilateral nephrocalcinosis with preserved renal function. Transitory liver dysfunction with elevated transaminase levels and parenchymal changes on ultrasound were registered. The clinical diagnosis was confirmed by detection of compound heterozygous mutations in SBDS using whole-exome sequencing: a recurrent intronic mutation causing aberrant splicing (c.258+2T>C) and a novel missense variant in a highly conserved codon (c.41A>G, p.Asn14Ser), considered to be damaging for the protein structure by in silico prediction programs. The carrier status of the parents has been confirmed. This case illustrates the challenges in differential diagnosis of pronounced neonatal hypotonia with asphyxia and highlights the muscular involvement in SDS. To our knowledge, this is the first report of myopathy evidenced in a patient with clinically and molecularly confirmed SDS.


Subject(s)
Bone Marrow Diseases/genetics , Exocrine Pancreatic Insufficiency/genetics , Lipomatosis/genetics , Muscular Diseases/genetics , Myofibrils/genetics , Proteins/genetics , Biopsy , Bone Marrow Diseases/physiopathology , Exocrine Pancreatic Insufficiency/physiopathology , Exome/genetics , Humans , Infant, Newborn , Lipomatosis/physiopathology , Male , Muscular Diseases/physiopathology , Mutation, Missense , Myofibrils/pathology , Myosins/biosynthesis , Myosins/genetics , Sequence Analysis, DNA , Shwachman-Diamond Syndrome
11.
Exp Eye Res ; 139: 90-6, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26240967

ABSTRACT

Epiretinal membrane (ERM) contraction is associated with a variety of ocular diseases that cause macular dysfunction. Trans-differentiated Müller cells have been identified in ERMs, and have been implicated to be involved in the contractile process. In this study, we tested the effect of dasatinib, an FDA-approved tyrosine kinase inhibitor, on matrix contraction caused by Müller cells, and examined molecular mechanism of action. Type I collagen matrix contraction assays were used to examine the effect of drugs on matrix contraction by trans-differentiated Müller cells. Fluophore-conjugated phalloidin was used for the detection of actin cytoskeleton, and Western-blot analyses were carried out to examine protein expression and phosphorylation status. Dasatinib inhibited collagen matrix contraction by trans-differentiated Müller cells that was associated with decreased cell spreading and reduction of actomyosin stress fibers. Concomitantly, dasatinib-treated Müller cells had reduced phosphorylation of Src family kinase, paxillin, as well as myosin II light chain. Specific inhibitors of Rho/ROCK and myosin II confirmed the critical role played by this pathway in Müller cell contraction. Our data demonstrate that dasatinib significantly reduced matrix contraction by Müller cells via inhibition of focal adhesion, as well as actomyosin contraction.


Subject(s)
Dasatinib/pharmacology , Ependymoglial Cells/metabolism , Extracellular Matrix/drug effects , Macular Degeneration/drug therapy , Myosins/genetics , Animals , Apoptosis , Cell Adhesion/drug effects , Disease Models, Animal , Ependymoglial Cells/pathology , Extracellular Matrix/metabolism , Extracellular Matrix/pathology , Immunoblotting , In Situ Nick-End Labeling , Macular Degeneration/metabolism , Macular Degeneration/pathology , Myosins/biosynthesis , Protein Kinase Inhibitors/pharmacology , Signal Transduction/drug effects , Stress Fibers/drug effects , Stress Fibers/metabolism , Swine
12.
Gene Expr Patterns ; 19(1-2): 36-44, 2015.
Article in English | MEDLINE | ID: mdl-26212629

ABSTRACT

The auditory-vestibular ganglion (AVG) is formed by the division of otic placode-derived neuroblasts, which then differentiate into auditory and vestibular afferent neurons. The developmental mechanisms that regulate neuronal cell fate determination, axonal pathfinding and innervation of otic neurons are poorly understood. The present study characterized the expression of myosin VIIA, along with the neuronal markers, Islet1, NeuroD1 and TuJ1, in the developing avian ear, during Hamburger-Hamilton (HH) stages 16-40. At early stages, when neuroblasts are delaminating from the otic epithelium, myosin VIIA expression was not observed. Myosin VIIA was initially detected in a subset of neurons during the early phase of neuronal differentiation (HH stage 20). As the AVG segregates into the auditory and vestibular portions, myosin VIIA was restricted to a subset of vestibular neurons, but was not present in auditory neurons. Myosin VIIA expression in the vestibular ganglion was maintained through HH stage 33 and was downregulated by stage 36. Myosin VIIA was also observed in the migrating processes of vestibular afferents as they begin to innervate the otic epithelium HH stage 22/23. Notably, afferents targeting hair cells of the cristae were positive for myosin VIIA while afferents targeting the utricular and saccular maculae were negative (HH stage 26-28). Although previous studies have reported that myosin VIIA is restricted to sensory hair cells, our data shows that myosin VIIA is also expressed in neurons of the developing chick ear. Our study suggests a possible role for myosin VIIA in axonal migration/pathfinding and/or innervation of vestibular afferents. In addition, myosin VIIA could be used as an early marker for vestibular neurons during the development of the avian AVG.


Subject(s)
Ear, Inner/embryology , Myosins/biosynthesis , Neurons/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/biosynthesis , Cell Differentiation/physiology , Chick Embryo , Ear, Inner/innervation , Ear, Inner/metabolism , Epithelium/metabolism , Myosin VIIa , Myosins/genetics , Neurogenesis/physiology , Neurons/cytology , Spiral Ganglion/cytology , Spiral Ganglion/metabolism , Tubulin/biosynthesis
13.
Am J Hum Genet ; 96(5): 841-9, 2015 May 07.
Article in English | MEDLINE | ID: mdl-25957469

ABSTRACT

Multiple pterygium syndrome (MPS) is a phenotypically and genetically heterogeneous group of rare Mendelian conditions characterized by multiple pterygia, scoliosis, and congenital contractures of the limbs. MPS typically segregates as an autosomal-recessive disorder, but rare instances of autosomal-dominant transmission have been reported. Whereas several mutations causing recessive MPS have been identified, the genetic basis of dominant MPS remains unknown. We identified four families affected by dominantly transmitted MPS characterized by pterygia, camptodactyly of the hands, vertebral fusions, and scoliosis. Exome sequencing identified predicted protein-altering mutations in embryonic myosin heavy chain (MYH3) in three families. MYH3 mutations underlie distal arthrogryposis types 1, 2A, and 2B, but all mutations reported to date occur in the head and neck domains. In contrast, two of the mutations found to cause MPS in this study occurred in the tail domain. The phenotypic overlap among persons with MPS, coupled with physical findings distinct from other conditions caused by mutations in MYH3, suggests that the developmental mechanism underlying MPS differs from that of other conditions and/or that certain functions of embryonic myosin might be perturbed by disruption of specific residues and/or domains. Moreover, the vertebral fusions in persons with MPS, coupled with evidence of MYH3 expression in bone, suggest that embryonic myosin plays a role in skeletal development.


Subject(s)
Arthrogryposis/genetics , Cytoskeletal Proteins/genetics , Myosins/biosynthesis , Arthrogryposis/physiopathology , Cytoskeletal Proteins/biosynthesis , Exome/genetics , Genetic Predisposition to Disease , High-Throughput Nucleotide Sequencing , Humans , Mutation , Myosins/genetics , Osteogenesis/genetics
14.
BMC Cancer ; 15: 275, 2015 Apr 12.
Article in English | MEDLINE | ID: mdl-25879875

ABSTRACT

BACKGROUND: AF-6/afadin plays an important role in the formation of adherence junctions. In breast and colon cancer, loss of AF-6/afadin induces cell migration and cell invasion. We aimed to elucidate the role of AF-6/afadin in human endometrial cancer. METHODS: Morphology and AF-6/afadin expression in endometrial cancer cell lines was investigated by 3-dimensional culture. We used Matrigel invasion assay to demonstrate AF-6/afadin knockdown induced invasive capability. Cell proliferation assay was performed to estimate chemoresistance to doxorubicin, paclitaxel and cisplatin induced by AF-6/afadin knockdown. The associations between AF-6/afadin expression and clinicopathological status were determined by immunohistochemical analysis in endometrial cancer tissues. Informed consent was obtained from all patients before the study. RESULTS: The majority of cell clumps in 3-dimensional cultures of Ishikawa cells that strongly expressed AF-6/afadin showed round gland-like structures. In contrast, the cell clumps in 3-dimensional cultures of HEC1A and AN3CA cells-both weakly expressing AF-6/afadin-showed irregular gland-like structures and disorganized colonies with no gland-like structures, respectively. AF-6/afadin knockdown resulted in reduced number of gland-like structures in 3-dimensional cultures and enhancement of cell invasion and phosphorylation of ERK1/2 and Src in the highly AF-6/afadin-expressing endometrial cancer cell line. Inhibitors of MAPK/ERK kinase (MEK) (U0126) and Src (SU6656) suppressed the AF-6/afadin knockdown-induced invasive capability. AF-6/afadin knockdown induced chemoresistance to doxorubicin, paclitaxel and cisplatin in Ishikawa cells, not in HEC1A. Immunohistochemical analysis showed that AF-6/afadin expression was significantly associated with myometrial invasion and high histological grade. CONCLUSIONS: AF-6/afadin regulates cell morphology and invasiveness. Invasive capability is partly regulated through the ERK and Src pathway. The inhibitors to these pathways might be molecular-targeted drugs which suppress myometrial invasion in endometrial cancer. AF-6/afadin could be a useful selection marker for fertility-sparing therapy for patients with atypical hyperplasia or grade 1 endometrioid adenocarcinoma with no myometrial invasion. AF-6/afadin knockdown induced chemoresistance especially to cisplatin. Therefore, loss of AF-6/afadin might be a predictive marker of chemoresistance to cisplatin.


Subject(s)
Cell Proliferation/drug effects , Drug Resistance, Neoplasm/genetics , Endometrial Neoplasms/genetics , Kinesins/biosynthesis , Myosins/biosynthesis , Adult , Aged , Aged, 80 and over , Cell Line, Tumor , Cell Movement/drug effects , Cisplatin/administration & dosage , Endometrial Neoplasms/drug therapy , Endometrial Neoplasms/pathology , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Kinesins/genetics , MAP Kinase Signaling System/drug effects , Middle Aged , Myosins/genetics , Neoplasm Invasiveness/genetics , Paclitaxel/administration & dosage
15.
Tissue Eng Part A ; 21(11-12): 1795-809, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25760435

ABSTRACT

The transcription factor atonal homolog 1 (ATOH1) has multiple homologues that are functionally conserved across species and is responsible for the generation of sensory hair cells. To evaluate potential functional differences between homologues, human and mouse ATOH1 (HATH1 and MATH-1, respectively) were nonvirally delivered to human Wharton's jelly cells (hWJCs) for the first time. Delivery of HATH1 to hWJCs demonstrated superior expression of inner ear hair cell markers and characteristics than delivery of MATH-1. Inhibition of HES1 and HES5 signaling further increased the atonal effect. Transfection of hWJCs with HATH1 DNA, HES1 siRNA, and HES5 siRNA displayed positive identification of key hair cell and support cell markers found in the cochlea, as well as a variety of cell shapes, sizes, and features not native to hair cells, suggesting the need for further examination of other cell types induced by HATH1 expression. In the first side-by-side evaluation of HATH1 and MATH-1 in human cells, substantial differences were observed, suggesting that the two atonal homologues may not be interchangeable in human cells, and artificial expression of HATH1 in hWJCs requires further study. In the future, this line of research may lead to engineered systems that would allow for evaluation of drug ototoxicity or potentially even direct therapeutic use.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/physiology , Cellular Reprogramming Techniques/methods , Hair Cells, Auditory, Inner/cytology , Mesenchymal Stem Cells/cytology , Animals , Basic Helix-Loop-Helix Transcription Factors/biosynthesis , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Lineage , Female , Fluorescent Dyes/metabolism , Genetic Vectors/genetics , Homeodomain Proteins/biosynthesis , Homeodomain Proteins/genetics , Humans , Infant, Newborn , Male , Mesenchymal Stem Cells/metabolism , Mice , Myosin VIIa , Myosins/biosynthesis , Myosins/genetics , Pyridinium Compounds/metabolism , Quaternary Ammonium Compounds/metabolism , RNA Interference , RNA, Small Interfering/genetics , Repressor Proteins/biosynthesis , Repressor Proteins/genetics , Signal Transduction , Species Specificity , Transcription Factor HES-1 , Transfection
16.
J Tissue Eng Regen Med ; 9(7): 847-51, 2015 Jul.
Article in English | MEDLINE | ID: mdl-24596180

ABSTRACT

In this communication, we introduce boron nitride nanotube (BNNT)-functionalised muscle cell/microfibre mesh constructs, obtained via tissue engineering, as a three-dimensional (3D) platform to study a wireless stimulation system for electrically responsive cells and tissues. Our stimulation strategy exploits the piezoelectric behaviour of some classes of ceramic nanoparticles, such as BNNTs, able to polarize under mechanical stress, e.g. using low-frequency ultrasound (US). In the microfibre scaffolds, C2C12 myoblasts were able to differentiate into viable myotubes and to internalize BNNTs, also upon US irradiation, so as to obtain a nanotech-assisted 3D in vitro model. We then tested our stimulatory system on 2D and 3D cellular models by investigating the expression of connexin 43 (Cx43), as a molecule involved in cell crosstalk and mechanotransduction, and myosin, as a myogenic differentiation marker. Cx43 gene expression revealed a marked model dependency. In control samples (without US and/or BNNTs), Cx43 was upregulated under 2D culture conditions (10.78 ± 1.05-fold difference). Interactions with BNNTs increased Cx43 expression in 3D samples. Cx43 mRNA dropped in 2D under the 'BNNTs + US' regimen, while it was best enhanced in 3D samples (3.58 ± 1.05 vs 13.74 ± 1.42-fold difference, p = 0.0001). At the protein level, the maximal expressions of Cx43 and myosin were detected in the 3D model. In contrast with the 3D model, in 2D cultures, BNNTs and US exerted a synergistic depletive effect upon myosin synthesis. These findings indicate that model dimensionality and stimulatory regimens can strongly affect the responses of signalling and differentiation molecules, proving the importance of developing proper in vitro platforms for biological modelling.


Subject(s)
Boron Compounds/chemistry , Mechanotransduction, Cellular , Muscle Fibers, Skeletal/metabolism , Myoblasts, Skeletal/metabolism , Tissue Engineering , Tissue Scaffolds/chemistry , Animals , Antigens, Differentiation/biosynthesis , Cell Line , Connexin 43/biosynthesis , Gene Expression Regulation , Mice , Muscle Fibers, Skeletal/cytology , Myoblasts, Skeletal/cytology , Myosins/biosynthesis , Nanotubes
17.
J Cell Biochem ; 115(2): 410-9, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24339252

ABSTRACT

Podocytes are a terminally differentiated and highly specialized cell type in the glomerulus that forms a crucial component of the glomerular filtration barrier. Recently, Myo1e was identified in the podocytes of glomeruli. Myo1e podocyte-specific knockout mice exhibit proteinuria, podocyte foot process effacement, glomerular basement membrane disorganization, signs of chronic renal injury, and kidney inflammation. After overexpression of Myo1e in a conditionally immortalized mouse podocyte cell line (MPC5), podocyte migration was evaluated via transwell assay, endocytosis was evaluated using FITC-transferrin, and adhesion was evaluated using a detachment assay after puromycin aminonucleoside treatment. Myo1e overexpression significantly increased the adherence of podocytes. ANOVA analysis indicated significant differences for cell adhesion between the overexpression and control groups (overexpression vs. control, t = 11.3199, P = 0.005; overexpression vs. negative control, t = 12.0570, P = 0.0006). Overexpression of Myo1e inhibited puromycin aminonucleoside-induced podocyte detachment, and the number of cells remaining on the bottom of the culture plate increased. Cell migration was enhanced in Myo1e-overexpressing podocytes in the transwell migration assay. Internalization of FITC-transferrin also increased in Myo1e-overexpressing podocytes relative to control cells. Overexpression of Myo1e can enhance podocyte migration ability, endocytosis, and attachment to the glomerular basement membrane. Restoration of Myo1e expression in podocytes may therefore strengthen their functional integrity against environmental and mechanical injury.


Subject(s)
Myosins/biosynthesis , Podocytes/metabolism , Animals , Cell Adhesion/genetics , Cell Line , Cell Movement/genetics , Endocytosis/genetics , Gene Expression Regulation , Kidney/cytology , Kidney/metabolism , Kidney Glomerulus , Mice , Mice, Knockout , Myosin Type I , Myosins/genetics , Zona Glomerulosa
18.
Curr Biol ; 23(23): 2434-9, 2013 Dec 02.
Article in English | MEDLINE | ID: mdl-24268417

ABSTRACT

In development and differentiation, morphological changes often accompany mechanical changes [1], but it is unclear whether or when cells in embryos sense tissue elasticity. The earliest embryo is uniformly pliable, while adult tissues vary widely in mechanics from soft brain and stiff heart to rigid bone [2]. However, cell sensitivity to microenvironment elasticity is debated based in part on results from complex three-dimensional culture models [3]. Regenerative cardiology provides strong motivation to clarify any cell-level sensitivities to tissue elasticity because rigid postinfarct regions limit pumping by the adult heart [4]. Here, we focus on the spontaneously beating embryonic heart and sparsely cultured cardiomyocytes, including cells derived from pluripotent stem cells. Tissue elasticity, Et, increases daily for heart to 1-2 kPa by embryonic day 4 (E4), and although this is ~10-fold softer than adult heart, the beating contractions of E4 cardiomyocytes prove optimal at ~Et,E4 both in vivo and in vitro. Proteomics reveals daily increases in a small subset of proteins, namely collagen plus cardiac-specific excitation-contraction proteins. Rapid softening of the heart's matrix with collagenase or stiffening it with enzymatic crosslinking suppresses beating. Sparsely cultured E4 cardiomyocytes on collagen-coated gels likewise show maximal contraction on matrices with native E4 stiffness, highlighting cell-intrinsic mechanosensitivity. While an optimal elasticity for striation proves consistent with the mathematics of force-driven sarcomere registration, contraction wave speed is linear in Et as theorized for excitation-contraction coupled to matrix elasticity. Pluripotent stem cell-derived cardiomyocytes also prove to be mechanosensitive to matrix and thus generalize the main observation that myosin II organization and contractile function are optimally matched to the load contributed by matrix elasticity.


Subject(s)
Extracellular Matrix Proteins/biosynthesis , Heart Rate , Heart/embryology , Myocardial Contraction/physiology , Myosins/biosynthesis , Cardiac Myosins/antagonists & inhibitors , Cell Differentiation , Cells, Cultured , Collagen/biosynthesis , Collagenases/pharmacology , Elasticity , Embryonic Stem Cells/metabolism , Heterocyclic Compounds, 4 or More Rings/pharmacology , Humans , Induced Pluripotent Stem Cells/metabolism , Myocytes, Cardiac/cytology , Myofibrils/physiology , Sarcomeres/physiology
19.
Eur J Obstet Gynecol Reprod Biol ; 171(1): 171-9, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24054828

ABSTRACT

OBJECTIVE: The molecular mechanisms underlying stress urinary incontinence (SUI) are not clear. In light of the limited availability of human tissue for study, we explored the changes in the urethra of C57BL/6 mice with experimentally induced SUI. STUDY DESIGN: Twelve virgin female mice were randomized into two groups: one group undergoing vaginal distension (VD) for 1h with an 8-mm dilator, and a non-instrumented control group. Four days after VD, leak point pressures (LPP) and maximum urethral closure pressure (MUCP) were assessed in these mice under urethane (1g/kg, i.p.) anesthesia. After measuring LPP and MUCP, the animals were sacrificed, and the urethras were removed for proteomic analysis using 2-dimensional differential gel electrophoresis (2D DIGE) and liquid chromatography-tandem mass spectrometry (LC-MS/MS) technology. Lastly, interaction between these proteins was further analyzed using MetaCore. RESULTS: LPP and MUCP values were significantly decreased in the 8-mm VD groups compared with the non-instrumented control group. Sixty-eight differentially expressed proteins of urethra from female mice with and without VD were identified. Of these, 19 proteins were up-regulated and 49 were down-regulated. The majority of the VD-induced proteins were involved in generation of precursor metabolites and energy, oxidation of reduction, regulation of apoptosis, and glycolysis. Myosin expression in the urethra was significantly decreased in the 8-mm VD group as compared with the control group. CONCLUSIONS: As a model of simulated birth trauma, VD can induce SUI in female mice. Under-expression of myosin plays a plausible role in the pathogenesis of SUI following vaginal trauma.


Subject(s)
Proteomics , Urethra/physiology , Urinary Incontinence, Stress/physiopathology , Vagina/injuries , Animals , Disease Models, Animal , Electrophoresis, Gel, Two-Dimensional , Female , Mice , Mice, Inbred C57BL , Myosins/biosynthesis , Parturition , Tandem Mass Spectrometry , Urethra/injuries , Urodynamics
20.
Exp Physiol ; 98(5): 1038-52, 2013 May.
Article in English | MEDLINE | ID: mdl-23291913

ABSTRACT

Skeletal muscle is a highly dynamic tissue that responds to endogenous and external stimuli, including alterations in mechanical loading and growth factors. In particular, the antigravity soleus muscle experiences significant muscle atrophy during disuse and extensive muscle damage upon reloading. Given that insulin-like growth factor-1 (IGF-1) has been implicated as a central regulator of muscle repair and modulation of muscle size, we examined the effect of virally mediated overexpression of IGF-1 on the soleus muscle following hindlimb cast immobilization and upon reloading. Recombinant IGF-1 cDNA virus was injected into one of the posterior hindlimbs of the mice, while the contralateral limb was injected with saline (control). At 20 weeks of age, both hindlimbs were immobilized for 2 weeks to induce muscle atrophy in the soleus and ankle plantarflexor muscle group. Subsequently, the mice were allowed to reambulate, and muscle damage and recovery were monitored over a period of 2-21 days. The primary finding of this study was that IGF-1 overexpression attenuated reloading-induced muscle damage in the soleus muscle, and accelerated muscle regeneration and force recovery. Muscle T2 assessed by magnetic resonance imaging, a non-specific marker of muscle damage, was significantly lower in IGF-1-injected compared with contralateral soleus muscles at 2 and 5 days reambulation (P<0.05). The reduced prevalence of muscle damage in IGF-1-injected soleus muscles was confirmed on histology, with a lower fractional area of abnormal muscle tissue in IGF-1-injected muscles at 2 days reambulation (33.2±3.3 versus 54.1±3.6%, P<0.05). Evidence of the effect of IGF-1 on muscle regeneration included timely increases in the number of central nuclei (21% at 5 days reambulation), paired-box transcription factor 7 (36% at 5 days), embryonic myosin (37% at 10 days) and elevated MyoD mRNA (7-fold at 2 days) in IGF-1-injected limbs (P<0.05). These findings demonstrate a potential role of IGF-1 in protecting unloaded skeletal muscles from damage and accelerating muscle repair and regeneration.


Subject(s)
Hindlimb Suspension/physiology , Insulin-Like Growth Factor I/biosynthesis , Muscle, Skeletal/injuries , Muscle, Skeletal/physiology , Muscular Atrophy/physiopathology , Animals , Female , Magnetic Resonance Imaging , Mice , Muscle, Skeletal/pathology , MyoD Protein/biosynthesis , Myosins/biosynthesis , Recovery of Function , Regeneration/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...