Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 7.946
Filtrar
1.
Cells ; 13(7)2024 Apr 08.
Artigo em Inglês | MEDLINE | ID: mdl-38607088

RESUMO

Muscle regeneration, representing an essential homeostatic process, relies mainly on the myogenic progress of resident satellite cells, and it is modulated by multiple physical and nutritional factors. Here, we investigated how myogenic differentiation-related factors and pathways respond to the first limiting amino acid lysine (Lys) in the fast and slow muscles, and their satellite cells (SCs), of swine. Thirty 28-day-old weaned piglets with similar body weights were subjected to three diet regimens: control group (d 0-28: 1.31% Lys, n = 12), Lys-deficient group (d 0-28: 0.83% Lys, n = 12), and Lys rescue group (d 0-14: 0.83% Lys; d 15-28: 1.31% Lys, n = 6). Pigs on d 15 and 29 were selectively slaughtered for muscular parameters evaluation. Satellite cells isolated from fast (semimembranosus) and slow (semitendinosus) muscles were also selected to investigate differentiation ability variations. We found Lys deficiency significantly hindered muscle development in both fast and slow muscles via the distinct manipulation of myogenic regulatory factors and the Wnt/Ca2+ pathway. In the SC model, Lys deficiency suppressed the Wnt/Ca2+ pathways and myosin heavy chain, myogenin, and myogenic regulatory factor 4 in slow muscle SCs but stimulated them in fast muscle SCs. When sufficient Lys was attained, the fast muscle-derived SCs Wnt/Ca2+ pathway (protein kinase C, calcineurin, calcium/calmodulin-dependent protein kinase II, and nuclear factor of activated T cells 1) was repressed, while the Wnt/Ca2+ pathway of its counterpart was stimulated to further the myogenic differentiation. Lys potentially manipulates the differentiation of porcine slow and fast muscle myofibers via the Wnt/Ca2+ pathway in opposite trends.


Assuntos
Lisina , Fatores de Regulação Miogênica , Animais , Suínos , Fatores de Regulação Miogênica/metabolismo , Lisina/metabolismo , Músculo Esquelético/metabolismo , Diferenciação Celular , Cadeias Pesadas de Miosina/metabolismo
2.
Mol Genet Genomics ; 299(1): 44, 2024 Apr 16.
Artigo em Inglês | MEDLINE | ID: mdl-38625590

RESUMO

Megacystis-microcolon-hypoperistalsis-syndrome (MMIHS) is a rare and early-onset congenital disease characterized by massive abdominal distension due to a large non-obstructive bladder, a microcolon and decreased or absent intestinal peristalsis. While in most cases inheritance is autosomal dominant and associated with heterozygous variant in ACTG2 gene, an autosomal recessive transmission has also been described including pathogenic bialellic loss-of-function variants in MYH11. We report here a novel family with visceral myopathy related to MYH11 gene, confirmed by whole genome sequencing (WGS). WGS was performed in two siblings with unusual presentation of MMIHS and their two healthy parents. The 38 years-old brother had severe bladder dysfunction and intestinal obstruction, whereas the 30 years-old sister suffered from end-stage kidney disease with neurogenic bladder and recurrent sigmoid volvulus. WGS was completed by retrospective digestive pathological analyses. Compound heterozygous variants of MYH11 gene were identified, associating a deletion of 1.2 Mb encompassing MYH11 inherited from the father and an in-frame variant c.2578_2580del, p.Glu860del inherited from the mother. Pathology analyses of the colon and the rectum revealed structural changes which significance of which is discussed. Cardiac and vascular assessment of the mother was normal. This is the second report of a visceral myopathy corresponding to late-onset form of MMIHS related to compound heterozygosity in MYH11; with complete gene deletion and a hypomorphic allele in trans. The hypomorphic allele harbored by the mother raised the question of the risk of aortic disease in adults. This case shows the interest of WGS in deciphering complex phenotypes, allowing adapted diagnosis and genetic counselling.


Assuntos
Anormalidades Múltiplas , Colo , Duodeno , Doenças Fetais , Obstrução Intestinal , Pseudo-Obstrução Intestinal , Bexiga Urinária , Adulto , Humanos , Masculino , Colo/anormalidades , Duodeno/anormalidades , Pseudo-Obstrução Intestinal/genética , Cadeias Pesadas de Miosina/genética , Estudos Retrospectivos , Bexiga Urinária/anormalidades , Feminino
3.
Cell Mol Life Sci ; 81(1): 195, 2024 Apr 24.
Artigo em Inglês | MEDLINE | ID: mdl-38653877

RESUMO

The Notch pathway is an evolutionarily conserved signaling system that is intricately regulated at multiple levels and it influences different aspects of development. In an effort to identify novel components involved in Notch signaling and its regulation, we carried out protein interaction screens which identified non-muscle myosin II Zipper (Zip) as an interacting partner of Notch. Physical interaction between Notch and Zip was further validated by co-immunoprecipitation studies. Immunocytochemical analyses revealed that Notch and Zip co-localize within same cytoplasmic compartment. Different alleles of zip also showed strong genetic interactions with Notch pathway components. Downregulation of Zip resulted in wing phenotypes that were reminiscent of Notch loss-of-function phenotypes and a perturbed expression of Notch downstream targets, Cut and Deadpan. Further, synergistic interaction between Notch and Zip resulted in highly ectopic expression of these Notch targets. Activated Notch-induced tumorous phenotype of larval tissues was enhanced by over-expression of Zip. Notch-Zip synergy resulted in the activation of JNK pathway that consequently lead to MMP activation and proliferation. Taken together, our results suggest that Zip may play an important role in regulation of Notch signaling.


Assuntos
Proteínas de Drosophila , Proteínas de Membrana , Cadeias Pesadas de Miosina , Receptores Notch , Transdução de Sinais , Animais , Proteínas de Drosophila/metabolismo , Proteínas de Drosophila/genética , Receptores Notch/metabolismo , Receptores Notch/genética , Drosophila melanogaster/metabolismo , Drosophila melanogaster/genética , Asas de Animais/metabolismo , Asas de Animais/crescimento & desenvolvimento , Drosophila/metabolismo , Drosophila/genética , Fenótipo , Metaloproteinases da Matriz/metabolismo , Metaloproteinases da Matriz/genética , Proliferação de Células , Miosina Tipo II/metabolismo , Miosina Tipo II/genética
4.
Genes (Basel) ; 15(3)2024 Mar 20.
Artigo em Inglês | MEDLINE | ID: mdl-38540440

RESUMO

BACKGROUND: Left ventricular hypertrophy (LVH) is a well-recognized cardiac dysfunction in infants of mothers with gestational diabetes mellitus (GDM). Left ventricular noncompaction (LVNC) is a cardiomyopathy that is morphologically characterized by numerous prominent trabeculations and deep intertrabecular recesses on cardiovascular imaging. However, there have been no case reports on neonates of mothers with GDM showing LVH and LVNC. CASE PRESENTATION: A patient, with LVH of a mother with GDM, was delivered at 36 weeks of gestation. Prominent trabeculations in the LV, suggesting LVNC, instead of LVH, were apparent 1 week after birth. A heterozygous deletion variant in the MYH7 gene (NM_000257.4: c.1090T>C, p.Phe364Leu) was discovered through genetic testing using a cardiomyopathy-associated gene panel in the patient and his father and the older brother who had LVNC. The patient is now 5 years old and does not have major cardiac events, although LVNC persisted. This is the first case of LVH secondary to a mother with GDM and LVNC with a novel variant in the MYH7 gene. CONCLUSION: Genetic testing should be conducted to obtain an accurate outcome and medical care in a patient with LVH and subsequently prominent hypertrabeculation in the LV.


Assuntos
Cardiomiopatias , Diabetes Gestacional , Cardiopatias Congênitas , Masculino , Lactente , Recém-Nascido , Feminino , Gravidez , Humanos , Pré-Escolar , Diabetes Gestacional/genética , Mães , Hipertrofia Ventricular Esquerda/genética , Cardiopatias Congênitas/genética , Cardiomiopatias/genética , Cadeias Pesadas de Miosina/genética , Miosinas Cardíacas/genética
5.
Front Biosci (Schol Ed) ; 16(1): 1, 2024 Feb 23.
Artigo em Inglês | MEDLINE | ID: mdl-38538344

RESUMO

BACKGROUND: Hypertrophic cardiomyopathy is the most frequent autosomal dominant disease, yet due to genetic heterogeneity, incomplete penetrance, and phenotype variability, the prognosis of the disease course in pathogenic variant carriers remains an issue. Identifying common patterns among the effects of different genetic variants is important. METHODS: We investigated the cause of familial hypertrophic cardiomyopathy (HCM) in a family with two patients suffering from a particularly severe disease. Searching for the genetic variants in HCM genes was performed using different sequencing methods. RESULTS: A new missense variant, p.Leu714Arg, was identified in exon 19 of the beta-myosin heavy chain gene (MYH7). The mutation was found in a region that encodes the 'converter domain' in the globular myosin head. This domain is essential for the conformational change of myosin during ATP cleavage and contraction cycle. Most reports on different mutations in this region describe severe phenotypic consequences. The two patients with the p.Leu714Arg mutation had heart failure early in life and died from HCM complications. CONCLUSIONS: This case presents a new likely pathogenic variant in MYH7 and supports the hypothesis that myosin converter mutations constitute a subclass of HCM mutations with a poor prognosis for the patient.


Assuntos
Cardiomiopatia Hipertrófica Familiar , Cardiomiopatia Hipertrófica , Humanos , Miosinas Cardíacas/genética , Cardiomiopatia Hipertrófica/genética , Cardiomiopatia Hipertrófica Familiar/diagnóstico por imagem , Cardiomiopatia Hipertrófica Familiar/genética , Mutação , Mutação de Sentido Incorreto/genética , Cadeias Pesadas de Miosina/genética , Fenótipo
6.
Genes Chromosomes Cancer ; 63(3): e23227, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38517106

RESUMO

AIMS: Kinase fusion-positive soft tissue tumors represent an emerging, molecularly defined group of mesenchymal tumors with a wide morphologic spectrum and diverse activating kinases. Here, we present two cases of soft tissue tumors with novel LTK fusions. METHODS AND RESULTS: Both cases presented as acral skin nodules (big toe and middle finger) in pediatric patients (17-year-old girl and 2-year-old boy). The tumors measured 2 and 3 cm in greatest dimension. Histologically, both cases exhibited bland-looking spindle cells infiltrating adipose tissue and accompanied by collagenous stroma. One case additionally displayed perivascular hyalinization and band-like stromal collagen. Both cases exhibited focal S100 staining, and one case had patchy coexpression of CD34. Targeted RNA-seq revealed the presence of novel in-frame MYH9::LTK and MYH10::LTK fusions, resulting in upregulation of LTK expression. Of interest, DNA methylation-based unsupervised clustering analysis in one case showed that the tumor clustered with dermatofibrosarcoma protuberans (DFSP). One tumor was excised with amputation with no local recurrence or distant metastasis at 18-month follow-up. The other case was initially marginally excised with local recurrence after one year, followed by wide local excision, with no evidence of disease at 10 years of follow-up. CONCLUSIONS: This is the first reported case series of soft tissue tumors harboring LTK fusion, expanding the molecular landscape of soft tissue tumors driven by activating kinase fusions. Furthermore, studies involving a larger number of cases and integrated genomic analyses will be warranted to fully elucidate the pathogenesis and classification of these tumors.


Assuntos
Neoplasias de Tecido Conjuntivo e de Tecidos Moles , Proteínas de Fusão Oncogênica , Neoplasias Cutâneas , Neoplasias de Tecidos Moles , Adolescente , Criança , Feminino , Humanos , Masculino , Antígenos CD34/metabolismo , Biomarcadores Tumorais/genética , Neoplasias de Tecido Conjuntivo e de Tecidos Moles/genética , Neoplasias de Tecido Conjuntivo e de Tecidos Moles/patologia , Receptores Proteína Tirosina Quinases , Neoplasias Cutâneas/patologia , Neoplasias de Tecidos Moles/genética , Neoplasias de Tecidos Moles/patologia , Proteínas de Fusão Oncogênica/genética , Cadeias Pesadas de Miosina/genética , Miosina não Muscular Tipo IIB/genética
7.
Breast Cancer Res ; 26(1): 48, 2024 Mar 19.
Artigo em Inglês | MEDLINE | ID: mdl-38504374

RESUMO

BACKGROUND: Breast cancer stem cell (CSC) expansion results in tumor progression and chemoresistance; however, the modulation of CSC pluripotency remains unexplored. Transmembrane protein 120B (TMEM120B) is a newly discovered protein expressed in human tissues, especially in malignant tissues; however, its role in CSC expansion has not been studied. This study aimed to determine the role of TMEM120B in transcriptional coactivator with PDZ-binding motif (TAZ)-mediated CSC expansion and chemotherapy resistance. METHODS: Both bioinformatics analysis and immunohistochemistry assays were performed to examine expression patterns of TMEM120B in lung, breast, gastric, colon, and ovarian cancers. Clinicopathological factors and overall survival were also evaluated. Next, colony formation assay, MTT assay, EdU assay, transwell assay, wound healing assay, flow cytometric analysis, sphere formation assay, western blotting analysis, mouse xenograft model analysis, RNA-sequencing assay, immunofluorescence assay, and reverse transcriptase-polymerase chain reaction were performed to investigate the effect of TMEM120B interaction on proliferation, invasion, stemness, chemotherapy sensitivity, and integrin/FAK/TAZ/mTOR activation. Further, liquid chromatography-tandem mass spectrometry analysis, GST pull-down assay, and immunoprecipitation assays were performed to evaluate the interactions between TMEM120B, myosin heavy chain 9 (MYH9), and CUL9. RESULTS: TMEM120B expression was elevated in lung, breast, gastric, colon, and ovarian cancers. TMEM120B expression positively correlated with advanced TNM stage, lymph node metastasis, and poor prognosis. Overexpression of TMEM120B promoted breast cancer cell proliferation, invasion, and stemness by activating TAZ-mTOR signaling. TMEM120B directly bound to the coil-coil domain of MYH9, which accelerated the assembly of focal adhesions (FAs) and facilitated the translocation of TAZ. Furthermore, TMEM120B stabilized MYH9 by preventing its degradation by CUL9 in a ubiquitin-dependent manner. Overexpression of TMEM120B enhanced resistance to docetaxel and doxorubicin. Conversely, overexpression of TMEM120B-∆CCD delayed the formation of FAs, suppressed TAZ-mTOR signaling, and abrogated chemotherapy resistance. TMEM120B expression was elevated in breast cancer patients with poor treatment outcomes (Miller/Payne grades 1-2) than in those with better outcomes (Miller/Payne grades 3-5). CONCLUSIONS: Our study reveals that TMEM120B bound to and stabilized MYH9 by preventing its degradation. This interaction activated the ß1-integrin/FAK-TAZ-mTOR signaling axis, maintaining stemness and accelerating chemotherapy resistance.


Assuntos
Neoplasias da Mama , Neoplasias Ovarianas , Humanos , Animais , Camundongos , Feminino , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Integrina beta1 , Linhagem Celular Tumoral , Serina-Treonina Quinases TOR/metabolismo , Proliferação de Células , Cadeias Pesadas de Miosina
9.
Zhonghua Yi Xue Yi Chuan Xue Za Zhi ; 41(3): 339-344, 2024 Mar 10.
Artigo em Chinês | MEDLINE | ID: mdl-38448026

RESUMO

OBJECTIVE: To explore the clinical and genetic characteristics of a neonate with Microvillus inclusion disease (MVID). METHODS: A neonate with MVID admitted to the First Affiliated Hospital of Zhengzhou University in May 2019 was selected as the study subject. Clinical data were collected. Whole exome sequencing (WES) was carried out, and candidate variants were verified by Sanger sequencing and multiple ligation-dependent probe amplification (MLPA). A literature was also carried out to summarize the clinical and genetic characteristics of MVID. RESULTS: The prematurely born neonate had presented with unexplained refractory diarrhea and metabolic acidosis. Active symptomatic treatment was ineffective, and the child had died at 2 months old. WES revealed that he had harbored compound heterozygous variants of the MYO5B gene, namely c.1591C>T (p.R531W) and deletion of exon 9. Sanger sequencing showed that the R531W variant was inherited form his father, and MLPA confirmed that the exon 9 deletion was inherited from his mother. Seven children with MVID were reported in China, of which one was lost during follow-up and six had deceased. One hundred eighty eight patients were reported worldwide and only one was cured. The clinical features of MVID had included refractory diarrhea, metabolic acidosis and poor prognosis. CONCLUSION: The child was diagnosed with MVID due to the compound heterozygous variants of the MYO5B gene, which has provided a basis for genetic counseling and prenatal diagnosis.


Assuntos
Acidose , Síndromes de Malabsorção , Microvilosidades , Mucolipidoses , Miosina Tipo V , Feminino , Humanos , Lactente , Recém-Nascido , Masculino , Gravidez , Diarreia/genética , Síndromes de Malabsorção/genética , Microvilosidades/patologia , Mucolipidoses/genética , Cadeias Pesadas de Miosina , Miosina Tipo V/genética
10.
Clin Lab ; 70(3)2024 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-38469783

RESUMO

BACKGROUND: The rapid development of automatic blood cell analyzers has greatly optimized complete blood count results. However, erroneous results relevant to automatic blood cell analyzers still exist. Pseudothrombocytopenia can be observed in both cases of anticoagulant-induced platelet aggregation, and the presence of large and giant platelets. METHODS: A rare case of a MYH9-related disorder, in which marked underestimation of platelet count was led by large and giant platelets using the impedance count by an automated hematology analyzer. Moreover, lancet-shaped and Dohle body-like cytoplasmic inclusions were detected in almost all white blood cells of the patient. RESULTS: The platelet count was done by an optical platelet counter or a fluorescence platelet counter, and peripheral blood smear was evaluated. In addition, the diagnosis of MYH9-related disorder was established by the molecular findings. CONCLUSIONS: Identification of the peripheral blood smear and familial history will eliminate the need for further laboratory testing and bone marrow examination.


Assuntos
Perda Auditiva Neurossensorial , Trombocitopenia , Trombocitopenia/congênito , Humanos , Contagem de Plaquetas/métodos , Trombocitopenia/diagnóstico , Trombocitopenia/genética , Plaquetas , Perda Auditiva Neurossensorial/diagnóstico , Cadeias Pesadas de Miosina/genética
11.
Circ Genom Precis Med ; 17(2): e004377, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38362799

RESUMO

BACKGROUND: Pathogenic autosomal-dominant missense variants in MYH7 (myosin heavy chain 7), which encodes the sarcomeric protein (ß-MHC [beta myosin heavy chain]) expressed in cardiac and skeletal myocytes, are a leading cause of hypertrophic cardiomyopathy and are clinically actionable. However, ≈75% of MYH7 missense variants are of unknown significance. While human-induced pluripotent stem cells (hiPSCs) can be differentiated into cardiomyocytes to enable the interrogation of MYH7 variant effect in a disease-relevant context, deep mutational scanning has not been executed using diploid hiPSC derivates due to low hiPSC gene-editing efficiency. Moreover, multiplexable phenotypes enabling deep mutational scanning of MYH7 variant hiPSC-derived cardiomyocytes are unknown. METHODS: To overcome these obstacles, we used CRISPRa On-Target Editing Retrieval enrichment to generate an hiPSC library containing 113 MYH7 codon variants suitable for deep mutational scanning. We first established that ß-MHC protein loss occurs in a hypertrophic cardiomyopathy human heart with a pathogenic MYH7 variant. We then differentiated the MYH7 missense variant hiPSC library to cardiomyocytes for multiplexed assessment of ß-MHC variant abundance by massively parallel sequencing and hiPSC-derived cardiomyocyte survival. RESULTS: Both the multiplexed assessment of ß-MHC abundance and hiPSC-derived cardiomyocyte survival accurately segregated all known pathogenic variants from synonymous variants. Functional data were generated for 4 variants of unknown significance and 58 additional MYH7 missense variants not yet detected in patients. CONCLUSIONS: This study leveraged hiPSC differentiation into disease-relevant cardiomyocytes to enable multiplexed assessments of MYH7 missense variants for the first time. Phenotyping strategies used here enable the application of deep mutational scanning to clinically actionable genes, which should reduce the burden of variants of unknown significance on patients and clinicians.


Assuntos
Cardiomiopatia Hipertrófica , Células-Tronco Pluripotentes Induzidas , Humanos , Miócitos Cardíacos/metabolismo , Cadeias Pesadas de Miosina/genética , Células-Tronco Pluripotentes Induzidas/metabolismo , Cardiomiopatia Hipertrófica/genética , Cardiomiopatia Hipertrófica/metabolismo , Diferenciação Celular/genética , Miosinas Cardíacas/genética
12.
Biosci Trends ; 18(1): 105-107, 2024 Mar 19.
Artigo em Inglês | MEDLINE | ID: mdl-38325822

RESUMO

Lactylation of α-myosin heavy chain (α-MHC) has recently been reported to preserve sarcomeric structure and function and attenuate the development of heart failure. Specifically, lactylation enhanced the interaction of α-MHC with the sarcomeric protein Titin, thereby maintaining normal sarcomeric structure and myocardial contractile function. Furthermore, the administration of lactate or inhibition of lactate efflux potentially treats heart failure by restoring lactylation of α-MHC and the interaction of α-MHC with Titin. This finding highlights the significant role of α-MHC lactylation in myocardial diseases and presents a new therapeutic target for the treatment of heart failure.


Assuntos
Insuficiência Cardíaca , Ácido Láctico , Humanos , Conectina/metabolismo , Ácido Láctico/metabolismo , Insuficiência Cardíaca/metabolismo , Miocárdio/metabolismo , Proteínas/metabolismo , Cadeias Pesadas de Miosina/metabolismo
13.
Cells ; 13(3)2024 Jan 26.
Artigo em Inglês | MEDLINE | ID: mdl-38334626

RESUMO

Myocarditis is one of the major causes of heart failure in children and young adults and can lead to dilated cardiomyopathy. Lymphocytic myocarditis could result from autoreactive CD4+ and CD8+ T cells, but defining antigen specificity in disease pathogenesis is challenging. To address this issue, we generated T cell receptor (TCR) transgenic (Tg) C57BL/6J mice specific to cardiac myosin heavy chain (Myhc)-α 334-352 and found that Myhc-α-specific TCRs were expressed in both CD4+ and CD8+ T cells. To investigate if the phenotype is more pronounced in a myocarditis-susceptible genetic background, we backcrossed with A/J mice. At the fourth generation of backcrossing, we observed that Tg T cells from naïve mice responded to Myhc-α 334-352, as evaluated by proliferation assay and carboxyfluorescein succinimidyl ester staining. The T cell responses included significant production of mainly pro-inflammatory cytokines, namely interferon (IFN)-γ, interleukin-17, and granulocyte macrophage-colony stimulating factor. While the naïve Tg mice had isolated myocardial lesions, immunization with Myhc-α 334-352 led to mild myocarditis, suggesting that further backcrossing to increase the percentage of A/J genome close to 99.99% might show a more severe disease phenotype. Further investigations led us to note that CD4+ T cells displayed the phenotype of cytotoxic T cells (CTLs) akin to those of conventional CD8+ CTLs, as determined by the expression of CD107a, IFN-γ, granzyme B natural killer cell receptor (NKG)2A, NKG2D, cytotoxic and regulatory T cell molecules, and eomesodermin. Taken together, the transgenic system described in this report may be a helpful tool to distinguish the roles of cytotoxic cardiac antigen-specific CD4+ T cells vs. those of CD8+ T cells in the pathogenesis of myocarditis.


Assuntos
Autoimunidade , Miocardite , Animais , Humanos , Camundongos , Linfócitos T CD4-Positivos , Linfócitos T CD8-Positivos/patologia , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Cadeias Pesadas de Miosina/genética , Receptores de Antígenos de Linfócitos T , Linfócitos T Citotóxicos
14.
Nat Commun ; 15(1): 1187, 2024 Feb 08.
Artigo em Inglês | MEDLINE | ID: mdl-38331992

RESUMO

Myosin VI is the only molecular motor that moves towards the minus end along actin filaments. Numerous cellular processes require myosin VI and tight regulations of the motor's activity. Defects in myosin VI activity are known to cause genetic diseases such as deafness and cardiomyopathy. However, the molecular mechanisms underlying the activity regulation of myosin VI remain elusive. Here, we determined the high-resolution cryo-electron microscopic structure of myosin VI in its autoinhibited state. Our structure reveals that autoinhibited myosin VI adopts a compact, monomeric conformation via extensive interactions between the head and tail domains, orchestrated by an elongated single-α-helix region resembling a "spine". This autoinhibited structure effectively blocks cargo binding sites and represses the motor's ATPase activity. Certain cargo adaptors such as GIPC can release multiple inhibitory interactions and promote motor activity, pointing to a cargo-mediated activation of the processive motor. Moreover, our structural findings allow rationalization of disease-associated mutations in myosin VI. Beyond the activity regulation mechanisms of myosin VI, our study also sheds lights on how activities of other myosin motors such as myosin VII and X might be regulated.


Assuntos
Cadeias Pesadas de Miosina , Miosinas , Microscopia Crioeletrônica , Cadeias Pesadas de Miosina/metabolismo , Miosinas/metabolismo , Citoesqueleto de Actina/metabolismo , Actinas/metabolismo
15.
Sci Rep ; 14(1): 3915, 2024 02 16.
Artigo em Inglês | MEDLINE | ID: mdl-38365813

RESUMO

Human induced pluripotent stem cells and their differentiation into cardiac myocytes (hiPSC-CMs) provides a unique and valuable platform for studies of cardiac muscle structure-function. This includes studies centered on disease etiology, drug development, and for potential clinical applications in heart regeneration/repair. Ultimately, for these applications to achieve success, a thorough assessment and physiological advancement of the structure and function of hiPSC-CMs is required. HiPSC-CMs are well noted for their immature and sub-physiological cardiac muscle state, and this represents a major hurdle for the field. To address this roadblock, we have developed a hiPSC-CMs (ß-MHC dominant) experimental platform focused on directed physiological enhancement of the sarcomere, the functional unit of cardiac muscle. We focus here on the myosin heavy chain (MyHC) protein isoform profile, the molecular motor of the heart, which is essential to cardiac physiological performance. We hypothesized that inducing increased expression of α-MyHC in ß-MyHC dominant hiPSC-CMs would enhance contractile performance of hiPSC-CMs. To test this hypothesis, we used gene editing with an inducible α-MyHC expression cassette into isogeneic hiPSC-CMs, and separately by gene transfer, and then investigated the direct effects of increased α-MyHC expression on hiPSC-CMs contractility and relaxation function. Data show improved cardiac functional parameters in hiPSC-CMs induced with α-MyHC. Positive inotropy and relaxation was evident in comparison to ß-MyHC dominant isogenic controls both at baseline and during pacing induced stress. This approach should facilitate studies of hiPSC-CMs disease modeling and drug screening, as well as advancing fundamental aspects of cardiac function parameters for the optimization of future cardiac regeneration, repair and re-muscularization applications.


Assuntos
Células-Tronco Pluripotentes Induzidas , Humanos , Miosinas Ventriculares/genética , Miosinas Ventriculares/metabolismo , Miosinas Ventriculares/farmacologia , Edição de Genes , Miocárdio , Miócitos Cardíacos/metabolismo , Diferenciação Celular , Miosinas/metabolismo , Cadeias Pesadas de Miosina/genética , Cadeias Pesadas de Miosina/metabolismo
16.
Biochemistry ; 63(5): 610-624, 2024 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-38357882

RESUMO

In Drosophila testis, myosin VI plays a special role, distinct from its motor function, by anchoring components to the unusual actin-based structures (cones) that are required for spermatid individualization. For this, the two calmodulin (CaM) light-chain molecules of myosin VI are replaced by androcam (ACaM), a related protein with 67% identity to CaM. Although ACaM has a similar bi-lobed structure to CaM, with two EF hand-type Ca2+ binding sites per lobe, only one functional Ca2+ binding site operates in the amino-terminus. To understand this light chain substitution, we used hydrogen-deuterium exchange mass spectrometry (HDX-MS) to examine dynamic changes in ACaM and CaM upon Ca2+ binding and interaction with the two CaM binding motifs of myosin VI (insert2 and IQ motif). HDX-MS reveals that binding of Ca2+ to ACaM destabilizes its N-lobe but stabilizes the entire C-lobe, whereas for CaM, Ca2+ binding induces a pattern of alternating stabilization/destabilization throughout. The conformation of this stable holo-C-lobe of ACaM seems to be a "prefigured" version of the conformation adopted by the holo-C-lobe of CaM for binding to insert2 and the IQ motif of myosin VI. Strikingly, the interaction of holo-ACaM with either peptide converts the holo-N-lobe to its Ca2+-free, more stable, form. Thus, ACaM in vivo should bind the myosin VI light chain sites in an apo-N-lobe/holo-C-lobe state that cannot fulfill the Ca2+-related functions of holo-CaM required for myosin VI motor assembly and activity. These findings indicate that inhibition of myosin VI motor activity is a precondition for transition to an anchoring function.


Assuntos
Calmodulina , Cadeias Pesadas de Miosina , Testículo , Masculino , Animais , Testículo/metabolismo , Deutério/metabolismo , Sequência de Aminoácidos , Calmodulina/metabolismo , Ligação Proteica , Drosophila/metabolismo , Espectrometria de Massas , Cálcio/metabolismo
17.
J Appl Physiol (1985) ; 136(5): 1015-1039, 2024 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-38328821

RESUMO

The efficacy of the NASA SPRINT exercise countermeasures program for quadriceps (vastus lateralis) and triceps surae (soleus) skeletal muscle health was investigated during 70 days of simulated microgravity. Individuals completed 6° head-down-tilt bedrest (BR, n = 9), bedrest with resistance and aerobic exercise (BRE, n = 9), or bedrest with resistance and aerobic exercise and low-dose testosterone (BRE + T, n = 8). All groups were periodically tested for muscle (n = 9 times) and aerobic (n = 4 times) power during bedrest. In BR, surprisingly, the typical bedrest-induced decrements in vastus lateralis myofiber size and power were either blunted (myosin heavy chain, MHC I) or eliminated (MHC IIa), along with no change (P > 0.05) in %MHC distribution and blunted quadriceps atrophy. In BRE, MHC I (vastus lateralis and soleus) and IIa (vastus lateralis) contractile performance was maintained (P > 0.05) or increased (P < 0.05). Vastus lateralis hybrid fiber percentage was reduced (P < 0.05) and energy metabolism enzymes and capillarization were generally maintained (P > 0.05), while not all of these positive responses were observed in the soleus. Exercise offsets 100% of quadriceps and approximately two-thirds of soleus whole muscle mass loss. Testosterone (BRE + T) did not provide any benefit over exercise alone for either muscle and for some myocellular parameters appeared detrimental. In summary, the periodic testing likely provided a partial exercise countermeasure for the quadriceps in the bedrest group, which is a novel finding given the extremely low exercise dose. The SPRINT exercise program appears to be viable for the quadriceps; however, refinement is needed to completely protect triceps surae myocellular and whole muscle health for astronauts on long-duration spaceflights.NEW & NOTEWORTHY This study provides unique exercise countermeasures development information for astronauts on long-duration spaceflights. The NASA SPRINT program was protective for quadriceps myocellular and whole muscle health, whereas the triceps surae (soleus) was only partially protected as has been shown with other programs. The bedrest control group data may provide beneficial information for overall exercise dose and targeting fast-twitch muscle fibers. Other unique approaches for the triceps surae are needed to supplement existing exercise programs.


Assuntos
Exercício Físico , Músculo Esquelético , Cadeias Pesadas de Miosina , Músculo Quadríceps , Simulação de Ausência de Peso , Humanos , Masculino , Músculo Quadríceps/fisiologia , Músculo Quadríceps/metabolismo , Simulação de Ausência de Peso/métodos , Adulto , Exercício Físico/fisiologia , Cadeias Pesadas de Miosina/metabolismo , Músculo Esquelético/fisiologia , Músculo Esquelético/metabolismo , United States National Aeronautics and Space Administration , Estados Unidos , Repouso em Cama/efeitos adversos , Testosterona/metabolismo , Testosterona/sangue , Voo Espacial/métodos , Atrofia Muscular/prevenção & controle , Atrofia Muscular/fisiopatologia , Treinamento de Força/métodos , Ausência de Peso/efeitos adversos , Força Muscular/fisiologia
18.
Eur J Orthod ; 46(2)2024 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-38364324

RESUMO

OBJECTIVE: Mouth breathing as a result of nasal obstruction affects craniofacial growth and development. This study aimed to investigate the effects of unilateral nasal obstruction and its recovery, along with the role of nitric oxide (NO) in masticatory muscle physiology. MATERIALS AND METHODS: Forty-eight 4-week-old male rats were divided into control and experimental groups. The five experimental groups were subjected to left-sided nasal obstruction by suturing the external nostril, and the sutures were removed after 1, 3, 5, 7, or 9 weeks to allow for varying recovery periods. We assessed morphological changes in masseter, temporalis, and digastric muscle, by examining cross-sectional area (CSA) and myosin heavy chain (MHC) isoform composition of muscle fibers. Reverse transcription-quantitative real-time polymerase chain reaction to measure messenger RNA (mRNA) levels for tumor necrosis factor-α (TNF-α), glucose transporter 4 (GLUT4), and neuronal nitric oxide synthase (nNOS) were conducted. RESULTS: The SpO2, CSA, and fibers showing MHC-2b isoforms were significantly lower, while RT-PCR showed higher mRNA levels in TNF-α and nNOS, and a decrease in GLUT4 mRNA in the jaw-closing muscles in the long-term nasal obstruction groups than that in the control group. LIMITATIONS: The study findings should be interpreted cautiously because of the functional differences between rodents and humans in terms of respiratory mechanisms. CONCLUSIONS: Unilateral nasal obstruction affects the morphology and contractile characteristics of the rat masticatory muscles during development, with possible involvement of NO in muscle hypofunction. These changes may revert to baseline levels if the nasal obstruction is eliminated before puberty in rats.


Assuntos
Obstrução Nasal , Humanos , Ratos , Masculino , Animais , Ratos Wistar , Fator de Necrose Tumoral alfa , Músculos da Mastigação , Cadeias Pesadas de Miosina/genética , RNA Mensageiro
19.
Invest Ophthalmol Vis Sci ; 65(2): 19, 2024 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-38334702

RESUMO

Purpose: The cytoskeleton of the extraocular muscles (EOMs) is significantly different from that of other muscles. We aimed to investigate the role of obscurin, a fundamental cytoskeletal protein, in the EOMs. Methods: The distribution of obscurin in human and zebrafish EOMs was compared using immunohistochemistry. The two obscurin genes in zebrafish, obscna and obscnb, were knocked out using CRISPR/Cas9, and the EOMs were investigated using immunohistochemistry, qPCR, and in situ hybridization. The optokinetic reflex (OKR) in five-day-old larvae and adult obscna-/-;obscnb-/- and sibling control zebrafish was analyzed. Swimming distance was recorded at the same age. Results: The obscurin distribution pattern was similar in human and zebrafish EOMs. The proportion of slow and fast myofibers was reduced in obscna-/-;obscnb-/- zebrafish EOMs but not in trunk muscle, whereas the number of myofibers containing cardiac myosin myh7 was significantly increased in EOMs of obscurin double mutants. Loss of obscurin resulted in less OKRs in zebrafish larvae but not in adult zebrafish. Conclusions: Obscurin expression is conserved in normal human and zebrafish EOMs. Loss of obscurin induces a myofiber type shift in the EOMs, with upregulation of cardiac myosin heavy chain, myh7, showing an adaptation strategy in EOMs. Our model will facilitate further studies in conditions related to obscurin.


Assuntos
Músculos Oculomotores , Proteínas Serina-Treonina Quinases , Fatores de Troca de Nucleotídeo Guanina Rho , Peixe-Zebra , Animais , Humanos , Imuno-Histoquímica , Músculo Esquelético/metabolismo , Cadeias Pesadas de Miosina/genética , Cadeias Pesadas de Miosina/metabolismo , Músculos Oculomotores/metabolismo , Fatores de Troca de Nucleotídeo Guanina Rho/genética , Proteínas Serina-Treonina Quinases/genética , Proteínas de Peixe-Zebra/genética
20.
Biomed Pharmacother ; 171: 116118, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38181716

RESUMO

The myosin heavy chain 9 (MYH9) gene encodes the heavy chain of non-muscle myosin IIA (NMIIA), which belongs to the myosin II subfamily of actin-based molecular motors. Previous studies have demonstrated that abnormal expression and mutations of MYH9 were correlated with MYH9-related diseases and tumors. Furthermore, earlier investigations identified MYH9 as a tumor suppressor. However, subsequent research revealed that MYH9 promoted tumorigenesis, progression and chemoradiotherapy resistance. Note-worthily, MYH9 has also been linked to viral infections, like severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), Epstein-Barr virus, and hepatitis B virus, as a receptor or co-receptor. In addition, MYH9 promotes the development of hepatocellular carcinoma by interacting with the hepatitis B virus-encoding X protein. Finally, various findings highlighted the role of MYH9 in the development of these illnesses, especially in tumors. This review summarizes the involvement of the MYH9-regulated signaling network in tumors and virus-related diseases and presents possible drug interventions on MYH9, providing insights for the use of MYH9 as a therapeutic target for tumors and virus-mediated diseases.


Assuntos
Infecções por Vírus Epstein-Barr , Neoplasias , Humanos , Herpesvirus Humano 4/metabolismo , Mutação , Actinas/genética , Cadeias Pesadas de Miosina/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...