Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Circ Res ; 133(5): 400-411, 2023 08 18.
Artículo en Inglés | MEDLINE | ID: mdl-37492967

RESUMEN

BACKGROUND: FLNC (filamin C), a member of the filamin family predominantly expressed in striated muscles, plays a crucial role in bridging the cytoskeleton and ECM (extracellular matrix) in cardiomyocytes, thereby maintaining heart integrity and function. Although genetic variants within the N-terminal ABD (actin-binding domain) of FLNC have been identified in patients with cardiomyopathy, the precise contribution of the actin-binding capability to FLNC's function in mammalian hearts remains poorly understood. METHODS: We conducted in silico analysis of the 3-dimensional structure of mouse FLNC to identify key amino acid residues within the ABD that are essential for FLNC's actin-binding capacity. Subsequently, we performed coimmunoprecipitation and immunofluorescent assays to validate the in silico findings and assess the impact of these mutations on the interactions with other binding partners and the subcellular localization of FLNC. Additionally, we generated and analyzed knock-in mouse models in which the FLNC-actin interaction was completely disrupted by these mutations. RESULTS: Our findings revealed that F93A/L98E mutations completely disrupted FLNC-actin interaction while preserving FLNC's ability to interact with other binding partners ITGB1 (ß1 integrin) and γ-SAG (γ-sarcoglycan), as well as maintaining FLNC subcellular localization. Loss of FLNC-actin interaction in embryonic cardiomyocytes resulted in embryonic lethality and cardiac developmental defects, including ventricular wall malformation and reduced cardiomyocyte proliferation. Moreover, disruption of FLNC-actin interaction in adult cardiomyocytes led to severe dilated cardiomyopathy, enhanced lethality and dysregulation of key cytoskeleton components. CONCLUSIONS: Our data strongly support the crucial role of FLNC as a bridge between actin filaments and ECM through its interactions with actin, ITGB1, γ-SAG, and other associated proteins in cardiomyocytes. Disruption of FLN-actin interaction may result in detachment of actin filaments from the extracellular matrix, ultimately impairing normal cardiac development and function. These findings also provide insights into mechanisms underlying cardiomyopathy associated with genetic variants in FLNC ABD and other regions.


Asunto(s)
Actinas , Cardiomiopatías , Ratones , Animales , Filaminas/genética , Filaminas/metabolismo , Actinas/genética , Actinas/metabolismo , Músculo Esquelético/metabolismo , Cardiomiopatías/genética , Miocitos Cardíacos/metabolismo , Mutación , Mamíferos
2.
PLoS Genet ; 19(1): e1010630, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36706168

RESUMEN

FLNC, encoding filamin C, is one of the most mutated genes in dilated and hypertrophic cardiomyopathy. However, the precise role of filamin C in mammalian heart remains unclear. In this study, we demonstrated Flnc global (FlncgKO) and cardiomyocyte-specific knockout (FlnccKO) mice died in utero from severely ruptured ventricular myocardium, indicating filamin C is required to maintain the structural integrity of myocardium in the mammalian heart. Contrary to the common belief that filamin C acts as an integrin inactivator, we observed attenuated activation of ß1 integrin specifically in the myocardium of FlncgKO mice. Although deleting ß1 integrin from cardiomyocytes did not recapitulate the heart rupture phenotype in Flnc knockout mice, deleting both ß1 integrin and filamin C from cardiomyocytes resulted in much more severe heart ruptures than deleting filamin C alone. Our results demonstrated that filamin C works in concert with ß1 integrin to maintain the structural integrity of myocardium during mammalian heart development.


Asunto(s)
Filaminas , Integrina beta1 , Miocardio , Animales , Ratones , Cardiomiopatía Hipertrófica , Filaminas/genética , Integrina beta1/genética , Miocitos Cardíacos
3.
Am J Physiol Heart Circ Physiol ; 324(3): H288-H292, 2023 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-36563012

RESUMEN

The use of digital image analysis and count regression models contributes to the reproducibility and rigor of histological studies in cardiovascular research. The use of formalized computer-based quantification strategies of histological images essentially removes potential researcher bias, allows for higher analysis throughput, and enables easy sharing of formalized quantification tools, contributing to research transparency, and data transferability. Moreover, the use of count regression models rather than ratios in statistical analysis of cell population data incorporates the extent of sampling into analysis and acknowledges the non-Gaussian nature of count distributions. Using quantification of proliferating cardiomyocytes in embryonic murine hearts as an example, we describe how these improvements can be implemented using open-source artificial intelligence-based image analysis tools and novel count regression models to efficiently analyze real-life data.


Asunto(s)
Inteligencia Artificial , Miocitos Cardíacos , Ratones , Animales , Reproducibilidad de los Resultados , Procesamiento de Imagen Asistido por Computador/métodos , Algoritmos
4.
Circulation ; 145(8): 586-602, 2022 02 22.
Artículo en Inglés | MEDLINE | ID: mdl-34915728

RESUMEN

BACKGROUND: Left ventricular noncompaction cardiomyopathy (LVNC) was discovered half a century ago as a cardiomyopathy with excessive trabeculation and a thin ventricular wall. In the decades since, numerous studies have demonstrated that LVNC primarily has an effect on left ventricles (LVs) and is often associated with LV dilation and dysfunction. However, in part because of the lack of suitable mouse models that faithfully mirror the selective LV vulnerability in patients, mechanisms underlying the susceptibility of LVs to dilation and dysfunction in LVNC remain unknown. Genetic studies have revealed that deletions and mutations in PRDM16 (PR domain-containing 16) cause LVNC, but previous conditional Prdm16 knockout mouse models do not mirror the LVNC phenotype in patients, and the underlying molecular mechanisms by which PRDM16 deficiency causes LVNC are still unclear. METHODS: Prdm16 cardiomyocyte-specific knockout (Prdm16cKO) mice were generated and analyzed for cardiac phenotypes. RNA sequencing and chromatin immunoprecipitation deep sequencing were performed to identify direct transcriptional targets of PRDM16 in cardiomyocytes. Single-cell RNA sequencing in combination with spatial transcriptomics was used to determine cardiomyocyte identity at the single-cell level. RESULTS: Cardiomyocyte-specific ablation of Prdm16 in mice caused LV-specific dilation and dysfunction, as well as biventricular noncompaction, which fully recapitulated LVNC in patients. PRDM16 functioned mechanistically as a compact myocardium-enriched transcription factor that activated compact myocardial genes while repressing trabecular myocardial genes in LV compact myocardium. Consequently, Prdm16cKO LV compact myocardial cardiomyocytes shifted from their normal transcriptomic identity to a transcriptional signature resembling trabecular myocardial cardiomyocytes or neurons. Chamber-specific transcriptional regulation by PRDM16 was attributable in part to its cooperation with LV-enriched transcription factors Tbx5 and Hand1. CONCLUSIONS: These results demonstrate that disruption of proper specification of compact cardiomyocytes may play a key role in the pathogenesis of LVNC. They also shed light on underlying mechanisms of the LV-restricted transcriptional program governing LV chamber growth and maturation, providing a tangible explanation for the susceptibility of LV in a subset of LVNC cardiomyopathies.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Ventrículos Cardíacos/metabolismo , Miocardio/metabolismo , Miocitos Cardíacos/metabolismo , Factores de Transcripción/metabolismo , Animales , Proteínas de Unión al ADN/genética , Ratones , Ratones Noqueados , Factores de Transcripción/genética
5.
J Clin Invest ; 130(10): 5109-5111, 2020 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-32865520

RESUMEN

The right ventricle (RV) is involved in systemic circulation in the fetal mammalian heart but quickly transitions to being solely responsible for pulmonary circulation after birth when the left ventricle (LV) becomes the systemic ventricle. To handle the increased workload, LV growth greatly outpaces that of the RV during postnatal stages. However, the molecular basis for this differential growth pattern between the 2 chambers is largely unknown. In this issue of the JCI, Yokota et al. reveal that the p38 mitogen-activated protein kinase (MAPK)/IRE1α/XBP1 axis specifically controls postnatal RV growth by suppressing cell cycle regulatory genes.


Asunto(s)
Ventrículos Cardíacos , Proteínas Quinasas p38 Activadas por Mitógenos , Animales , Endorribonucleasas , Femenino , Corazón , Embarazo , Proteínas Serina-Treonina Quinasas , Proteínas Quinasas p38 Activadas por Mitógenos/genética
6.
JCI Insight ; 5(16)2020 08 20.
Artículo en Inglés | MEDLINE | ID: mdl-32814711

RESUMEN

Nexilin (NEXN) was recently identified as a component of the junctional membrane complex required for development and maintenance of cardiac T-tubules. Loss of Nexn in mice leads to a rapidly progressive dilated cardiomyopathy (DCM) and premature death. A 3 bp deletion (1948-1950del) leading to loss of the glycine in position 650 (G650del) is classified as a variant of uncertain significance in humans and may function as an intermediate risk allele. To determine the effect of the G650del variant on cardiac structure and function, we generated a G645del-knockin (G645del is equivalent to human G650del) mouse model. Homozygous G645del mice express about 30% of the Nexn expressed by WT controls and exhibited a progressive DCM characterized by reduced T-tubule formation, with disorganization of the transverse-axial tubular system. On the other hand, heterozygous Nexn global KO mice and genetically engineered mice encoding a truncated Nexn missing the first N-terminal actin-binding domain exhibited normal cardiac function, despite expressing only 50% and 20% of the Nexn, respectively, expressed by WT controls, suggesting that not only quantity but also quality of Nexn is necessary for a proper function. These findings demonstrated that Nexn G645 is crucial for Nexn's function in tubular system organization and normal cardiac function.


Asunto(s)
Cardiomiopatías/genética , Corazón/fisiopatología , Proteínas de Microfilamentos/genética , Animales , Cardiomiopatías/fisiopatología , Cardiomiopatía Dilatada/genética , Modelos Animales de Enfermedad , Homocigoto , Ratones Mutantes , Proteínas de Microfilamentos/metabolismo , Mutación , Miocitos Cardíacos/patología
7.
PLoS Genet ; 16(4): e1008730, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32251422

RESUMEN

O-linked N-acetylglucosamine (GlcNAc) transferase (OGT) is the only enzyme catalyzing O-GlcNAcylation. Although it has been shown that OGT plays an essential role in maintaining postnatal heart function, its role in heart development remains unknown. Here we showed that loss of OGT in early fetal cardiomyocytes led to multiple heart developmental defects including hypertrabeculation, biventricular dilation, atrial septal defects, ventricular septal defects, and defects in coronary vessel development. In addition, RNA sequencing revealed that Angiopoietin-1, required within cardiomyocytes for both myocardial and coronary vessel development, was dramatically downregulated in cardiomyocyte-specific OGT knockout mouse hearts. In conclusion, our data demonstrated that OGT plays an essential role in regulating heart development through activating expression of cardiomyocyte Angiopoietin-1.


Asunto(s)
Corazón/embriología , Miocitos Cardíacos/metabolismo , N-Acetilglucosaminiltransferasas/metabolismo , Angiopoyetina 1/genética , Angiopoyetina 1/metabolismo , Animales , Células Cultivadas , Corazón/fisiología , Ratones , Ratones Endogámicos C57BL , N-Acetilglucosaminiltransferasas/genética
8.
Circulation ; 140(1): 55-66, 2019 07 02.
Artículo en Inglés | MEDLINE | ID: mdl-30982350

RESUMEN

BACKGROUND: Membrane contact sites are fundamental for transmission and translation of signals in multicellular organisms. The junctional membrane complexes in the cardiac dyads, where transverse (T) tubules are juxtaposed to the sarcoplasmic reticulum, are a prime example. T-tubule uncoupling and remodeling are well-known features of cardiac disease and heart failure. Even subtle alterations in the association between T-tubules and the junctional sarcoplasmic reticulum can cause serious cardiac disorders. NEXN (nexilin) has been identified as an actin-binding protein, and multiple mutations in the NEXN gene are associated with cardiac diseases, but the precise role of NEXN in heart function and disease is still unknown. METHODS: Nexn global and cardiomyocyte-specific knockout mice were generated. Comprehensive phenotypic and RNA sequencing and mass spectrometry analyses were performed. Heart tissue samples and isolated single cardiomyocytes were analyzed by electron and confocal microscopy. RESULTS: Global and cardiomyocyte-specific loss of Nexn in mice resulted in a rapidly progressive dilated cardiomyopathy. In vivo and in vitro analyses revealed that NEXN interacted with junctional sarcoplasmic reticulum proteins, was essential for optimal calcium transients, and was required for initiation of T-tubule invagination and formation. CONCLUSIONS: These results demonstrated that NEXN is a pivotal component of the junctional membrane complex and is required for initiation and formation of T-tubules, thus providing insight into mechanisms underlying cardiomyopathy in patients with mutations in NEXN.


Asunto(s)
Cardiomiopatía Dilatada/metabolismo , Membrana Celular/metabolismo , Uniones Intercelulares/metabolismo , Proteínas de Microfilamentos/deficiencia , Fibras Musculares Esqueléticas/metabolismo , Miocitos Cardíacos/metabolismo , Animales , Canales de Calcio Tipo L/metabolismo , Cardiomiopatía Dilatada/genética , Cardiomiopatía Dilatada/patología , Membrana Celular/genética , Membrana Celular/patología , Células Cultivadas , Uniones Intercelulares/genética , Uniones Intercelulares/patología , Ratones , Ratones Noqueados , Ratones Transgénicos , Proteínas de Microfilamentos/genética , Fibras Musculares Esqueléticas/patología , Miocitos Cardíacos/patología
9.
Proc Natl Acad Sci U S A ; 114(45): 11956-11961, 2017 11 07.
Artículo en Inglés | MEDLINE | ID: mdl-29078393

RESUMEN

Small heat shock protein HSPB7 is highly expressed in the heart. Several mutations within HSPB7 are associated with dilated cardiomyopathy and heart failure in human patients. However, the precise role of HSPB7 in the heart is still unclear. In this study, we generated global as well as cardiac-specific HSPB7 KO mouse models and found that loss of HSPB7 globally or specifically in cardiomyocytes resulted in embryonic lethality before embryonic day 12.5. Using biochemical and cell culture assays, we identified HSPB7 as an actin filament length regulator that repressed actin polymerization by binding to monomeric actin. Consistent with HSPB7's inhibitory effects on actin polymerization, HSPB7 KO mice had longer actin/thin filaments and developed abnormal actin filament bundles within sarcomeres that interconnected Z lines and were cross-linked by α-actinin. In addition, loss of HSPB7 resulted in up-regulation of Lmod2 expression and mislocalization of Tmod1. Furthermore, crossing HSPB7 null mice into an Lmod2 null background rescued the elongated thin filament phenotype of HSPB7 KOs, but double KO mice still exhibited formation of abnormal actin bundles and early embryonic lethality. These in vivo findings indicated that abnormal actin bundles, not elongated thin filament length, were the cause of embryonic lethality in HSPB7 KOs. Our findings showed an unsuspected and critical role for a specific small heat shock protein in directly modulating actin thin filament length in cardiac muscle by binding monomeric actin and limiting its availability for polymerization.


Asunto(s)
Citoesqueleto de Actina/metabolismo , Cardiomiopatías/genética , Proteínas de Choque Térmico HSP27/genética , Cardiopatías Congénitas/genética , Corazón/embriología , Citoesqueleto de Actina/genética , Animales , Proteínas del Citoesqueleto/biosíntesis , Proteínas del Citoesqueleto/genética , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Musculares/biosíntesis , Proteínas Musculares/genética , Miocardio/citología , Miocitos Cardíacos/citología , Organogénesis/genética , Sarcómeros/metabolismo , Tropomodulina/metabolismo
10.
J Clin Invest ; 127(8): 3189-3200, 2017 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-28737513

RESUMEN

Defective protein quality control (PQC) systems are implicated in multiple diseases. Molecular chaperones and co-chaperones play a central role in functioning PQC. Constant mechanical and metabolic stress in cardiomyocytes places great demand on the PQC system. Mutation and downregulation of the co-chaperone protein BCL-2-associated athanogene 3 (BAG3) are associated with cardiac myopathy and heart failure, and a BAG3 E455K mutation leads to dilated cardiomyopathy (DCM). However, the role of BAG3 in the heart and the mechanisms by which the E455K mutation leads to DCM remain obscure. Here, we found that cardiac-specific Bag3-KO and E455K-knockin mice developed DCM. Comparable phenotypes in the 2 mutants demonstrated that the E455K mutation resulted in loss of function. Further experiments revealed that the E455K mutation disrupted the interaction between BAG3 and HSP70. In both mutants, decreased levels of small heat shock proteins (sHSPs) were observed, and a subset of proteins required for cardiomyocyte function was enriched in the insoluble fraction. Together, these observations suggest that interaction between BAG3 and HSP70 is essential for BAG3 to stabilize sHSPs and maintain cardiomyocyte protein homeostasis. Our results provide insight into heart failure caused by defects in BAG3 pathways and suggest that increasing BAG3 protein levels may be of therapeutic benefit in heart failure.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Reguladoras de la Apoptosis/genética , Cardiomiopatías/metabolismo , Proteínas de Choque Térmico/metabolismo , Mutación , Animales , Cardiomiopatías/genética , Técnicas de Cocultivo , Ecocardiografía , Proteínas HSP70 de Choque Térmico/metabolismo , Insuficiencia Cardíaca/metabolismo , Estimación de Kaplan-Meier , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Chaperonas Moleculares/metabolismo , Miocitos Cardíacos/metabolismo , Fenotipo
11.
JCI Insight ; 1(16): e89908, 2016 Oct 06.
Artículo en Inglés | MEDLINE | ID: mdl-27734035

RESUMEN

Adipose tissue is a key endocrine organ that governs systemic homeostasis. PPARγ is a master regulator of adipose tissue signaling that plays an essential role in insulin sensitivity, making it an important therapeutic target. The selective PPARγ agonist rosiglitazone (RSG) has been used to treat diabetes. However, adverse cardiovascular effects have seriously hindered its clinical application. Experimental models have revealed that PPARγ activation increases cardiac hypertrophy. RSG stimulates cardiac hypertrophy and oxidative stress in cardiomyocyte-specific PPARγ knockout mice, implying that RSG might stimulate cardiac hypertrophy independently of cardiomyocyte PPARγ. However, candidate cell types responsible for RSG-induced cardiomyocyte hypertrophy remain unexplored. Utilizing cocultures of adipocytes and cardiomyocytes, we found that stimulation of PPARγ signaling in adipocytes increased miR-200a expression and secretion. Delivery of miR-200a in adipocyte-derived exosomes to cardiomyocytes resulted in decreased TSC1 and subsequent mTOR activation, leading to cardiomyocyte hypertrophy. Treatment with an antagomir to miR-200a blunted this hypertrophic response in cardiomyocytes. In vivo, specific ablation of PPARγ in adipocytes was sufficient to blunt hypertrophy induced by RSG treatment. By delineating mechanisms by which RSG elicits cardiac hypertrophy, we have identified pathways that mediate the crosstalk between adipocytes and cardiomyocytes to regulate cardiac remodeling.


Asunto(s)
Adipocitos/metabolismo , Cardiomegalia/genética , MicroARNs/genética , Miocitos Cardíacos/metabolismo , PPAR gamma/metabolismo , Células 3T3 , Animales , Cardiomegalia/inducido químicamente , Células Cultivadas , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , PPAR gamma/antagonistas & inhibidores , PPAR gamma/genética , Rosiglitazona , Serina-Treonina Quinasas TOR/metabolismo , Tiazolidinedionas/efectos adversos , Proteína 1 del Complejo de la Esclerosis Tuberosa , Proteínas Supresoras de Tumor/metabolismo
12.
PLoS One ; 10(9): e0138624, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26398569

RESUMEN

Skeletal muscle is the major site for glucose disposal, the impairment of which closely associates with the glucose intolerance in diabetic patients. Diabetes-related ankyrin repeat protein (DARP/Ankrd23) is a member of muscle ankyrin repeat proteins, whose expression is enhanced in the skeletal muscle under diabetic conditions; however, its role in energy metabolism remains poorly understood. Here we report a novel role of DARP in the regulation of glucose homeostasis through modulating AMP-activated protein kinase (AMPK) activity. DARP is highly preferentially expressed in skeletal muscle, and its expression was substantially upregulated during myotube differentiation of C2C12 myoblasts. Interestingly, DARP-/- mice demonstrated better glucose tolerance despite similar body weight, while their insulin sensitivity did not differ from that in wildtype mice. We found that phosphorylation of AMPK, which mediates insulin-independent glucose uptake, in skeletal muscle was significantly enhanced in DARP-/- mice compared to that in wildtype mice. Gene silencing of DARP in C2C12 myotubes enhanced AMPK phosphorylation, whereas overexpression of DARP in C2C12 myoblasts reduced it. Moreover, DARP-silencing increased glucose uptake and oxidation in myotubes, which was abrogated by the treatment with AICAR, an AMPK activator. Of note, improved glucose tolerance in DARP-/- mice was abolished when mice were treated with AICAR. Mechanistically, gene silencing of DARP enhanced protein expression of LKB1 that is a major upstream kinase for AMPK in myotubes in vitro and the skeletal muscle in vivo. Together with the altered expression under diabetic conditions, our data strongly suggest that DARP plays an important role in the regulation of glucose homeostasis under physiological and pathological conditions, and thus DARP is a new therapeutic target for the treatment of diabetes mellitus.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Glucosa/metabolismo , Músculo Esquelético/enzimología , Proteínas Nucleares/metabolismo , Proteínas Quinasas Activadas por AMP/química , Aminoimidazol Carboxamida/análogos & derivados , Aminoimidazol Carboxamida/farmacología , Animales , Peso Corporal , Diferenciación Celular , Línea Celular , Regulación hacia Abajo , Metabolismo Energético , Transportador de Glucosa de Tipo 1/metabolismo , Transportador de Glucosa de Tipo 4/metabolismo , Insulina/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fibras Musculares Esqueléticas/química , Fibras Musculares Esqueléticas/metabolismo , Músculo Esquelético/metabolismo , Proteínas Nucleares/deficiencia , Proteínas Nucleares/genética , Fosforilación/efectos de los fármacos , Proteínas Serina-Treonina Quinasas/metabolismo , Interferencia de ARN , Ribonucleótidos/farmacología , Regulación hacia Arriba
13.
RNA Biol ; 12(5): 479-85, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25901584

RESUMEN

The U2AF heterodimer is generally accepted to play a vital role in defining functional 3' splice sites in pre-mRNA splicing. Given prevalent mutations in U2AF, particularly in the U2AF1 gene (which encodes for the U2AF35 subunit) in blood disorders and other human cancers, there are renewed interests in these classic splicing factors to further understand their regulatory functions in RNA metabolism in both physiological and disease settings. We recently reported that U2AF has a maximal capacity to directly bind ˜88% of functional 3' splice sites in the human genome and that numerous U2AF binding events also occur in various exonic and intronic locations, thus providing additional mechanisms for the regulation of alternative splicing besides their traditional role in titrating weak splice sites in the cell. These findings, coupled with the existence of multiple related proteins to both U2AF65 and U2AF35, beg a series of questions on the universal role of U2AF in functional 3' splice site definition, their binding specificities in vivo, potential mechanisms to bypass their requirement for certain intron removal events, contribution of splicing-independent functions of U2AF to important cellular functions, and the mechanism for U2AF mutations to invoke specific diseases in humans.


Asunto(s)
Empalme Alternativo/genética , Genoma , Proteínas Nucleares/metabolismo , Ribonucleoproteínas/metabolismo , Secuencia Conservada , Enfermedad/genética , Humanos , Modelos Biológicos , Proteínas Nucleares/química , Unión Proteica , Estructura Terciaria de Proteína , Ribonucleoproteínas/química , Empalmosomas/metabolismo , Factor de Empalme U2AF
14.
Nat Struct Mol Biol ; 21(11): 997-1005, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25326705

RESUMEN

The U2AF heterodimer has been well studied for its role in defining functional 3' splice sites in pre-mRNA splicing, but many fundamental questions still remain unaddressed regarding the function of U2AF in mammalian genomes. Through genome-wide analysis of U2AF-RNA interactions, we report that U2AF has the capacity to directly define ~88% of functional 3' splice sites in the human genome, but numerous U2AF binding events also occur in intronic locations. Mechanistic dissection reveals that upstream intronic binding events interfere with the immediate downstream 3' splice site associated either with the alternative exon, to cause exon skipping, or with the competing constitutive exon, to induce exon inclusion. We further demonstrate partial functional impairment with leukemia-associated mutations in U2AF35, but not U2AF65, in regulated splicing. These findings reveal the genomic function and regulatory mechanism of U2AF in both normal and disease states.


Asunto(s)
Empalme Alternativo , Genoma Humano , Proteínas Nucleares/metabolismo , Sitios de Empalme de ARN , Ribonucleoproteínas/metabolismo , Secuencia de Bases , Sitios de Unión , Exones , Células HeLa , Humanos , Intrones , Datos de Secuencia Molecular , Mutación , Proteínas Nucleares/genética , Unión Proteica , Ribonucleoproteínas/genética , Factor de Empalme U2AF
15.
Hum Mol Genet ; 23(1): 209-25, 2014 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-23975679

RESUMEN

Recent human genetic studies have provided evidences that sporadic or inherited missense mutations in four-and-a-half LIM domain protein 1 (FHL1), resulting in alterations in FHL1 protein expression, are associated with rare congenital myopathies, including reducing body myopathy and Emery-Dreifuss muscular dystrophy. However, it remains to be clarified whether mutations in FHL1 cause skeletal muscle remodeling owing to gain- or loss of FHL1 function. In this study, we used FHL1-null mice lacking global FHL1 expression to evaluate loss-of-function effects on skeletal muscle homeostasis. Histological and functional analyses of soleus, tibialis anterior and sternohyoideus muscles demonstrated that FHL1-null mice develop an age-dependent myopathy associated with myofibrillar and intermyofibrillar (mitochondrial and sarcoplasmic reticulum) disorganization, impaired muscle oxidative capacity and increased autophagic activity. A longitudinal study established decreased survival rates in FHL1-null mice, associated with age-dependent impairment of muscle contractile function and a significantly lower exercise capacity. Analysis of primary myoblasts isolated from FHL1-null muscles demonstrated early muscle fiber differentiation and maturation defects, which could be rescued by re-expression of the FHL1A isoform, highlighting that FHL1A is necessary for proper muscle fiber differentiation and maturation in vitro. Overall, our data show that loss of FHL1 function leads to myopathy in vivo and suggest that loss of function of FHL1 may be one of the mechanisms underlying muscle dystrophy in patients with FHL1 mutations.


Asunto(s)
Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas con Dominio LIM/genética , Proteínas con Dominio LIM/metabolismo , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Músculo Esquelético/patología , Distrofias Musculares/patología , Miofibrillas/patología , Factores de Edad , Animales , Diferenciación Celular , Femenino , Humanos , Masculino , Ratones , Ratones Transgénicos , Actividad Motora , Músculo Esquelético/metabolismo , Distrofias Musculares/genética , Distrofia Muscular de Emery-Dreifuss/patología , Mioblastos Esqueléticos/metabolismo , Mioblastos Esqueléticos/patología , Miofibrillas/metabolismo
16.
Mol Cell ; 36(6): 996-1006, 2009 Dec 25.
Artículo en Inglés | MEDLINE | ID: mdl-20064465

RESUMEN

Recent transcriptome analysis indicates that > 90% of human genes undergo alternative splicing, underscoring the contribution of differential RNA processing to diverse proteomes in higher eukaryotic cells. The polypyrimidine tract-binding protein PTB is a well-characterized splicing repressor, but PTB knockdown causes both exon inclusion and skipping. Genome-wide mapping of PTB-RNA interactions and construction of a functional RNA map now reveal that dominant PTB binding near a competing constitutive splice site generally induces exon inclusion, whereas prevalent binding close to an alternative site often causes exon skipping. This positional effect was further demonstrated by disrupting or creating a PTB-binding site on minigene constructs and testing their responses to PTB knockdown or overexpression. These findings suggest a mechanism for PTB to modulate splice site competition to produce opposite functional consequences, which may be generally applicable to RNA-binding splicing factors to positively or negatively regulate alternative splicing in mammalian cells.


Asunto(s)
Empalme Alternativo , Exones/genética , Genoma Humano , Proteína de Unión al Tracto de Polipirimidina/metabolismo , ARN , Animales , Sitios de Unión/genética , Perfilación de la Expresión Génica , Células HeLa , Humanos , Datos de Secuencia Molecular , Proteína de Unión al Tracto de Polipirimidina/genética , Unión Proteica , ARN/genética , ARN/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...