Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 124
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
J Biol Chem ; 291(42): 21913-21924, 2016 Oct 14.
Artículo en Inglés | MEDLINE | ID: mdl-27566547

RESUMEN

GRK2, a G protein-coupled receptor kinase, plays a critical role in cardiac physiology. Adrenergic receptors are the primary target for GRK2 activity in the heart; phosphorylation by GRK2 leads to desensitization of these receptors. As such, levels of GRK2 activity in the heart directly correlate with cardiac contractile function. Furthermore, increased expression of GRK2 after cardiac insult exacerbates injury and speeds progression to heart failure. Despite the importance of this kinase in both the physiology and pathophysiology of the heart, relatively little is known about the role of GRK2 in skeletal muscle function and disease. In this study we generated a novel skeletal muscle-specific GRK2 knock-out (KO) mouse (MLC-Cre:GRK2fl/fl) to gain a better understanding of the role of GRK2 in skeletal muscle physiology. In isolated muscle mechanics testing, GRK2 ablation caused a significant decrease in the specific force of contraction of the fast-twitch extensor digitorum longus muscle yet had no effect on the slow-twitch soleus muscle. Despite these effects in isolated muscle, exercise capacity was not altered in MLC-Cre:GRK2fl/fl mice compared with wild-type controls. Skeletal muscle hypertrophy stimulated by clenbuterol, a ß2-adrenergic receptor (ß2AR) agonist, was significantly enhanced in MLC-Cre:GRK2fl/fl mice; mechanistically, this seems to be due to increased clenbuterol-stimulated pro-hypertrophic Akt signaling in the GRK2 KO skeletal muscle. In summary, our study provides the first insights into the role of GRK2 in skeletal muscle physiology and points to a role for GRK2 as a modulator of contractile properties in skeletal muscle as well as ß2AR-induced hypertrophy.


Asunto(s)
Clenbuterol/efectos adversos , Quinasa 2 del Receptor Acoplado a Proteína-G/metabolismo , Contracción Muscular/efectos de los fármacos , Músculo Esquelético/enzimología , Enfermedades Musculares/enzimología , Transducción de Señal/efectos de los fármacos , Animales , Clenbuterol/farmacocinética , Quinasa 2 del Receptor Acoplado a Proteína-G/genética , Hipertrofia/inducido químicamente , Hipertrofia/enzimología , Hipertrofia/genética , Hipertrofia/patología , Ratones , Ratones Noqueados , Contracción Muscular/genética , Músculo Esquelético/patología , Enfermedades Musculares/inducido químicamente , Enfermedades Musculares/genética , Enfermedades Musculares/patología , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptores Adrenérgicos beta 2/genética , Receptores Adrenérgicos beta 2/metabolismo , Transducción de Señal/genética
2.
Am J Dermatopathol ; 39(10): 747-752, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28030367

RESUMEN

Port wine stain (PWS) is a congenital, progressive vascular malformation. Many patients with PWS develop hypertrophy and discrete nodularity during their adult life, but the mechanism(s) remain incompletely understood. In this study, we attempted to investigate activation status of PKCα, PI3K, PDPK1 and PLC-γ and protein levels of PP2A and DAG to explore their potential roles in the formation of hypertrophic and nodular PWS lesions. We found phosphorylated levels of PKCα, PI3K, PDPK1, and PLC-γ and protein levels of PP2A and DAG showed moderate increases in the endothelial cells of hypertrophic PWS as compared to the adjacent normal skin. These increases extended throughout the entire stroma of blood vessels in PWS nodules. Many proliferating cells, such as fibroblasts, also showed strong activation of PKCα, PI3K, PDPK1 and PLC-γ and upregulations of PP2A and DAG in nodular PWS lesions. Our data showed that there is aberrant activation of PKCα, PI3K, PDPK1 and PLC-γ and upregulation of PP2A and DAG mainly in endothelial cells in hypertrophic PWS areas, but presenting in the entire vasculatures and surrounding fibroblasts in PWS nodules. Our data suggest that both PKCα and PI3K signaling pathways contribute to the development of hypertrophy and nodularity in adult PWS.


Asunto(s)
Fosfatidilinositol 3-Quinasas/metabolismo , Mancha Vino de Oporto/enzimología , Mancha Vino de Oporto/patología , Proteína Quinasa C-alfa/metabolismo , Adulto , Femenino , Humanos , Hipertrofia/enzimología , Hipertrofia/patología , Masculino
3.
Proc Natl Acad Sci U S A ; 111(52): 18643-8, 2014 Dec 30.
Artículo en Inglés | MEDLINE | ID: mdl-25512490

RESUMEN

Without a fully developed initial segment, the most proximal region of the epididymis, male infertility results. Therefore, it is important to understand the development and regulation of this crucial region. In addition to distinctively high activity levels of the components of the ERK pathway, which are essential for initial-segment differentiation, the initial segment exhibits high protein and activity levels of phosphatase and tensin homolog (PTEN). To understand the role of PTEN in the regulation of the initial segment, we generated a mouse model with a conditional deletion of Pten from the epithelial cells of the proximal epididymis from postnatal day 17 (P17) onward. Shortly after Pten deletion, hypertrophy of the proximal epididymis became evident. Loss of Pten resulted in activation of the AKT (protein kinase B) pathway components from P28 onward, which in turn gradually suppressed RAF1 proto-oncogene serine/threonine kinase (RAF1)/ERK signaling through the interaction between AKT and RAF1. Consistent with progressive changes in RAF1/ERK signaling, loss of Pten progressively altered cell shape, size, organization, proliferation, and survival in the initial-segment epithelium and resulted in dedifferentiation and extensive epithelial folding. Most importantly, knockout males progressively lost fertility and became infertile from 6 to 12 mo. Spermatozoa from older knockout mice showed a lower percentage of motility and a higher percentage of flagellar angulation compared with controls, suggesting compromised sperm maturation. Therefore, under normal physiological conditions, PTEN suppresses AKT activity to maintain activation of the RAF1/ERK signaling pathway, which in turn maintains normal function of the initial segment and therefore, normal sperm maturation.


Asunto(s)
Epidídimo/enzimología , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Fertilidad/fisiología , Sistema de Señalización de MAP Quinasas/fisiología , Fosfohidrolasa PTEN/metabolismo , Quinasas raf/metabolismo , Animales , Proliferación Celular/genética , Forma de la Célula/genética , Quinasas MAP Reguladas por Señal Extracelular/genética , Eliminación de Gen , Hipertrofia/enzimología , Hipertrofia/genética , Hipertrofia/patología , Masculino , Ratones , Ratones Noqueados , Fosfohidrolasa PTEN/genética , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Quinasas raf/genética
4.
Biochim Biophys Acta ; 1842(7): 945-58, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24583340

RESUMEN

Podocytes are terminally differentiated cells of the glomerular filtration barrier that react with hypertrophy in the course of injury such as in membranous nephropathy (MGN). The neuronal deubiquitinase ubiquitin C-terminal hydrolase L1 (UCH-L1) is expressed and activated in podocytes of human and rodent MGN. UCH-L1 regulates the mono-ubiquitin pool and induces accumulation of poly-ubiquitinated proteins in affected podocytes. Here, we investigated the role of UCH-L1 in podocyte hypertrophy and in the homeostasis of the hypertrophy associated "model protein" p27(Kip1). A better understanding of the basic mechanisms leading to podocyte hypertrophy is crucial for the development of specific therapies in MGN. In human and rat MGN, hypertrophic podocytes exhibited a simultaneous up-regulation of UCH-L1 and of cytoplasmic p27(Kip1) content. Functionally, inhibition of UCH-L1 activity and knockdown or inhibition of UCH-L1 attenuated podocyte hypertrophy by decreasing the total protein content in isolated glomeruli and in cultured podocytes. In contrast, UCH-L1 levels and activity increased podocyte hypertrophy and total protein content in culture, specifically of cytoplasmic p27(Kip1). UCH-L1 enhanced cytoplasmic p27(Kip1) levels by nuclear export and decreased poly-ubiquitination and proteasomal degradation of p27(Kip1). In parallel, UCH-L1 increased podocyte turnover, migration and cytoskeletal rearrangement, which are associated with known oncogenic functions of cytoplasmic p27(Kip1) in cancer. We propose that UCH-L1 induces podocyte hypertrophy in MGN by increasing the total protein content through altered degradation and accumulation of proteins such as p27(Kip1) in the cytoplasm of podocytes. Modification of both UCH-L1 activity and levels could be a new therapeutic avenue to podocyte hypertrophy in MGN.


Asunto(s)
Hipertrofia/metabolismo , Enfermedades Renales/metabolismo , Podocitos/metabolismo , Ubiquitina Tiolesterasa/metabolismo , Animales , Células Cultivadas , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/genética , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , Citoplasma/enzimología , Citoplasma/genética , Citoplasma/metabolismo , Humanos , Hipertrofia/enzimología , Hipertrofia/genética , Enfermedades Renales/enzimología , Enfermedades Renales/genética , Masculino , Podocitos/enzimología , Complejo de la Endopetidasa Proteasomal/genética , Complejo de la Endopetidasa Proteasomal/metabolismo , Ratas , Ratas Sprague-Dawley , Ubiquitina Tiolesterasa/genética , Ubiquitinación , Regulación hacia Arriba/genética
5.
Brain ; 137(Pt 2): 520-36, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24271323

RESUMEN

The aim of this study was to elucidate the characteristics, pathogenesis and treatment strategy of hypertrophic pachymeningitis that is associated with myeloperoxidase anti-neutrophil cytoplasmic antibody (ANCA). We retrospectively investigated clinical, radiological, immunological and pathological profiles of 36 patients with immune-mediated or idiopathic hypertrophic pachymeningitis, including 17 patients with myeloperoxidase-ANCA, four patients with proteinase 3-ANCA, six patients with other immune-mediated disorders, and nine patients with 'idiopathic' variety. Myeloperoxidase-ANCA-positive hypertrophic pachymeningitis was characterized by: (i) an elderly female predominance; (ii) 82% of patients diagnosed with granulomatosis with polyangiitis (previously known as Wegener's granulomatosis) according to Watts' algorithm; (iii) a high frequency of patients with lesions limited to the dura mater and upper airways, developing headaches, chronic sinusitis, otitis media or mastoiditis; (iv) a low frequency of patients with the 'classical or generalized form' of granulomatosis with polyangiitis involving the entire upper and lower airways and kidney, or progressing to generalized disease, in contrast to proteinase 3-ANCA-positive hypertrophic pachymeningitis; (v) less severe neurological damage according to the modified Rankin Scale and low disease activity according to the Birmingham Vasculitis Activity Score compared with proteinase 3-ANCA-positive hypertrophic pachymeningitis; (vi) increased levels of CXCL10, CXCL8 and interleukin 6 in cerebrospinal fluids, and increased numbers of T cells, neutrophils, eosinophils, plasma cells and monocytes/macrophages in autopsied or biopsied dura mater with pachymeningitis, suggesting TH1-predominant granulomatous lesions in hypertrophic pachymeningitis, as previously reported in pulmonary or renal lesions of granulomatosis with polyangiitis; and (vii) greater efficacy of combination therapy with prednisolone and cyclophosphamide compared with monotherapy with prednisolone. Proteinase 3-ANCA may be considered a marker for more severe neurological damage, higher disease activity and a higher frequency of the generalized form compared with myeloperoxidase-ANCA-positive hypertrophic pachymeningitis. However, categorization into 'granulomatosis with polyangiitis' according to Watts' algorithm and immunological or pathological features were common in both proteinase 3- and myeloperoxidase-ANCA-positive hypertrophic pachymeningitis. These data indicate that most patients with myeloperoxidase-ANCA-positive hypertrophic pachymeningitis should be categorized as having the central nervous system-limited form of ANCA-associated vasculitis, consistent with the concept of ophthalmic-, pulmonary- or renal-limited vasculitis.


Asunto(s)
Anticuerpos Anticitoplasma de Neutrófilos/sangre , Anticuerpos Anticitoplasma de Neutrófilos/líquido cefalorraquídeo , Hipertrofia/sangre , Hipertrofia/líquido cefalorraquídeo , Meningitis/sangre , Meningitis/líquido cefalorraquídeo , Peroxidasa/sangre , Peroxidasa/líquido cefalorraquídeo , Vasculitis/enzimología , Adulto , Anciano , Biomarcadores/sangre , Biomarcadores/líquido cefalorraquídeo , Femenino , Humanos , Hipertrofia/enzimología , Masculino , Meningitis/enzimología , Persona de Mediana Edad , Estudios Retrospectivos , Vasculitis/sangre , Vasculitis/líquido cefalorraquídeo
6.
Zhongguo Zhong Yao Za Zhi ; 40(16): 3125-31, 2015 Aug.
Artículo en Zh | MEDLINE | ID: mdl-26790279

RESUMEN

Glomerular hypertrophy is the main pathological characteristic in the early stage of diabetic nephropathy (DN), and its regulatory mechanism is closely related to mammalian target of rapamycin (mTOR) signaling pathway activity. mTOR includes mTOR complex 1 (mTORC1) and mTOR complex 2(mTORC2), in which, the upstream pathway of mTORC1 is phosphatidylinositol-3-kinase (PI3K)/serine-threonine kinase(Akt)/adenosine monophosphate activated protein kinase(AMPK), and the representative signaling molecules in the downstream pathway of mTORC1 are 4E-binding proteins(4EBP) and phosphoprotein 70 S6Kinase(p70S6K). Some Chinese herbal extracts could improve cell proliferation via intervening the expressions of the key molecules in the upstream or downstream of PIK/Akt/mTOR signaling pathway in vivo. As for glomerular mesangial cells(MC) and podocyte, mTOR plays an important role in regulating glomerular inherent cells, including adjusting cell cycle, energy metabolism and matrix protein synthesis. Rapamycin, the inhibitor of mTOR, could suppress glomerular inherent cell hypertrophy, cell proliferation, glomerular basement membrane (GBM) thickening and mesangial matrix deposition in model rats with DN. Some Chinese herbal extracts could alleviate glomerular lesions by intervening mTOR signaling pathway activity in renal tissue of DN animal models or in renal inherent cells in vivo and in vitro.


Asunto(s)
Nefropatías Diabéticas/tratamiento farmacológico , Nefropatías Diabéticas/enzimología , Medicamentos Herbarios Chinos/administración & dosificación , Serina-Treonina Quinasas TOR/metabolismo , Animales , Nefropatías Diabéticas/genética , Nefropatías Diabéticas/patología , Humanos , Hipertrofia/tratamiento farmacológico , Hipertrofia/enzimología , Hipertrofia/genética , Hipertrofia/patología , Glomérulos Renales/efectos de los fármacos , Glomérulos Renales/metabolismo , Glomérulos Renales/patología , Transducción de Señal/efectos de los fármacos , Serina-Treonina Quinasas TOR/genética
7.
J Biol Chem ; 288(45): 32119-32125, 2013 Nov 08.
Artículo en Inglés | MEDLINE | ID: mdl-24056368

RESUMEN

The exact molecular mechanisms governing articular chondrocytes remain unknown in skeletal biology. In this study, we have found that ESET (an ERG-associated protein with a SET domain, also called SETDB1) histone methyltransferase is expressed in articular cartilage. To test whether ESET regulates articular chondrocytes, we carried out mesenchyme-specific deletion of the ESET gene in mice. ESET knock-out did not affect generation of articular chondrocytes during embryonic development. Two weeks after birth, there was minimal qualitative difference at the knee joints between wild-type and ESET knock-out animals. At 1 month, ectopic hypertrophy, proliferation, and apoptosis of articular chondrocytes were seen in the articular cartilage of ESET-null animals. At 3 months, additional signs of terminal differentiation such as increased alkaline phosphatase activity and an elevated level of matrix metalloproteinase (MMP)-13 were found in ESET-null cartilage. Staining for type II collagen and proteoglycan revealed that cartilage degeneration became progressively worse from 2 weeks to 12 months at the knee joints of ESET knock-out mutants. Analysis of over 14 pairs of age- and sex-matched wild-type and knock-out mice indicated that the articular chondrocyte phenotype in ESET-null mutants is 100% penetrant. Our results demonstrate that expression of ESET plays an essential role in the maintenance of articular cartilage by preventing articular chondrocytes from terminal differentiation and may have implications in joint diseases such as osteoarthritis.


Asunto(s)
Cartílago Articular/enzimología , Diferenciación Celular , Condrocitos/enzimología , N-Metiltransferasa de Histona-Lisina/metabolismo , Articulación de la Rodilla/enzimología , Osteoartritis de la Rodilla/enzimología , Fosfatasa Alcalina/genética , Fosfatasa Alcalina/metabolismo , Animales , Cartílago Articular/patología , Condrocitos/patología , Colágeno Tipo II/genética , Colágeno Tipo II/metabolismo , N-Metiltransferasa de Histona-Lisina/genética , Hipertrofia/enzimología , Hipertrofia/genética , Hipertrofia/patología , Articulación de la Rodilla/patología , Metaloproteinasa 13 de la Matriz/genética , Metaloproteinasa 13 de la Matriz/metabolismo , Ratones , Ratones Noqueados , Especificidad de Órganos/genética , Osteoartritis de la Rodilla/genética , Osteoartritis de la Rodilla/patología
8.
Am J Physiol Cell Physiol ; 304(1): C56-67, 2013 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-23076795

RESUMEN

Arachidonic acid (AA) is the metabolic precursor to a diverse range of downstream bioactive lipid mediators. A positive or negative influence of individual eicosanoid species [e.g., prostaglandins (PGs), leukotrienes, and hydroxyeicosatetraenoic acids] has been implicated in skeletal muscle cell growth and development. The collective role of AA-derived metabolites in physiological states of skeletal muscle growth/atrophy remains unclear. The present study aimed to determine the direct effect of free AA supplementation and subsequent eicosanoid biosynthesis on skeletal myocyte growth in vitro. C2C12 (mouse) skeletal myocytes induced to differentiate with supplemental AA exhibited dose-dependent increases in the size, myonuclear content, and protein accretion of developing myotubes, independent of changes in cell density or the rate/extent of myogenic differentiation. Nonselective (indomethacin) or cyclooxygenase 2 (COX-2)-selective (NS-398) nonsteroidal anti-inflammatory drugs blunted basal myogenesis, an effect that was amplified in the presence of supplemental free AA substrate. The stimulatory effects of AA persisted in preexisting myotubes via a COX-2-dependent (NS-389-sensitive) pathway, specifically implying dependency on downstream PG biosynthesis. AA-stimulated growth was associated with markedly increased secretion of PGF(2α) and PGE(2); however, incubation of myocytes with PG-rich conditioned medium failed to mimic the effects of direct AA supplementation. In vitro AA supplementation stimulates PG release and skeletal muscle cell hypertrophy via a COX-2-dependent pathway.


Asunto(s)
Ácido Araquidónico/fisiología , Ciclooxigenasa 2/fisiología , Fibras Musculares Esqueléticas/metabolismo , Transducción de Señal/fisiología , Animales , Diferenciación Celular/fisiología , Aumento de la Célula , Supervivencia Celular/fisiología , Células Cultivadas , Dinoprost/metabolismo , Dinoprost/fisiología , Dinoprostona/metabolismo , Dinoprostona/fisiología , Hipertrofia/enzimología , Hipertrofia/metabolismo , Hipertrofia/patología , Ratones , Fibras Musculares Esqueléticas/citología , Fibras Musculares Esqueléticas/patología
9.
Tohoku J Exp Med ; 231(1): 45-56, 2013 09.
Artículo en Inglés | MEDLINE | ID: mdl-24042457

RESUMEN

Glomerular epithelial cells (GECs) are known to play a key role in maintaining the structure and function of the glomerulus. GEC injury induced by hyperglycemia is present in early-stage diabetic nephropathy (DN), which is the most common cause of renal failure. In an attempt to identify target proteins involved in the pathogenesis of GEC injury at early DN, we performed the proteomic analysis using primary cultures of GECs, prepared from the dissected rat glomeruli. The protein expression profiles in the two-dimensional electrophoresis gels were compared between GECs treated for three days with normal glucose (5 mM) and those with high glucose (30 mM) concentrations. These concentrations correspond to blood glucose concentrations under normoglycemia and hyperglycemia, respectively. Proteins with differential expression levels were identified using ESI-Q-TOF tandem mass spectrometry. The primary GECs cultured in hyperglycemic conditions showed cellular hypertrophy and increased production of reactive oxygen species, both of which reflect the GEC injury. Our proteomic analysis identified eight proteins with differential expression profiles, depending on glucose concentrations. Among them, we selected ATP synthase ß subunit and enolase 2 that are related to energy metabolism and are down-regulated under hyperglycemia, and confirmed that hyperglycemia decreased the expression levels of ATP synthase ß subunit and enolase 2 proteins by western blotting analysis. Hyperglycemia may impair mitochondrial function and alter glycolysis in GECs by down-regulating the expression of ATP synthase ß subunit and enolase 2. The present study may provide a better understanding of the pathogenic mechanisms of GEC injury in early DN.


Asunto(s)
Células Epiteliales/enzimología , Hiperglucemia/enzimología , Glomérulos Renales/patología , ATPasas de Translocación de Protón Mitocondriales/metabolismo , Fosfopiruvato Hidratasa/metabolismo , Animales , Western Blotting , Células Cultivadas , Electroforesis en Gel Bidimensional , Células Epiteliales/efectos de los fármacos , Glucosa/farmacología , Hiperglucemia/patología , Hipertrofia/enzimología , Hipertrofia/patología , Masculino , Proteómica , Ratas , Ratas Sprague-Dawley , Especies Reactivas de Oxígeno/metabolismo , Espectrometría de Masa por Ionización de Electrospray
10.
EMBO J ; 27(8): 1266-76, 2008 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-18354498

RESUMEN

In response to cancer, AIDS, sepsis and other systemic diseases inducing muscle atrophy, the E3 ubiquitin ligase Atrogin1/MAFbx (MAFbx) is dramatically upregulated and this response is necessary for rapid atrophy. However, the precise function of MAFbx in muscle wasting has been questioned. Here, we present evidence that during muscle atrophy MAFbx targets the eukaryotic initiation factor 3 subunit 5 (eIF3-f) for ubiquitination and degradation by the proteasome. Ectopic expression of MAFbx in myotubes induces atrophy and degradation of eIF3-f. Conversely, blockade of MAFbx expression by small hairpin RNA interference prevents eIF3-f degradation in myotubes undergoing atrophy. Furthermore, genetic activation of eIF3-f is sufficient to cause hypertrophy and to block atrophy in myotubes, whereas genetic blockade of eIF3-f expression induces atrophy in myotubes. Finally, eIF3-f induces increasing expression of muscle structural proteins and hypertrophy in both myotubes and mouse skeletal muscle. We conclude that eIF3-f is a key target that accounts for MAFbx function during muscle atrophy and has a major role in skeletal muscle hypertrophy. Thus, eIF3-f seems to be an attractive therapeutic target.


Asunto(s)
Factor 3 de Iniciación Eucariótica/metabolismo , Proteínas Musculares/fisiología , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Atrofia Muscular/metabolismo , Atrofia Muscular/patología , Proteínas Ligasas SKP Cullina F-box/fisiología , Animales , Línea Celular , Modelos Animales de Enfermedad , Femenino , Humanos , Hipertrofia/enzimología , Hipertrofia/metabolismo , Ratones , Ratones Endogámicos C57BL , Fibras Musculares Esqueléticas/enzimología , Fibras Musculares Esqueléticas/metabolismo , Fibras Musculares Esqueléticas/patología , Músculo Esquelético/enzimología , Atrofia Muscular/enzimología , Complejo de la Endopetidasa Proteasomal/metabolismo , Complejo de la Endopetidasa Proteasomal/fisiología , Mapeo de Interacción de Proteínas , Ubiquitinación
11.
BJU Int ; 109(1): 132-40, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21883831

RESUMEN

OBJECTIVE: To study muscarinic/purinergic receptor activation and Rho-kinase/protein kinase C (PKC) signalling during smooth muscle contraction in normal and hypertrophic mouse urinary bladders. METHODS: Partial urinary outflow obstruction was induced in adult female (10-12 weeks) C57Bl/6 mice and comparisons were made with sham-operated controls. Bladder preparations were examined in vitro. Expression of signalling proteins was examined using Western blot analysis. RESULTS: Obstructed bladders increased more than threefold in weight and were found to have enhanced muscarinic and attenuated purinergic components during nerve-induced contractions. The contractile response to carbachol was shifted towards lower concentrations of carbachol for the peak response and had a markedly enhanced sustained component. The amplitude of the α,ß-methylene ATP-induced responses was lowered. Rho-kinase inhibitor Y27632 (10 µM) inhibited peak and sustained contractile responses to carbachol in control bladders (peak by 38%; plateau 57%) and obstructed bladders (peak 37% plateau 47%). PKC inhibitor GF109203X (1 µM) inhibited carbachol contractions in controls (peak by 29%; plateau 29%) and obstructed bladders (peak 17%; plateau 12%). Inhibition by a similar extent was observed after nerve stimulation. Sensitivity to Ca(2+) in high-K(+) depolarized intact tissues increased in obstructed bladders. This increased receptor-independent Ca(2+)-sensitivity was abolished by Y27632. Tissue contents of the myosin-binding phosphatase subunit MYPT-1 and catalytic phosphatase subunit PP1ß, were decreased and the contents of RhoGDI, RhoA and CPI-17 increased. A decrease in the Rho-kinase isoform ROCK-1 was observed. CONCLUSION: Based on these results, one can speculate that Rho-kinase inhibition would preferentially target the pathological phasic activity in the urinary bladder rather than inhibit the physiological receptor-mediated bladder emptying.


Asunto(s)
Contracción Muscular/fisiología , Proteína Quinasa C/metabolismo , Obstrucción del Cuello de la Vejiga Urinaria/enzimología , Vejiga Urinaria/fisiopatología , Quinasas Asociadas a rho/metabolismo , Animales , Western Blotting , Modelos Animales de Enfermedad , Femenino , Hipertrofia/enzimología , Hipertrofia/patología , Ratones , Ratones Endogámicos C57BL , Vejiga Urinaria/enzimología , Vejiga Urinaria/patología , Obstrucción del Cuello de la Vejiga Urinaria/patología , Obstrucción del Cuello de la Vejiga Urinaria/fisiopatología
12.
Curr Hypertens Rep ; 14(1): 88-96, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22081429

RESUMEN

One of the major challenges for hypertension research is to identify the mechanisms that cause the comorbidities encountered in many hypertensive patients, as seen in the metabolic syndrome. An emerging body of evidence suggests that human and experimental hypertensives may exhibit uncontrolled activity of proteinases, including the family of matrix metalloproteinases, recognized for their ability to restructure the extracellular matrix proteins and to play a role in hypertrophy. We propose a new hypothesis that provides a molecular framework for the comorbidities of hypertension, diabetes, capillary rarefaction, immune suppression, and other cell and organ dysfunctions due to early and uncontrolled extracellular receptor cleavage by active proteinases. The proteinase and signaling activity in hypertensives requires further detailed analysis of the proteinase expression, the mechanisms causing proenzyme activation, and identification of the proteinase substrate. This work may open the opportunity for reassessment of old interventions and development of new interventions to manage hypertension and its comorbidities.


Asunto(s)
Proteínas de la Matriz Extracelular/metabolismo , Hipertensión/enzimología , Metaloproteinasas de la Matriz/metabolismo , Síndrome Metabólico/enzimología , Receptores de Superficie Celular/metabolismo , Transducción de Señal/fisiología , Comorbilidad , Humanos , Hipertensión/complicaciones , Hipertensión/fisiopatología , Hipertrofia/enzimología , Tolerancia Inmunológica/fisiología , Síndrome Metabólico/complicaciones , Síndrome Metabólico/fisiopatología , Microcirculación/fisiología , Modelos Biológicos , Sistema Renina-Angiotensina/fisiología , Vasoconstricción/fisiología
13.
Proc Natl Acad Sci U S A ; 106(18): 7548-52, 2009 May 05.
Artículo en Inglés | MEDLINE | ID: mdl-19376970

RESUMEN

Nonsteroidal anti-inflammatory drugs selective for inhibition of COX-2 increase heart failure and elevate blood pressure. The COX-2 gene was floxed and crossed into merCremer mice under the alpha-myosin heavy-chain promoter. Tamoxifen induced selective deletion of COX-2 in cardiomyocytes depressed cardiac output, and resulted in weight loss, diminished exercise tolerance, and enhanced susceptibility to induced arrhythmogenesis. The cardiac dysfunction subsequent to pressure overload recovered progressively in the knockouts coincident with increasing cardiomyocyte hypertrophy and interstitial and perivascular fibrosis. Inhibition of COX-2 in cardiomyocytes may contribute to heart failure in patients receiving nonsteroidal anti-inflammatory drugs specific for inhibition of COX-2.


Asunto(s)
Ciclooxigenasa 2/fisiología , Frecuencia Cardíaca , Miocitos Cardíacos/patología , Miocitos Cardíacos/fisiología , Animales , Antiinflamatorios no Esteroideos/efectos adversos , Ciclooxigenasa 2/genética , Eliminación de Gen , Frecuencia Cardíaca/genética , Hipertrofia/inducido químicamente , Hipertrofia/enzimología , Ratones , Ratones Noqueados , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/enzimología
14.
J Vasc Surg Venous Lymphat Disord ; 10(2): 527-538.e2, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34358672

RESUMEN

BACKGROUND: PIK3CA (activating mutations of the p110α subunit of phosphatidylinositol 3-kinases)-related overgrowth spectrums (PROS) include a variety of clinical presentations that are associated with hypertrophy of different parts of the body. We performed a systematic literature review to assess the current treatment options and their efficacy and safety for PROS. METHODS: A literature search was performed in Embase, MEDLINE (Ovid), Web of Science Core Collection, Cochrane Central Register of Controlled Trials, ClinicalTrials.gov, and Google Scholar to retrieve studies on the treatment of hypertrophy in PROS. Randomized controlled trials, cohort studies, and case series with ≥10 patients were included in the present review. The titles, abstracts, and full text were assessed by two reviewers independently. The risk of bias was assessed using the Newcastle-Ottawa scale. RESULTS: We included 16 studies of the treatment of hypertrophy in PROS patients, 13 (81.3%) from clinical retrospective studies and 3 (13.7%) from prospective cohort studies. The risk of bias grade was low for 2, medium for 12, and high for 2 studies. Of the 16 studies, 13 reported on surgical treatment and 3 reported pharmacologic treatment using phosphatidylinositol-3-kinase (PI3K)/mammalian target of rapamycin (mTOR) pathway inhibitors in PROS patients. In 3 studies, PROS was defined by a mutation in the PIK3CA gene, and 13 studies relied on a clinical definition of PROS. Surgical therapy was beneficial for a specific subgroup of PROS (macrodactyly). However, little has been reported concerning surgery and the potential benefits for other PROS entities. The reported side effects after surgical therapy were mostly prolonged wound healing or scarring. PI3K/mTOR pathway inhibition was beneficial in patients with PROS by reducing hypertrophy and systemic symptoms. The adverse effects reported included infection, changes in blood count, liver enzymes, and metabolic measures. CONCLUSIONS: Surgery is a locally limited treatment option for specific types of PROS. A promising treatment option for PROS is pharmacologic PIK3CA inhibition. However, the level of evidence on the treatment of overgrowth in PROS patients is limited.


Asunto(s)
Fosfatidilinositol 3-Quinasa Clase I/antagonistas & inhibidores , Hipertrofia/terapia , Inhibidores mTOR/uso terapéutico , Inhibidores de las Quinasa Fosfoinosítidos-3/uso terapéutico , Procedimientos Quirúrgicos Operativos , Fosfatidilinositol 3-Quinasa Clase I/genética , Fosfatidilinositol 3-Quinasa Clase I/metabolismo , Predisposición Genética a la Enfermedad , Humanos , Hipertrofia/diagnóstico , Hipertrofia/enzimología , Hipertrofia/genética , Inhibidores mTOR/efectos adversos , Terapia Molecular Dirigida , Mutación , Fenotipo , Inhibidores de las Quinasa Fosfoinosítidos-3/efectos adversos , Transducción de Señal , Procedimientos Quirúrgicos Operativos/efectos adversos , Síndrome , Resultado del Tratamiento
15.
Mol Pharmacol ; 79(3): 381-8, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21127130

RESUMEN

Cardiac-directed expression of AC6 has pronounced favorable effects on cardiac function possibly not linked with cAMP production. To determine rigorously whether cAMP generation is required for the beneficial effects of increased AC6 expression, we generated a catalytically inactive AC6 mutant (AC6mut) that has markedly diminished cAMP generating capacity by replacing aspartic acid with alanine at position 426 in the C1 domain (catalytic region) of AC6. Gene transfer of AC6 or AC6mut (adenovirus-mediated) in adult rat cardiac myocytes resulted in similar expression levels and intracellular distribution, but AC6mut expression was associated with marked reduction in cAMP production. Despite marked reduction in cAMP generation, AC6mut influenced intracellular signaling events similarly to that observed after expression of catalytically intact AC6. For example, both AC6 and AC6mut reduced phenylephrine-induced cardiac myocyte hypertrophy and apoptosis (p < 0.001), expression of cardiac ankyrin repeat protein (p < 0.01), and phospholamban (p < 0.05). AC6mut expression, similar to its catalytically intact cohort, was associated with increased Ca2+ transients in cardiac myocytes after isoproterenol stimulation. Many of the biological effects of AC6 expression are replicated by a catalytically inactive AC6 mutant, indicating that the mechanisms for these effects do not require increased cAMP generation.


Asunto(s)
Adenilil Ciclasas/fisiología , Miocitos Cardíacos/enzimología , Mutación Puntual/fisiología , Adenilil Ciclasas/biosíntesis , Adenilil Ciclasas/genética , Adenilil Ciclasas/metabolismo , Animales , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Calcineurina/metabolismo , Calcio/metabolismo , Calcio/fisiología , Proteínas de Unión al Calcio/biosíntesis , Proteínas de Unión al Calcio/metabolismo , AMP Cíclico/fisiología , Regulación de la Expresión Génica/efectos de los fármacos , Hipertrofia/enzimología , Hipertrofia/fisiopatología , Proteínas Musculares , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/fisiología , Proteínas Nucleares/metabolismo , Fenilefrina/farmacología , Fosforilación , Mutación Puntual/genética , Ratas , Ratas Sprague-Dawley , Proteínas Represoras/metabolismo , Troponina I/biosíntesis , Troponina I/metabolismo
16.
J Biol Chem ; 285(38): 29336-47, 2010 Sep 17.
Artículo en Inglés | MEDLINE | ID: mdl-20525681

RESUMEN

Airway smooth muscle hypertrophy is one of the hallmarks of airway remodeling in severe asthma. Several human diseases have been now associated with dysregulated microRNA (miRNA) expression. miRNAs are a class of small non-coding RNAs, which negatively regulate gene expression at the post-transcriptional level. Here, we identify miR-26a as a hypertrophic miRNA of human airway smooth muscle cells (HASMCs). We show that stretch selectively induces the transcription of miR-26a located in the locus 3p21.3 of human chromosome 3. The transcription factor CCAAT enhancer-binding protein α (C/EBPα) directly activates miR-26a expression through the transcriptional machinery upon stretch. Furthermore, stretch or enforced expression of miR-26a induces HASMC hypertrophy, and miR-26 knockdown reverses this effect, suggesting that miR-26a is a hypertrophic gene. We identify glycogen synthase kinase-3ß (GSK-3ß), an anti-hypertrophic protein, as a target gene of miR-26a. Luciferase reporter assays demonstrate that miR-26a directly interact with the 3'-untranslated repeat of the GSK-3ß mRNA. Stretch or enforced expression of miR-26a attenuates the endogenous GSK-3ß protein levels followed by the induction of HASMC hypertrophy. miR-26 knockdown reverses this effect, suggesting that miR-26a-induced hypertrophy occurs via its target gene GSK-3ß. Overall, as a first time, our study unveils that miR-26a is a mechanosensitive gene, and it plays an important role in the regulation of HASMC hypertrophy.


Asunto(s)
Glucógeno Sintasa Quinasa 3/metabolismo , Hipertrofia/enzimología , Hipertrofia/metabolismo , MicroARNs/fisiología , Músculo Liso/metabolismo , Sistema Respiratorio/metabolismo , Estrés Mecánico , Regiones no Traducidas 3'/genética , Regiones no Traducidas 3'/fisiología , Western Blotting , Células Cultivadas , Inmunoprecipitación de Cromatina , Glucógeno Sintasa Quinasa 3/genética , Glucógeno Sintasa Quinasa 3 beta , Humanos , Hipertrofia/genética , MicroARNs/genética , Músculo Liso/enzimología , Análisis de Secuencia por Matrices de Oligonucleótidos , Reacción en Cadena de la Polimerasa , Sistema Respiratorio/enzimología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
17.
Proc Natl Acad Sci U S A ; 105(34): 12457-62, 2008 Aug 26.
Artículo en Inglés | MEDLINE | ID: mdl-18711143

RESUMEN

G protein-coupled receptor (GPCR) kinases (GRKs) are critical regulators of cellular signaling and function. In cardiomyocytes, GRK2 and GRK5 are two GRKs important for myocardial regulation, and both have been shown to be up-regulated in the dysfunctional heart. We report that increased levels and activity of GRK5 in failing myocardium may have unique significance due to its nuclear localization, a property not shared by GRK2. We find that transgenic mice with elevated cardiac GRK5 levels have exaggerated hypertrophy and early heart failure compared with control mice after pressure overload. This pathology is not present in cardiac GRK2-overexpressing mice or in mice with overexpression of a mutant GRK5 that is excluded from the nucleus. Nuclear accumulation of GRK5 is enhanced in myocytes after aortic banding in vivo and in vitro in myocytes after increased G alpha q activity, the trigger for pressure-overload hypertrophy. GRK5 enhances activation of MEF2 in concert with Gq signals, demonstrating that nuclear localized GRK5 regulates gene transcription via a pathway critically linked to myocardial hypertrophy. Mechanistically, we show that this is due to GRK5 acting, in a non-GPCR manner, as a class II histone deacetylase (HDAC) kinase because it can associate with and phosphorylate the myocyte enhancer factor-2 repressor, HDAC5. Moreover, significant HDAC activity can be found with GRK5 in the heart. Our data show that GRK5 is a nuclear HDAC kinase that plays a key role in maladaptive cardiac hypertrophy apparently independent of any action directly on GPCRs.


Asunto(s)
Núcleo Celular/enzimología , Quinasa 5 del Receptor Acoplado a Proteína-G/fisiología , Miocitos Cardíacos/enzimología , Animales , Quinasa 5 del Receptor Acoplado a Proteína-G/análisis , Quinasa 5 del Receptor Acoplado a Proteína-G/genética , Quinasa 5 del Receptor Acoplado a Proteína-G/metabolismo , Insuficiencia Cardíaca/enzimología , Insuficiencia Cardíaca/etiología , Histona Desacetilasas/metabolismo , Hipertrofia/enzimología , Hipertrofia/etiología , Factores de Transcripción MEF2 , Ratones , Ratones Transgénicos , Miocitos Cardíacos/ultraestructura , Factores Reguladores Miogénicos/metabolismo , Regulación hacia Arriba
19.
J Cell Biol ; 151(7): 1537-48, 2000 Dec 25.
Artículo en Inglés | MEDLINE | ID: mdl-11134081

RESUMEN

Ras is a universal eukaryotic intracellular protein integrating extracellular signals from multiple receptor types. To investigate its role in the adult central nervous system, constitutively activated V12-Ha-Ras was expressed selectively in neurons of transgenic mice via a synapsin promoter. Ras-transgene protein expression increased postnatally, reaching a four- to fivefold elevation at day 40 and persisting at this level, thereafter. Neuronal Ras was constitutively active and a corresponding activating phosphorylation of mitogen-activated kinase was observed, but there were no changes in the activity of phosphoinositide 3-kinase, the phosphorylation of its target kinase Akt/PKB, or expression of the anti-apoptotic proteins Bcl-2 or Bcl-X(L). Neuronal Ras activation did not alter the total number of neurons, but induced cell soma hypertrophy, which resulted in a 14.5% increase of total brain volume. Choline acetyltransferase and tyrosine hydroxylase activities were increased, as well as neuropeptide Y expression. Degeneration of motorneurons was completely prevented after facial nerve lesion in Ras-transgenic mice. Furthermore, neurotoxin-induced degeneration of dopaminergic substantia nigra neurons and their striatal projections was greatly attenuated. Thus, the Ras signaling pathway mimics neurotrophic effects and triggers neuroprotective mechanisms in adult mice. Neuronal Ras activation might become a tool to stabilize donor neurons for neural transplantation and to protect neuronal populations in neurodegenerative diseases.


Asunto(s)
Hipertrofia/patología , Neuronas Motoras/metabolismo , Neuronas Motoras/patología , Enfermedades Neurodegenerativas/patología , Fármacos Neuroprotectores/metabolismo , Proteínas Serina-Treonina Quinasas , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , 1-Metil-4-fenil-1,2,3,6-Tetrahidropiridina/farmacología , 1-Metil-4-fenilpiridinio/farmacología , Animales , Axotomía , Encéfalo/efectos de los fármacos , Encéfalo/enzimología , Encéfalo/patología , Recuento de Células , Tamaño de la Célula , Células Cultivadas , Colina O-Acetiltransferasa/metabolismo , Activación Enzimática , Hipertrofia/enzimología , Ratones , Ratones Transgénicos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Neuronas Motoras/efectos de los fármacos , Neuronas Motoras/enzimología , Mutación/genética , Enfermedades Neurodegenerativas/enzimología , Neuropéptido Y/genética , Neuropéptido Y/metabolismo , Oxidopamina/farmacología , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-akt , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Proteínas Proto-Oncogénicas p21(ras)/genética , ARN Mensajero/análisis , ARN Mensajero/genética , Transducción de Señal/efectos de los fármacos , Sustancia Negra/efectos de los fármacos , Sustancia Negra/patología , Tirosina 3-Monooxigenasa/metabolismo
20.
Acta Neurochir (Wien) ; 151(9): 1127-34, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19444374

RESUMEN

BACKGROUND: Although there are still some unresolved aspects, current research has revealed that vascular cell proliferation probably plays an important part in the pathological formation process of cerebral vasospasm. Using a "two-hemorrhage" model of subarachnoid hemorrhage (SAH), this study investigated the function of ERK1/2 and vascular wall cell proliferation in pathological development of cerebral vasospasm. METHODS: Fifty rabbits were randomly divided into five groups: (1) SAH day 1, (2) SAH day 3, (3) SAH day 7, (4) SAH + DMSO (dimethyl sufoxide) solution, (5) SAH + PD98059 (a mitogen-activated protein kinase inhibitor) dissolved in DMSO solution. In the SAH + PD98059/DMSO group and SAH + DMSO control group, PD98059 in DMSO (2 mmol/l) or an equal quantity of DMSO, respectively, was injected into the cisterna magna, once a day from SAH day 1 to day 3. Western protein blotting was used to detect the expression of proliferating cell nuclear antigen (PCNA) and extracellular signal-regulated protein kinases 1 and 2 (ERK1/2) in each group's basilar arteries. Light microscopy and electron microscopy were used for dynamic histological detection at each observation point of the SAH vascular wall under the effects of SAH and the mitogen-activated protein kinase inhibitor. Another 18 rabbits were randomly divided into three groups: SAH, SAH + DMSO and SAH + PD98059/DMSO; cerebral angiograpathy was conducted on SAH days 1 and 7, and the progression of angiographic vasospasm evaluated. RESULTS: Compared with the control group, the extent of vasospasm after SAH increased with time. PD98059 significantly reduced angiographic and morphological vasospasm. In cerebral vasospasm, the expression of T-ERK1/2 showed no significant change. However, expression of p-ERK1/2 and PCNA began to increase significantly on day 3, and achieved a peak on day 7. PD98059 significantly inhibited the expression of p-ERK1/2 and PCNA (p < 0.05). CONCLUSIONS: Cell proliferation on the vascular wall plays an important part in the pathological formation process of cerebral vasospasm. ERK1/2 phosphorylation, as an important signaling pathway, taking part in the process of vascular-wall pathological proliferation of cerebral vasospasm.


Asunto(s)
Proliferación Celular , Hipertrofia/enzimología , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Músculo Liso Vascular/enzimología , Hemorragia Subaracnoidea/complicaciones , Vasoespasmo Intracraneal/enzimología , Animales , Arteria Basilar/citología , Arteria Basilar/enzimología , Modelos Animales de Enfermedad , Activación Enzimática/fisiología , Inhibidores Enzimáticos/farmacología , Flavonoides/farmacología , Hipertrofia/tratamiento farmacológico , Hipertrofia/fisiopatología , Microscopía Electrónica , Músculo Liso Vascular/citología , Miocitos del Músculo Liso/citología , Miocitos del Músculo Liso/enzimología , Antígeno Nuclear de Célula en Proliferación , Conejos , Regulación hacia Arriba/fisiología , Vasoespasmo Intracraneal/tratamiento farmacológico , Vasoespasmo Intracraneal/fisiopatología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA