Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
J Clin Endocrinol Metab ; 108(10): 2500-2509, 2023 09 18.
Artículo en Inglés | MEDLINE | ID: mdl-37022983

RESUMEN

CONTEXT: Glycemic variation had been demonstrated to be associated with several complications of diabetes. OBJECTIVE: Investigation of the association between visit to visit hemoglobin A1c (HbA1c) variation and the long-term risk of major adverse limb events (MALEs). METHODS: Retrospective database study. Average real variability was used to represent glycemic variations with all the HbA1c measurements during the 4 following years after the initial diagnosis of type 2 diabetes. Participants were followed from the beginning of the fifth year until death or the end of the follow-up. The association between HbA1c variations and MALEs was evaluated after adjusting for mean HbA1c and baseline characteristics. Included were 56 872 patients at the referral center with a first diagnosis of type 2 diabetes, no lower extremity arterial disease, and at least 1 HbA1c measurement in each of the 4 following years were identified from a multicenter database. The main outcome measure was incidence of a MALE, which was defined as the composite of revascularization, foot ulcers, and lower limb amputations. RESULTS: The average number of HbA1c measurements was 12.6. The mean follow-up time was 6.1 years. The cumulative incidence of MALEs was 9.25 per 1000 person-years. Visit to visit HbA1c variations were significantly associated with MALEs and lower limb amputation after multivariate adjustment. People in the highest quartile of variations had increased risks for MALEs (HR 1.25, 95% CI 1.10-1.41) and lower limb amputation (HR 3.05, 95% CI 1.97-4.74). CONCLUSION: HbA1c variation was independently associated with a long-term risk of MALEs and lower limb amputations in patients with type 2 diabetes.


Asunto(s)
Diabetes Mellitus Tipo 2 , Masculino , Humanos , Diabetes Mellitus Tipo 2/complicaciones , Diabetes Mellitus Tipo 2/epidemiología , Diabetes Mellitus Tipo 2/diagnóstico , Hemoglobina Glucada , Factores de Riesgo , Estudios Retrospectivos , Glucemia , Extremidad Inferior/cirugía
2.
Cancer Med ; 12(3): 2958-2969, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36069126

RESUMEN

BACKGROUND: Acute myocardial infarction (AMI) and colon cancer share similar risk factors. Studies have suggested an association between AMI and colon cancer; however, evidence is conflicting. Whether sex disparities exist in this association in the real world remains unknown. METHODS: In this population-based retrospective cohort study, 94,780 and 97,987 male patients and 38,697 and 72,007 female patients with and without new-onset AMI, respectively, from January 1, 2001, to December 31, 2012, were enrolled from Taiwan's National Health Insurance Research Database. Inverse probability of treatment weighting (IPTW) was used to balance covariates across study groups. The primary outcome was a new diagnosis of colon cancer. RESULTS: The incidence rate of colon cancer was 1.54 (95% confidence interval [CI] = 1.46-1.62) and 1.40 (95% CI = 1.32-1.48) per 1000 person-years in the male patients and 1.62 (95% CI = 1.50-1.74) and 1.22 (95% CI = 1.13-1.32) in the female patients, in the AMI and non-AMI groups, respectively. AMI was associated with a significantly higher risk of colon cancer in the female patients (hazard ratio [HR] = 1.31, 95% CI = 1.06-1.61) but not in the male patients (HR = 1.09, 95% CI = 0.95-1.26). In the subgroup analysis, the association between AMI and colon cancer in the female patients was stronger in those aged ≥65 years (HR = 1.28, 95% CI = 1.13-1.44). CONCLUSIONS: An increased risk of colon cancer was observed only in the female patients with AMI. The association between AMI and colon cancer in the female patients was the most evident in those aged ≥65 years.


Asunto(s)
Neoplasias del Colon , Infarto del Miocardio , Humanos , Masculino , Femenino , Estudios Retrospectivos , Factores de Riesgo , Neoplasias del Colon/complicaciones , Incidencia
3.
Commun Biol ; 5(1): 364, 2022 04 19.
Artículo en Inglés | MEDLINE | ID: mdl-35440618

RESUMEN

Vascular calcification occurs in arterial aging, atherosclerosis, diabetes mellitus, and chronic kidney disease. Transforming growth factor-ß1 (TGF-ß1) is a key modulator driving the osteogenic transdifferentiation of vascular smooth muscle cells (VSMCs), leading to vascular calcification. We hypothesize that milk fat globule-epidermal growth factor 8 (MFG-E8), a glycoprotein expressed in VSMCs, promotes the osteogenic transdifferentiation of VSMCs through the activation of TGF-ß1-mediated signaling. We observe that the genetic deletion of MFG-E8 prevents calcium chloride-induced vascular calcification in common carotid arteries (CCAs). The exogenous application of MFG-E8 to aged CCAs promotes arterial wall calcification. MFG-E8-deficient cultured VSMCs exhibit decreased biomineralization and phenotypic transformation to osteoblast-like cells in response to osteogenic medium. MFG-E8 promotes ß1 integrin-dependent MMP2 expression, causing TGF-ß1 activation and subsequent VSMC osteogenic transdifferentiation and biomineralization. Thus, the established molecular link between MFG-E8 and vascular calcification suggests that MFG-E8 can be therapeutically targeted to mitigate vascular calcification.


Asunto(s)
Factor de Crecimiento Transformador beta1 , Calcificación Vascular , Anciano , Antígenos de Superficie/genética , Antígenos de Superficie/metabolismo , Transdiferenciación Celular , Factor VIII/metabolismo , Glucolípidos , Glicoproteínas/metabolismo , Humanos , Gotas Lipídicas , Proteínas de la Leche/genética , Proteínas de la Leche/metabolismo , Miocitos del Músculo Liso/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Calcificación Vascular/metabolismo
4.
J Am Heart Assoc ; 10(11): e020870, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-34041925

RESUMEN

Background Migration of vascular smooth muscle cells (VSMCs) is the main contributor to neointimal formation. The Arp2/3 (actin-related proteins 2 and 3) complex activates actin polymerization and is involved in lamellipodia formation during VSMC migration. Milk fat globule-epidermal growth factor 8 (MFG-E8) is a glycoprotein expressed in VSMCs. We hypothesized that MFG-E8 regulates VSMC migration through modulation of Arp2/3-mediated actin polymerization. Methods and Results To determine whether MFG-E8 is essential for VSMC migration, a model of neointimal hyperplasia was induced in the common carotid artery of wild-type and MFG-E8 knockout mice, and the extent of neointimal formation was evaluated. Genetic deletion of MFG-E8 in mice attenuated injury-induced neointimal hyperplasia. Cultured VSMCs deficient in MFG-E8 exhibited decreased cell migration. Immunofluorescence and immunoblotting revealed decreased Arp2 but not Arp3 expression in the common carotid arteries and VSMCs deficient in MFG-E8. Exogenous administration of recombinant MFG-E8 biphasically and dose-dependently regulated the cultured VSMCs. At a low concentration, MFG-E8 upregulated Arp2 expression. By contrast, MFG-E8 at a high concentration reduced the Arp2 level and significantly attenuated actin assembly. Arp2 upregulation mediated by low-dose MFG-E8 was abolished by treating cultured VSMCs with ß1 integrin function-blocking antibody and Rac1 inhibitors. Moreover, treatment of the artery with a high dose of recombinant MFG-E8 diminished injury-induced neointimal hyperplasia and reduced VSMC migration. Conclusions MFG-E8 plays a critical role in VSMC migration through dose-dependent regulation of Arp2-mediated actin polymerization. These findings suggest that high doses of MFG-E8 may have therapeutic potential for treating vascular occlusive diseases.


Asunto(s)
Actinas/metabolismo , Antígenos de Superficie/genética , Arteriopatías Oclusivas/tratamiento farmacológico , Arteria Carótida Común/metabolismo , ADN/genética , Regulación de la Expresión Génica , Proteínas de la Leche/genética , Animales , Antígenos de Superficie/metabolismo , Antígenos de Superficie/uso terapéutico , Apoptosis , Arteriopatías Oclusivas/genética , Arteriopatías Oclusivas/patología , Arteria Carótida Común/patología , Movimiento Celular/genética , Proliferación Celular , Células Cultivadas , Modelos Animales de Enfermedad , Femenino , Masculino , Ratones , Ratones Noqueados , Microscopía Confocal , Proteínas de la Leche/metabolismo , Proteínas de la Leche/uso terapéutico , Músculo Liso Vascular/patología , Polimerizacion
5.
J Biomed Sci ; 26(1): 61, 2019 Aug 30.
Artículo en Inglés | MEDLINE | ID: mdl-31470852

RESUMEN

BACKGROUND: Among older adults, arterial aging is the major factor contributing to increased risk for cardiovascular disease-related morbidity and mortality. The chronic vascular inflammation that accompanies aging causes diffuse intimal-medial thickening of the arterial wall, thus increasing the vulnerability of aged vessels to vascular insults. Milk fat globule-epidermal growth factor 8 (MFG-E8) is a biomarker for aging arteries. This integrin-binding glycoprotein, induced by angiotensin II, facilitates vascular smooth muscle cell (VSMC) proliferation and invasion in aging vasculatures. This study investigated whether MFG-E8 directly mediates the initial inflammatory responses in aged arteries or VSMCs. METHODS: A model of neointimal hyperplasia was induced in the common carotid artery (CCA) of aged mice to exacerbate age-associated vascular remodeling. Recombinant MFG-E8 (rMFG-E8) was administered to the injured artery using Pluronic gel to accentuate the effect on age-related vascular pathophysiology. The MFG-E8 level, leukocyte infiltration, and proinflammatory cell adhesion molecule (CAM) expression in the arterial wall were evaluated through immunohistochemistry. By using immunofluorescence and immunoblotting, the activation of the critical proinflammatory transcription factor nuclear factor (NF)-κB in the injured CCAs was analyzed. Immunofluorescence, immunoblotting, and quantitative real-time polymerase chain reaction were conducted using VSMCs isolated from the aortas of young and aged mice to assess NF-κB nuclear translocation, NF-κB-dependent gene expression, and cell proliferation. The extent of intimal-medial thickening in the injured vessels was analyzed morphometrically. Finally, Transwell migration assay was used to examine VSMC migration. RESULTS: Endogenous MFG-E8 expression in aged CCAs was significantly induced by ligation injury. Aged CCAs treated with rMFG-E8 exhibited increased leukocyte extravasation, CAM expression, and considerably increased NF-κB activation induced by rMFG-E8 in the ligated vessels. Exposure of early passage VSMCs from aged aortas to rMFG-E8 substantially increased NF-κB activation, proinflammatory gene expression, and cell proliferation. However, rMFG-E8 attenuated VSMC migration. CONCLUSIONS: MFG-E8 promoted the proinflammatory phenotypic shift of aged VSMCs and arteries, rendering the vasculature prone to vascular diseases. MFG-E8 may constitute a novel therapeutic target for retarding the aging processes in such vessels.


Asunto(s)
Envejecimiento/genética , Antígenos de Superficie/genética , Arterias/fisiología , Inflamación/genética , Proteínas de la Leche/genética , Músculo Liso Vascular/inmunología , Animales , Antígenos de Superficie/metabolismo , Inflamación/fisiopatología , Ratones , Proteínas de la Leche/metabolismo , Músculo Liso Vascular/metabolismo , Fenotipo , Proteínas Recombinantes
6.
J Biomed Sci ; 26(1): 37, 2019 May 16.
Artículo en Inglés | MEDLINE | ID: mdl-31096970

RESUMEN

BACKGROUND: The blood-spinal cord barrier (BSCB) is composed of a monolayer of endothelium linked with tight junctions and extracellular matrix (ECM)-rich basement membranes and is surrounded by astrocyte foot processes. Endothelial permeability is regulated by interaction between endothelial cells and ECM proteins. Fibronectin (FN) is a principal ECM component of microvessels. Excessive FN deposition disrupts cell-cell adhesion in fibroblasts through ß1 integrin ligation. To determine whether excessive FN deposition contributes to the disruption of endothelial integrity, we used an in vitro model of the endothelial monolayer to investigate whether the FN inhibitor pUR4 prevents FN deposition into the subendothelial matrix and attenuates endothelial leakage. METHODS: To correlate the effects of excessive FN accumulation in microvessels on BSCB disruption, spinal nerve ligation-which induces BSCB leakage-was applied, and FN expression in the spinal cord was evaluated through immunohistochemistry and immunoblotting. To elucidate the effects by which pUR4 modulates endothelial permeability, brain-derived endothelial (bEND.3) cells treated with tumor necrosis factor (TNF)-α were used to mimic a leaky BSCB. A bEND.3 monolayer was preincubated with pUR4 before TNF-α treatment. The transendothelial electrical resistance (TEER) measurement and transendothelial permeability assay were applied to assess the endothelial integrity of the bEND.3 monolayer. Immunofluorescence analysis and immunoblotting were performed to evaluate the inhibitory effects of pUR4 on TNF-α-induced FN deposition. To determine the mechanisms underlying pUR4-mediated endothelial permeability, cell morphology, stress fiber formation, myosin light chain (MLC) phosphorylation, and ß1 integrin-mediated signaling were evaluated through immunofluorescence analysis and immunoblotting. RESULTS: Excessive FN was accumulated in the microvessels of the spinal cord after spinal nerve ligation; moreover, pUR4 inhibited TNF-α-induced FN deposition in the bEND.3 monolayer and maintained intact TEER and endothelial permeability. Furthermore, pUR4 reduced cell morphology alteration, actin stress fiber formation, and MLC phosphorylation, thereby attenuating paracellular gap formation. Moreover, pUR4 reduced ß1 integrin activation and downstream signaling. CONCLUSIONS: pUR4 reduces TNF-α-induced ß1 integrin activation by depleting ECM FN, leading to a decrease in endothelial hyperpermeability and maintenance of monolayer integrity. These findings suggest therapeutic benefits of pUR4 in pathological vascular leakage treatment.


Asunto(s)
Fibronectinas/antagonistas & inhibidores , Integrina beta1/metabolismo , Fragmentos de Péptidos/farmacología , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Adhesión Celular/efectos de los fármacos , Línea Celular , Permeabilidad de la Membrana Celular/efectos de los fármacos , Células Endoteliales/metabolismo , Endotelio Vascular/metabolismo , Matriz Extracelular/metabolismo , Humanos , Masculino , Ratas , Ratas Sprague-Dawley
7.
PLoS One ; 12(10): e0185811, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28968441

RESUMEN

Circulating fibrocytes play a key role in the pathogenesis of pulmonary fibrosis. Fibrocytes are bone marrow-derived leukocytes, which enter the lungs in response to their chemoattractant CXCL12 and differentiate into fibroblasts or myofibroblasts, leading to excess deposition of the collagen-rich extracellular matrix. Matrix metalloproteinase (MMP)-9 and MMP-2, secreted by fibrocytes, degrade the subendothelial basement membrane and promote fibrocyte influx into the lungs. Here, we demonstrate that R1R2, a novel peptide derived from the bacterial adhesin SFS, attenuates pulmonary fibrosis by preventing the differentiation of fibrocytes into myofibroblasts and by reducing the invasion of fibrocytes through basement membrane-like proteins. Moreover, our findings reveal dual regulation of R1R2 on MMP-9 through reduced enzymatic activity on gelatin and increased cleavage of CXCL12. These data suggest that R1R2 has potent anti-fibrotic effects against pulmonary fibrosis.


Asunto(s)
Diferenciación Celular , Movimiento Celular , Fibroblastos/metabolismo , Péptidos/farmacología , Fibrosis Pulmonar/prevención & control , Animales , Fibroblastos/citología , Ratones , Ratones Endogámicos C57BL
8.
PLoS One ; 10(2): e0117356, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25675397

RESUMEN

The extracellular matrix (ECM) is a major constituent of the vessel wall. In addition to providing a structural scaffold, the ECM controls numerous cellular functions in both physiologic and pathologic settings. Vascular remodeling occurs after injury and is characterized by endothelial cell activation, inflammatory cell infiltration, phenotypic modulation of smooth muscle cells (SMCs), and augmented deposition of collagen-rich ECM. R1R2, a peptide derived from the bacterial adhesin SFS, with sequence homology to collagen, is known to inhibit collagen type I deposition in vitro by inhibiting the binding of fibronectin to collagen. However, the inhibitory effects of R1R2 during vascular remodeling have not been explored. We periadventitially delivered R1R2 to carotid arteries using pluronic gel in a vascular remodeling mouse model induced by blood flow cessation, and evaluated its effects on intima-media thickening, ECM deposition, SMC activation, and inflammatory cell infiltration. Morphometric analysis demonstrated that R1R2 reduced intima-media thickening compared to the control groups. R1R2 treatment also decreased collagen type I deposition in the vessel wall, and maintained SMC in the contractile phenotype. Interestingly, R1R2 dramatically reduced inflammatory cell infiltration into the vessel by ∼ 78%. This decrease was accompanied by decreased VCAM-1 and ICAM-1 expression. Our in vitro studies revealed that R1R2 attenuated SMC proliferation and migration, and also decreased monocyte adhesion and transendothelial migration through endothelial cells. Together, these data suggest that R1R2 attenuates vascular remodeling responses by decreasing inflammation and by modulating SMC proliferation and migration, and suggest that the R1R2 peptide may have therapeutic potential in treating occlusive vascular diseases.


Asunto(s)
Adhesinas Bacterianas/química , Colágeno/antagonistas & inhibidores , Inflamación/metabolismo , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/metabolismo , Fragmentos de Péptidos/farmacología , Remodelación Vascular/efectos de los fármacos , Animales , Adhesión Celular/efectos de los fármacos , Moléculas de Adhesión Celular/metabolismo , Diferenciación Celular/efectos de los fármacos , Línea Celular , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Quimiotaxis de Leucocito/efectos de los fármacos , Colágeno/metabolismo , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Matriz Extracelular/metabolismo , Fibronectinas/metabolismo , Humanos , Hiperplasia , Inflamación/tratamiento farmacológico , Inflamación/inmunología , Ratones , Monocitos/efectos de los fármacos , Monocitos/metabolismo , Monocitos/patología , Contracción Muscular/efectos de los fármacos , Miocitos del Músculo Liso/citología , Neointima/patología , Fenotipo , Unión Proteica/efectos de los fármacos , Migración Transendotelial y Transepitelial/efectos de los fármacos
9.
Mol Biol Cell ; 25(5): 583-93, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24403604

RESUMEN

Tissue fibrosis occurs when matrix production outpaces matrix degradation. Degradation of collagen, the main component of fibrotic tissue, is mediated through an extracellular proteolytic pathway and intracellular pathway of cellular uptake and lysosomal digestion. Recent studies demonstrate that disruption of the intracellular pathways can exacerbate fibrosis. These pathways are poorly characterized. Here we identify novel mediators of the intracellular pathway of collagen turnover through a genome-wide RNA interference screen in Drosophila S2 cells. Screening of 7505 Drosophila genes conserved among metazoans identified 22 genes that were required for efficient internalization of type I collagen. These included proteins involved in vesicle transport, the actin cytoskeleton, and signal transduction. We show further that the flotillin genes have a conserved and central role in collagen uptake in Drosophila and human cells. Short hairpin RNA-mediated silencing of flotillins in human monocyte and fibroblasts impaired collagen uptake by promoting lysosomal degradation of the endocytic collagen receptors uPARAP/Endo180 and mannose receptor. These data provide an initial characterization of intracellular pathways of collagen turnover and identify the flotillin genes as critical regulators of this process. A better understanding of these pathways may lead to novel therapies that reduce fibrosis by increasing collagen turnover.


Asunto(s)
Colágeno/metabolismo , Proteínas de Drosophila/fisiología , Drosophila/genética , Proteínas de la Membrana/fisiología , Animales , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Técnicas de Silenciamiento del Gen , Genoma de los Insectos , Humanos , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Interferencia de ARN
10.
Am J Physiol Lung Cell Mol Physiol ; 304(11): L709-21, 2013 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-23564511

RESUMEN

Pulmonary fibrosis is a vexing clinical problem with no proven therapeutic options. In the normal lung there is continuous collagen synthesis and collagen degradation, and these two processes are precisely balanced to maintain normal tissue architecture. With lung injury there is an increase in the rate of both collagen production and collagen degradation. The increase in collagen degradation is critical in preventing the formation of permanent scar tissue each time the lung is exposed to injury. In pulmonary fibrosis, collagen degradation does not keep pace with collagen production, resulting in extracellular accumulation of fibrillar collagen. Collagen degradation occurs through both extracellular and intracellular pathways. The extracellular pathway involves cleavage of collagen fibrils by proteolytic enzyme including the metalloproteinases. The less-well-described intracellular pathway involves binding and uptake of collagen fragments by fibroblasts and macrophages for lysosomal degradation. The relationship between these two pathways and their relevance to the development of fibrosis is complex. Fibrosis in the lung, liver, and skin has been associated with an impaired degradative environment. Much of the current scientific effort in fibrosis is focused on understanding the pathways that regulate increased collagen production. However, recent reports suggest an important role for collagen turnover and degradation in regulating the severity of tissue fibrosis. The objective of this review is to evaluate the roles of the extracellular and intracellular collagen degradation pathways in the development of fibrosis and to examine whether pulmonary fibrosis can be viewed as a disease of impaired matrix degradation rather than a disease of increased matrix production.


Asunto(s)
Colágeno/metabolismo , Pulmón/metabolismo , Fibrosis Pulmonar/metabolismo , Animales , Colágeno/biosíntesis , Colagenasas/metabolismo , Matriz Extracelular/metabolismo , Fibroblastos/metabolismo , Humanos , Lisosomas/metabolismo , Macrófagos/metabolismo , Fibrosis Pulmonar/fisiopatología
11.
Proc Natl Acad Sci U S A ; 110(2): 660-5, 2013 Jan 08.
Artículo en Inglés | MEDLINE | ID: mdl-23269839

RESUMEN

Airway obstruction is a hallmark of allergic asthma and is caused primarily by airway smooth muscle (ASM) hypercontractility. Airway inflammation leads to the release of cytokines that enhance ASM contraction by increasing ras homolog gene family, member A (RhoA) activity. The protective mechanisms that prevent or attenuate the increase in RhoA activity have not been well studied. Here, we report that mice lacking the gene that encodes the protein Milk Fat Globule-EGF factor 8 (Mfge8(-/-)) develop exaggerated airway hyperresponsiveness in experimental models of asthma. Mfge8(-/-) ASM had enhanced contraction after treatment with IL-13, IL-17A, or TNF-α. Recombinant Mfge8 reduced contraction in murine and human ASM treated with IL-13. Mfge8 inhibited IL-13-induced NF-κB activation and induction of RhoA. Mfge8 also inhibited rapid activation of RhoA, an effect that was eliminated by an inactivating point mutation in the RGD integrin-binding site in recombinant Mfge8. Human subjects with asthma had decreased Mfge8 expression in airway biopsies compared with healthy controls. These data indicate that Mfge8 binding to integrin receptors on ASM opposes the effect of allergic inflammation on RhoA activity and identify a pathway for specific inhibition of ASM hypercontractility in asthma.


Asunto(s)
Antígenos de Superficie/metabolismo , Asma/fisiopatología , Hiperreactividad Bronquial/fisiopatología , Proteínas de la Leche/metabolismo , Contracción Muscular/fisiología , Músculo Liso/fisiología , Análisis de Varianza , Animales , Antígenos de Superficie/genética , Western Blotting , Lavado Broncoalveolar , Calcio/metabolismo , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Humanos , Interleucina-13/farmacología , Pulmón/patología , Ratones , Ratones Noqueados , Proteínas de la Leche/genética , FN-kappa B/metabolismo , Mutación Puntual/genética , Proteínas de Unión al GTP rho/metabolismo , Proteína de Unión al GTP rhoA
12.
Arterioscler Thromb Vasc Biol ; 31(10): 2172-80, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21817093

RESUMEN

OBJECTIVE: Smooth muscle calponin (CNN1) contains multiple conserved intronic CArG elements that bind serum response factor and display enhancer activity in vitro. The objectives here were to evaluate these CArG elements for activity in transgenic mice and determine the effect of human CNN1 on injury-induced vascular remodeling. METHODS AND RESULTS: Mice carrying a lacZ reporter under control of intronic CArG elements in the human CNN1 gene failed to show smooth muscle cell (SMC)-restricted activity. However, deletion of the orthologous sequences in mice abolished endogenous Cnn1 promoter activity, suggesting their necessity for in vivo Cnn1 expression. Mice carrying a 38-kb bacterial artificial chromosome (BAC) harboring the human CNN1 gene displayed SMC- restricted expression of the corresponding CNN1 protein, as measured by immunohistochemistry and Western blotting. Extensive BAC recombineering studies revealed the absolute necessity of a single intronic CArG element for correct SMC-restricted expression of human CNN1. Overexpressing human CNN1 suppressed neointimal formation following arterial injury. Mice with an identical BAC carrying mutations in CArG elements that inhibit human CNN1 expression showed outward remodeling and neointimal formation. CONCLUSIONS: A single intronic CArG element is necessary but insufficient for proper CNN1 expression in vivo. CNN1 overexpression antagonizes arterial injury-induced neointimal formation.


Asunto(s)
Proteínas de Unión al Calcio/genética , Traumatismos de las Arterias Carótidas/metabolismo , Proliferación Celular , Proteínas de Microfilamentos/genética , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Regiones Promotoras Genéticas , Túnica Íntima/metabolismo , Animales , Sitios de Unión , Western Blotting , Proteínas de Unión al Calcio/deficiencia , Proteínas de Unión al Calcio/metabolismo , Arterias Carótidas/metabolismo , Arterias Carótidas/patología , Traumatismos de las Arterias Carótidas/genética , Traumatismos de las Arterias Carótidas/patología , Línea Celular , Cromosomas Artificiales Bacterianos , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Genes Reporteros , Humanos , Inmunohistoquímica , Intrones , Operón Lac , Luciferasas/genética , Luciferasas/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Proteínas de Microfilamentos/deficiencia , Proteínas de Microfilamentos/metabolismo , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/patología , Ratas , Elemento de Respuesta al Suero , Factor de Respuesta Sérica/metabolismo , Transfección , Túnica Íntima/patología , Calponinas
13.
PLoS One ; 6(4): e18538, 2011 Apr 05.
Artículo en Inglés | MEDLINE | ID: mdl-21483686

RESUMEN

Retinoids are used clinically to treat a number of hyper-proliferative disorders and have been shown in experimental animals to attenuate vascular occlusive diseases, presumably through nuclear receptors bound to retinoic acid response elements (RARE) located in target genes. Here, we show that natural or synthetic retinoids rapidly induce mRNA and protein expression of a specific isoform of A-Kinase Anchoring Protein 12 (AKAP12ß) in cultured smooth muscle cells (SMC) as well as the intact vessel wall. Expression kinetics and actinomycin D studies indicate Akap12ß is a retinoid-induced, immediate-early gene. Akap12ß promoter analyses reveal a conserved RARE mildly induced with atRA in a region that exhibits hyper-acetylation. Immunofluorescence microscopy and protein kinase A (PKA) regulatory subunit overlay assays in SMC suggest a physical association between AKAP12ß and PKA following retinoid treatment. Consistent with its designation as a tumor suppressor, inducible expression of AKAP12ß attenuates SMC growth in vitro. Further, immunohistochemistry studies establish marked decreases in AKAP12 expression in experimentally-injured vessels of mice as well as atheromatous lesions in humans. Collectively, these results demonstrate a novel role for retinoids in the induction of an AKAP tumor suppressor that blocks vascular SMC growth thus providing new molecular insight into how retiniods may exert their anti-proliferative effects in the injured vessel wall.


Asunto(s)
Proteínas de Anclaje a la Quinasa A/genética , Proteínas de Anclaje a la Quinasa A/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Genes Supresores de Tumor , Músculo Liso Vascular/citología , Músculo Liso Vascular/metabolismo , Retinoides/farmacología , Animales , Línea Celular , Proliferación Celular/efectos de los fármacos , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Humanos , Masculino , Ratones , Músculo Liso Vascular/efectos de los fármacos , Regiones Promotoras Genéticas/genética , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Ratas , Elementos de Respuesta/genética , Tretinoina/farmacología
14.
Nature ; 460(7256): 705-10, 2009 Aug 06.
Artículo en Inglés | MEDLINE | ID: mdl-19578358

RESUMEN

MicroRNAs (miRNAs) are regulators of myriad cellular events, but evidence for a single miRNA that can efficiently differentiate multipotent stem cells into a specific lineage or regulate direct reprogramming of cells into an alternative cell fate has been elusive. Here we show that miR-145 and miR-143 are co-transcribed in multipotent murine cardiac progenitors before becoming localized to smooth muscle cells, including neural crest stem-cell-derived vascular smooth muscle cells. miR-145 and miR-143 were direct transcriptional targets of serum response factor, myocardin and Nkx2-5 (NK2 transcription factor related, locus 5) and were downregulated in injured or atherosclerotic vessels containing proliferating, less differentiated smooth muscle cells. miR-145 was necessary for myocardin-induced reprogramming of adult fibroblasts into smooth muscle cells and sufficient to induce differentiation of multipotent neural crest stem cells into vascular smooth muscle. Furthermore, miR-145 and miR-143 cooperatively targeted a network of transcription factors, including Klf4 (Kruppel-like factor 4), myocardin and Elk-1 (ELK1, member of ETS oncogene family), to promote differentiation and repress proliferation of smooth muscle cells. These findings demonstrate that miR-145 can direct the smooth muscle fate and that miR-145 and miR-143 function to regulate the quiescent versus proliferative phenotype of smooth muscle cells.


Asunto(s)
Linaje de la Célula , MicroARNs/metabolismo , Miocitos del Músculo Liso/citología , Miocitos del Músculo Liso/metabolismo , Animales , Diferenciación Celular , Proliferación Celular , Femenino , Regulación de la Expresión Génica , Proteína Homeótica Nkx-2.5 , Proteínas de Homeodominio/metabolismo , Factor 4 Similar a Kruppel , Masculino , Ratones , Ratones Transgénicos , MicroARNs/genética , Modelos Biológicos , Miocardio/metabolismo , Factores de Transcripción/metabolismo , Transcripción Genética , Enfermedades Vasculares/metabolismo , Proteína Elk-4 del Dominio ets/metabolismo
15.
Circ Res ; 105(3): 271-8, 2009 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-19574551

RESUMEN

RATIONALE: We previously identified a novel serine carboxypeptidase, SCPEP1, that undergoes cleavage across all tissues where it is expressed. SCPEP1 bears the signature catalytic triad found in all serine carboxypeptidases, but its biological function is completely unknown. OBJECTIVE: To begin elucidating the functions of SCPEP1 in vitro and in the vessel wall after injury. METHODS AND RESULTS: Cultured smooth muscle cells were transduced with adenovirus carrying wild-type Scpep1, a short hairpin RNA to Scpep1, or variants of Scpep1 with mutations that disrupt the catalytic triad domain or SCPEP1 cleavage. Western blotting of key growth regulators or growth and migratory responses were assessed following SCPEP1 gain- or loss-of-function in smooth muscle cells. Vascular injury-induced remodeling and cell proliferation were evaluated in wild-type or newly created Scpep1 knockout mice. Overexpression of wild-type or cleavage-defective SCPEP1, but not a catalytic triad mutant SCPEP1, promotes smooth muscle cell proliferation and migration in vitro. A short hairpin RNA to Scpep1 blunts endogenous growth, which is rescued on concurrent expression of Scpep1 carrying silent mutations that evade knockdown. SCPEP1 protein is highly expressed in the neointima of 2 models of vascular remodeling. Scpep1-null mice show decreases in medial and intimal cell proliferation as well as vessel remodeling following arterial injury. CONCLUSIONS: SCPEP1 promotes smooth muscle cell proliferation and migration in a catalytic triad-dependent, cleavage-independent manner. SCPEP1 represents a new mediator of vascular remodeling and a potential therapeutic target for the treatment of vascular occlusive diseases.


Asunto(s)
Carboxipeptidasas/metabolismo , Movimiento Celular/fisiología , Proliferación Celular , Músculo Liso Vascular/citología , Músculo Liso Vascular/metabolismo , Adenoviridae , Animales , Carboxipeptidasas/genética , Células Cultivadas , Ratones , Ratones Noqueados , Mutación/genética , Transducción Genética , Túnica Íntima/citología , Túnica Íntima/metabolismo , Túnica Media/citología , Túnica Media/metabolismo
16.
J Histochem Cytochem ; 54(6): 701-11, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16461364

RESUMEN

We previously identified a novel gene designated retinoid-inducible serine carboxypeptidase (RISC or Scpep1). Here we characterize a polyclonal antibody raised to Scpep1 and assess its localization in mouse cells and tissues. Western blot analysis revealed an immunospecific approximately 35-kDa protein corresponding to endogenous Scpep1. This protein is smaller than the predicted approximately 51-kDa, suggesting that Scpep1 is proteolytically cleaved to a mature enzyme. Immunohistochemical studies demonstrate Scpep1 expression in embryonic heart and vasculature as well as in adult aortic smooth muscle cells and endothelial cells. Scpep1 displays a broad expression pattern in adult tissues with detectable levels in epithelia of digestive tract and urinary bladder, islet of Langerhans, type II alveolar cells and macrophages of lung, macrophage-like cells of lymph nodes and spleen, Leydig cells of testis, and nerve fibers in brain and ganglia. Consistent with previous mRNA studies in kidney, Scpep1 protein is restricted to proximal convoluted tubular epithelium (PCT). Immunoelectron microscopy shows enriched Scpep1 within lysosomes of the PCT, and immunofluorescence microscopy colocalizes Scpep1 with lysosomal-associated membrane protein-2. These results suggest that Scpep1 is a widely distributed lysosomal protease requiring proteolytic cleavage for activity. The highly specific Scpep1 antibody characterized herein provides a necessary reagent for elucidating Scpep1 function.


Asunto(s)
Carboxipeptidasas/biosíntesis , Animales , Anticuerpos , Carboxipeptidasas/inmunología , Células Cultivadas , Embrión de Mamíferos/enzimología , Humanos , Inmunohistoquímica , Túbulos Renales Proximales/enzimología , Proteína 2 de la Membrana Asociada a los Lisosomas , Proteínas de Membrana de los Lisosomas/metabolismo , Lisosomas/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Microscopía Fluorescente , Microscopía Inmunoelectrónica , Especificidad de Órganos
17.
J Exp Med ; 198(12): 1899-908, 2003 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-14676301

RESUMEN

Phenotype analysis of female mice lacking androgen receptor (AR) deficient (AR-/-) indicates that the development of mammary glands is retarded with reduced ductal branching in the prepubertal stages, and fewer Cap cells in the terminal end buds, as well as decreased lobuloalveolar development in adult females, and fewer milk-producing alveoli in the lactating glands. The defective development of AR-/- mammary glands involves the defects of insulin-like growth factor I-insulin-like growth factor I receptor and mitogen-activated protein kinase (MAPK) signals as well as estrogen receptor (ER) activity. Similar growth retardation and defects in growth factor-mediated Ras/Raf/MAPK cascade and ER signaling are also found in AR-/- MCF7 breast cancer cells. The restoration assays show that AR NH2-terminal/DNA-binding domain, but not the ligand-binding domain, is essential for normal MAPK function in MCF7 cells, and an AR mutant (R608K), found in male breast cancer, is associated with the excessive activation of MAPK. Together, our data provide the first in vivo evidence showing that AR-mediated MAPK and ER activation may play important roles for mammary gland development and MCF7 breast cancer cell proliferation.


Asunto(s)
Glándulas Mamarias Animales/crecimiento & desarrollo , Receptores Androgénicos/fisiología , Animales , Sitios de Unión , División Celular , Línea Celular Tumoral , ADN/metabolismo , Femenino , Humanos , Factor I del Crecimiento Similar a la Insulina/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Morfogénesis , Fenotipo , Receptor IGF Tipo 1/fisiología , Receptores de Estrógenos/análisis , Transcripción Genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...