Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 56
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
J Pharmacol Exp Ther ; 378(1): 1-9, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33879542

RESUMEN

Aging is a progressive, multifactorial, degenerative process in which deleterious changes occur in the biochemistry and function of organs. We showed that angiotensin II (AngII)-induced pathologies in the heart and kidney of young (3-month-old) mice are suppressed by the caveolin-1 scaffolding domain (CSD) peptide. Because AngII mediates many aging-associated changes, we explored whether CSD could reverse pre-existing pathologies and improve organ function in aged mice. Using 18-month-old mice (similar to 60-year-old humans), we found that >5-fold increases in leakage of serum proteins and >2-fold increases in fibrosis are associated with aging in the heart, kidney, and brain. Because tyrosine phosphorylation of cell junction proteins leads to the loss of microvascular barrier function, we analyzed the activation of the receptor tyrosine kinase PDGFR and the nonreceptor tyrosine kinases c-Src and Pyk2. We observed 5-fold activation of PDGFR and 2- to 3-fold activation of c-Src and Pyk2 in aged mice. Treatment with CSD for 4 weeks reversed these pathologic changes (microvascular leakage, fibrosis, kinase activation) in all organs almost down to the levels in healthy, young mice. In studies of heart function, CSD reduced the aging-associated increase in cardiomyocyte cross-sectional area and enhanced ventricular compliance in that echocardiographic studies demonstrated improved ejection fraction and fractional shortening and reduced isovolumic relation time. These results suggest that versions of CSD may be developed as treatments for aging-associated diseases in human patients based on the concept that CSD inhibits tyrosine kinases, leading to the inhibition of microvascular leakage and associated fibrosis, thereby improving organ function. SIGNIFICANCE STATEMENT: The caveolin-1 scaffolding domain (CSD) peptide reverses aging-associated fibrosis, microvascular leakage, and organ dysfunction in the heart, kidneys, and brain via a mechanism that involves the suppression of the activity of multiple tyrosine kinases, suggesting that CSD can be developed as a treatment for a wide range of diseases found primarily in the aged.


Asunto(s)
Envejecimiento/efectos de los fármacos , Envejecimiento/metabolismo , Caveolina 1/farmacología , Corazón/efectos de los fármacos , Riñón/efectos de los fármacos , Riñón/metabolismo , Envejecimiento/patología , Secuencia de Aminoácidos , Animales , Caveolina 1/química , Caveolina 1/genética , Femenino , Riñón/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas Tirosina Quinasas
2.
Lab Invest ; 97(4): 370-382, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28112757

RESUMEN

Chronic ventricular pressure overload (PO) results in congestive heart failure (CHF) in which myocardial fibrosis develops in concert with ventricular dysfunction. Caveolin-1 is important in fibrosis in various tissues due to its decreased expression in fibroblasts and monocytes. The profibrotic effects of low caveolin-1 can be blocked with the caveolin-1 scaffolding domain peptide (CSD, a caveolin-1 surrogate) using both mouse models and human cells. We have studied the beneficial effects of CSD on mice in which PO was induced by trans-aortic constriction (TAC). Beneficial effects observed in TAC mice receiving CSD injections daily included: improved ventricular function (increased ejection fraction, stroke volume, and cardiac output; reduced wall thickness); decreased collagen I, collagen chaperone HSP47, fibronectin, and CTGF levels; decreased activation of non-receptor tyrosine kinases Pyk2 and Src; and decreased activation of eNOS. To determine the source of cells that contribute to fibrosis in CHF, flow cytometric studies were performed that suggested that myofibroblasts in the heart are in large part bone marrow-derived. Two CD45+ cell populations were observed. One (Zone 1) contained CD45+/HSP47-/macrophage marker+ cells (macrophages). The second (Zone 2) contained CD45moderate/HSP47+/macrophage marker- cells often defined as fibrocytes. TAC increased the number of cells in Zones 1 and 2 and the level of HSP47 in Zone 2. These studies are a first step in elucidating the mechanism of action of CSD in heart fibrosis and promoting the development of CSD as a novel treatment to reduce fibrosis and improve ventricular function in CHF patients.


Asunto(s)
Caveolina 1/farmacología , Corazón/efectos de los fármacos , Miocardio/patología , Fragmentos de Péptidos/farmacología , Función Ventricular/efectos de los fármacos , Animales , Aorta/patología , Aorta/fisiopatología , Western Blotting , Colágeno Tipo I/genética , Colágeno Tipo I/metabolismo , Constricción Patológica/fisiopatología , Fibrosis/prevención & control , Citometría de Flujo , Quinasa 2 de Adhesión Focal/metabolismo , Expresión Génica/efectos de los fármacos , Proteínas del Choque Térmico HSP47/genética , Proteínas del Choque Térmico HSP47/metabolismo , Corazón/fisiopatología , Humanos , Integrina beta3/metabolismo , Antígenos Comunes de Leucocito/metabolismo , Masculino , Ratones Endogámicos C57BL , Miocardio/metabolismo , Óxido Nítrico Sintasa de Tipo III/metabolismo , Presión , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Familia-src Quinasas/metabolismo
3.
J Biol Chem ; 289(12): 8545-61, 2014 Mar 21.
Artículo en Inglés | MEDLINE | ID: mdl-24469446

RESUMEN

Periostin (PN), a novel fasciclin-related matricellular protein, has been implicated in cardiac development and postnatal remodeling, but the mechanism remains unknown. We examined the role of PN in mediating intracellular kinase activation for atrioventricular valve morphogenesis using well defined explant cultures, gene transfection systems, and Western blotting. The results show that valve progenitor (cushion) cells secrete PN into the extracellular matrix, where it can bind to INTEGRINs and activate INTEGRIN/focal adhesion kinase signaling pathways and downstream kinases, PI3K/AKT and ERK. Functional assays with prevalvular progenitor cells showed that activating these signaling pathways promoted adhesion, migration, and anti-apoptosis. Through activation of PI3K/ERK, PN directly enhanced collagen expression. Comparing PN-null to WT mice also revealed that expression of hyaluronan (HA) and activation of hyaluronan synthase-2 (Has2) are also enhanced upon PN/INTEGRIN/focal adhesion kinase-mediated activation of PI3K and/or ERK, an effect confirmed by the reduction of HA synthase-2 in PN-null mice. We also identified in valve progenitor cells a potential autocrine signaling feedback loop between PN and HA through PI3K and/or ERK. Finally, in a three-dimensional assay to simulate normal valve maturation in vitro, PN promoted collagen compaction in a kinase-dependent fashion. In summary, this study provides the first direct evidence that PN can act to stimulate a valvulogenic signaling pathway.


Asunto(s)
Moléculas de Adhesión Celular/metabolismo , Válvulas Cardíacas/embriología , Ácido Hialurónico/metabolismo , Transducción de Señal , Animales , Adhesión Celular , Moléculas de Adhesión Celular/genética , Movimiento Celular , Proliferación Celular , Células Cultivadas , Embrión de Pollo , Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Eliminación de Gen , Válvulas Cardíacas/citología , Válvulas Cardíacas/metabolismo , Integrinas/metabolismo , Sistema de Señalización de MAP Quinasas , Ratones , Ratones Endogámicos C57BL , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ovinos
4.
J Biol Chem ; 289(11): 7856-72, 2014 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-24324260

RESUMEN

The hepatocyte growth factor (HGF) and the HGF receptor Met pathway are important in the pathogenesis of interstitial lung disease (ILD). Alternatively spliced isoforms of CD44 containing variable exon 6 (CD44v6) and its ligand hyaluronan (HA) alter cellular function in response to interaction between CD44v6 and HGF. TGF-ß1 is the crucial cytokine that induces fibrotic action in ILD fibroblasts (ILDFbs). We have identified an autocrine TGF-ß1 signaling that up-regulates both Met and CD44v6 mRNA and protein expression. Western blot analysis, flow cytometry, and immunostaining revealed that CD44v6 and Met colocalize in fibroblasts and in tissue sections from ILD patients and in lungs of bleomycin-treated mice. Interestingly, cell proliferation induced by TGF-ß1 is mediated through Met and CD44v6. Further, cell proliferation mediated by TGF-ß1/CD44v6 is ERK-dependent. In contrast, action of Met on ILDFb proliferation does not require ERK but does require p38(MAPK). ILDFbs were sorted into CD44v6(+)/Met(+) and CD44v6(-)/Met(+) subpopulations. HGF inhibited TGF-ß1-stimulated collagen-1 and α-smooth muscle cell actin expression in both of these subpopulations by interfering with TGF-ß1 signaling. HGF alone markedly stimulated CD44v6 expression, which in turn regulated collagen-1 synthesis. Our data with primary lung fibroblast cultures with respect to collagen-1, CD44v6, and Met expressions were supported by immunostaining of lung sections from bleomycin-treated mice and from ILD patients. These results define the relationships between CD44v6, Met, and autocrine TGF-ß1 signaling and the potential modulating influence of HGF on TGF-ß1-induced CD44v6-dependent fibroblast function in ILD fibrosis.


Asunto(s)
Receptores de Hialuranos/metabolismo , Enfermedades Pulmonares Intersticiales/metabolismo , Proteínas Proto-Oncogénicas c-met/metabolismo , Fibrosis Pulmonar/metabolismo , Transducción de Señal , Factor de Crecimiento Transformador beta1/metabolismo , Animales , Comunicación Autocrina , Núcleo Celular/metabolismo , Proliferación Celular , Células Cultivadas , Medios de Cultivo/química , Ensayo de Inmunoadsorción Enzimática , Femenino , Fibroblastos/metabolismo , Citometría de Flujo , Regulación de la Expresión Génica , Factor de Crecimiento de Hepatocito/metabolismo , Humanos , Pulmón/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Fibrosis Pulmonar/patología , ARN Mensajero/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa
5.
Am J Physiol Lung Cell Mol Physiol ; 306(8): L736-48, 2014 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-24583879

RESUMEN

The interstitial lung diseases (ILD) include a large number of chronic, progressive, irreversible respiratory disorders involving pulmonary fibrosis, the most common of which are idiopathic pulmonary fibrosis and scleroderma lung disease (SSc ILD). Because bleomycin causes lung fibrosis when used in cancer chemotherapy, it is used to model human ILD in rodents. In most studies, bleomycin has been delivered directly into the lung by intratracheal or intraoral administration. Here we have compared the effects in mice of bleomycin delivered directly into the lungs (direct model) or systemically using osmotic minipumps (pump model) to determine which more closely resembles human ILD. The pump model is more similar to human SSc ILD in that: 1) lung injury/fibrosis is limited to the subpleural portion of the lung in the pump model and in SSc ILD, whereas the entire lung is affected in the direct model; 2) conversely, there is massive inflammation throughout the lung in the direct model, whereas inflammation is limited in the pump model and in SSc ILD; 3) hypertrophic type II alveolar epithelial cells are present at high levels in SSc ILD and in the pump model but not in the direct model; and 4) lung fibrosis is accompanied by dermal fibrosis. The pump model is also move convenient and humane than the direct model because there is less weight loss and mortality.


Asunto(s)
Antibióticos Antineoplásicos/administración & dosificación , Bleomicina/administración & dosificación , Sistemas de Liberación de Medicamentos , Bombas de Infusión , Enfermedades Pulmonares Intersticiales/tratamiento farmacológico , Esclerodermia Sistémica/tratamiento farmacológico , Animales , Caveolina 1/metabolismo , Proteínas de la Matriz Extracelular/metabolismo , Técnica del Anticuerpo Fluorescente , Humanos , Técnicas para Inmunoenzimas , Enfermedades Pulmonares Intersticiales/metabolismo , Enfermedades Pulmonares Intersticiales/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ósmosis , Alveolos Pulmonares/efectos de los fármacos , Alveolos Pulmonares/patología , Esclerodermia Sistémica/metabolismo , Esclerodermia Sistémica/patología , Pérdida de Peso/efectos de los fármacos
6.
Respir Res ; 14: 90, 2013 Sep 08.
Artículo en Inglés | MEDLINE | ID: mdl-24011378

RESUMEN

BACKGROUND: In fibrotic lung diseases, expression of caveolin-1 is decreased in fibroblasts and monocytes. The effects of this deficiency are reversed by treating cells or animals with the caveolin-1 scaffolding domain peptide (CSD, amino acids 82-101 of caveolin-1) which compensates for the lack of caveolin-1. Here we compare the function of CSD subdomains (Cav-A, Cav-B, Cav-C, Cav-AB, and Cav-BC) and mutated versions of CSD (F92A and T90A/T91A/F92A). METHODS: Migration toward the chemokine CXCL12 and the associated expression of F-actin, CXCR4, and pSmad 2/3 were studied in monocytes from healthy donors and SSc patients. Fibrocyte differentiation was studied using PBMC from healthy donors and SSc patients. Collagen I secretion and signaling were studied in fibroblasts derived from the lung tissue of healthy subjects and SSc patients. RESULTS: Cav-BC and CSD at concentrations as low as 0.01 µM inhibited the hypermigration of SSc monocytes and TGFß-activated Normal monocytes and the differentiation into fibrocytes of SSc and Normal monocytes. While CSD also inhibited the migration of poorly migrating Normal monocytes, Cav-A (and other subdomains to a lesser extent) promoted the migration of Normal monocytes while inhibiting the hypermigration of TGFß-activated Normal monocytes. The effects of versions of CSD on migration may be mediated in part via their effects on CXCR4, F-actin, and pSmad 2/3 expression. Cav-BC was as effective as CSD in inhibiting fibroblast collagen I and ASMA expression and MEK/ERK signaling. Cav-C and Cav-AB also inhibited collagen I expression, but in many cases did not affect ASMA or MEK/ERK. Cav-A increased collagen I expression in scleroderma lung fibroblasts. Full effects on fibroblasts of versions of CSD required 5 µM peptide. CONCLUSIONS: Cav-BC retains most of the anti-fibrotic functions of CSD; Cav-A exhibits certain pro-fibrotic functions. Results obtained with subdomains and mutated versions of CSD further suggest that the critical functional residues in CSD depend on the cell type and readout being studied. Monocytes may be more sensitive to versions of CSD than fibroblasts and endothelial cells because the baseline level of caveolin-1 in monocytes is much lower than in these other cell types.


Asunto(s)
Caveolina 1/farmacología , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Esclerodermia Sistémica/metabolismo , Actinas/metabolismo , Adolescente , Adulto , Anciano , Estudios de Casos y Controles , Diferenciación Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Células Cultivadas , Femenino , Fibroblastos/patología , Humanos , Pulmón/patología , Masculino , Regiones de Fijación a la Matriz , Persona de Mediana Edad , Monocitos/efectos de los fármacos , Monocitos/metabolismo , Monocitos/patología , Estructura Terciaria de Proteína , Receptores CXCR4/metabolismo , Esclerodermia Sistémica/patología , Proteína Smad2/metabolismo , Proteína smad3/metabolismo , Adulto Joven
7.
Front Immunol ; 14: 1116644, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37822942

RESUMEN

Introduction: In the US, despite the recent decline in breast cancer deaths, a persistent mortality disparity exists between black and white women with breast cancer, with black women having a 41% higher death rate. Several studies are now reporting that racial disparities can exist independent of socioeconomic and standard of care issues, suggesting that biological factors may be involved. Caveolin-1 (Cav1) loss in the tumor stromal compartment is a novel clinical biomarker for predicting poor outcome in breast cancer including triple negative subtype, however the mechanism of Cav1 loss is unknown. We previously identified miR-510-5p as a novel oncomir and propose here that the high levels observed in patients is a novel mechanism leading to stromal Cav1 loss and worse outcomes. Methods: Cav1 was identified as a direct target of miR-510-5p through luciferase, western blot and qPCR assays. Stromal cross talk between epithelial cells and fibroblasts was assessed in vitro using transwell co-culture assays and in vivo using xenograft assays. Results: We found that Cav1 is a direct target of miR-510-5p and that expression in fibroblasts results in an 'activated' phenotype. We propose that this could be important in the context of cancer disparities as we also observed increased levels of circulating miR-510-5p and reduced levels of stromal Cav1 in black women compared to white women with breast cancer. Finally, we observed a significant increase in tumor growth when tumor cells were co-injected with miR-510-5p expressing cancer associated fibroblasts in vivo. Conclusion: We propose that miR-510-5p mediated negative regulation of Cav1 in fibroblasts is a novel mechanism of aggressive tumor growth and may be a driver of breast cancer disparity.


Asunto(s)
Neoplasias de la Mama , Caveolina 1 , MicroARNs , Femenino , Humanos , Neoplasias de la Mama/patología , Caveolina 1/genética , Caveolina 1/metabolismo , Línea Celular Tumoral , Fibroblastos/metabolismo , MicroARNs/genética , MicroARNs/metabolismo
8.
Int J Cancer ; 131(9): 2034-43, 2012 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-22322590

RESUMEN

Invasiveness is one of the key features of aggressive prostate cancer; however, our understanding of the precise mechanisms effecting invasion remains limited. The ceramide hydrolyzing enzyme acid ceramidase (AC), overexpressed in most prostate tumors, causes an aggressive and invasive phenotype through downstream effectors that have not yet been well characterized. Here, we demonstrate that AC, through generation of sphingosine-1-phosphate (S1P), promotes Ets1 nuclear expression and binding to the promoter region of matrix-degrading protease cathepsin B. Through confocal microscopy and flow cytometry, we found that AC overexpression promotes pericellular localization of cathepsin B and its translocation to the outer leaflet of the cell membrane. AC overexpressing cells have an increased abundance of cathepsin B-enriched invasive structures and enhanced ability to invade through a collagen matrix, but not in the presence of an inhibitor of cathepsin B. In human prostate tissues, AC and cathepsin B overexpression were strongly associated and may relate to poor outcome. These results demonstrate a novel pathway by which AC, through S1P, promotes an invasive phenotype in prostate cancer by causing overexpression and secretion of cathepsin B through activation and nuclear expression of Ets1. As prostate cancer prognosis is dramatically worse when invasion has occurred, this study provides critical insight into the progression toward lethal prostate cancer.


Asunto(s)
Ceramidasa Ácida/metabolismo , Catepsina B/metabolismo , Lisofosfolípidos/biosíntesis , Invasividad Neoplásica , Neoplasias de la Próstata/metabolismo , Proteína Proto-Oncogénica c-ets-1/metabolismo , Esfingosina/análogos & derivados , Catepsina B/antagonistas & inhibidores , Catepsina B/genética , Línea Celular Tumoral , Membrana Celular/metabolismo , Movimiento Celular , Humanos , Masculino , Regiones Promotoras Genéticas , Neoplasias de la Próstata/genética , Esfingosina/biosíntesis , Regulación hacia Arriba
9.
Curr Opin Rheumatol ; 24(6): 642-8, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22955018

RESUMEN

PURPOSE OF REVIEW: Racial disparities appear to exist in the susceptibility and severity of systemic sclerosis (SSc, scleroderma) and are responsible for a greater health burden in blacks as compared with whites. Disparities in socioeconomic status and access to healthcare do not sufficiently explain the observed differences in prevalence and mortality. It is important to determine whether there might be a biologic basis for the racial disparities observed in SSc. RECENT FINDINGS: We present data to suggest that the increased susceptibility and severity of SSc in blacks may result in part from an imbalance of profibrotic and antifibrotic factors. Racial differences in the expression of transforming growth factor-ß1 (TGF-ß1) and caveolin-1, as well as differences in the expression of hepatocyte growth factor and PPAR-γ, have been demonstrated in blacks with SSc, as well as in normal black individuals. A genetic predisposition to fibrosis may account for much of the racial disparities between black and white patients with SSc. SUMMARY: A better understanding of the biologic basis for the racial disparities observed in SSc may lead to improved therapies, along with the recognition that different therapies may need to be adapted for different groups of patients.


Asunto(s)
Negro o Afroamericano/etnología , Disparidades en el Estado de Salud , Esclerodermia Sistémica/etnología , Población Blanca/etnología , Negro o Afroamericano/genética , Caveolina 1/genética , Susceptibilidad a Enfermedades/etnología , Predisposición Genética a la Enfermedad/etnología , Predisposición Genética a la Enfermedad/genética , Factor de Crecimiento de Hepatocito/genética , Humanos , PPAR gamma/genética , Esclerodermia Sistémica/genética , Esclerodermia Sistémica/terapia , Factor de Crecimiento Transformador beta1/genética , Población Blanca/genética
10.
PLoS One ; 17(2): e0264413, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35213624

RESUMEN

The caveolin-1 scaffolding domain (CSD, amino acids 82-101 of caveolin-1) has been shown to suppress bleomycin-induced lung and skin fibrosis and angiotensin II (AngII)-induced myocardial fibrosis. To identify active subregions within CSD, we split its sequence into three slightly overlapping 8-amino acid subregions (82-89, 88-95, and 94-101). Interestingly, all three peptides showed activity. In bleomycin-treated mice, all three subregions suppressed the pathological effects on lung and skin tissue morphology. In addition, while bone marrow monocytes isolated from bleomycin-treated mice showed greatly enhanced migration in vitro toward CXCL12, treatment in vivo with CSD and its subregions almost completely suppressed this enhanced migration. In AngII-induced heart failure, both 82-89 and 88-95 significantly suppressed fibrosis (both Col I and HSP47 levels), microvascular leakage, and heart weight/ body weight ratio (HW/BW) while improving ventricular function. In contrast, while 94-101 suppressed the increase in Col I, it did not improve the other parameters. The idea that all three subregions can be active depending on the assay was further supported by experiments studying the in vitro migration of human monocytes in which all three subregions were extremely active. These studies are very novel in that it has been suggested that there is only one active region within CSD that is centered on amino acids 90-92. In contrast, we demonstrate here the presence of other active regions within CSD.


Asunto(s)
Caveolina 1/metabolismo , Movimiento Celular , Monocitos/metabolismo , Fibrosis Pulmonar/metabolismo , Enfermedades de la Piel/metabolismo , Animales , Bleomicina/efectos adversos , Bleomicina/farmacología , Ratones , Fibrosis Pulmonar/inducido químicamente , Enfermedades de la Piel/inducido químicamente
11.
PLoS One ; 17(1): e0262479, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35015787

RESUMEN

Heart failure is a leading cause of hospitalizations and mortality worldwide. Heart failure with a preserved ejection fraction (HFpEF) represents a significant clinical challenge due to the lack of available treatment modalities for patients diagnosed with HFpEF. One symptom of HFpEF is impaired diastolic function that is associated with increases in left ventricular stiffness. Increases in myocardial fibrillar collagen content is one factor contributing to increases in myocardial stiffness. Cardiac fibroblasts are the primary cell type that produce fibrillar collagen in the heart. However, relatively little is known regarding phenotypic changes in cardiac fibroblasts in HFpEF myocardium. In the current study, cardiac fibroblasts were established from left ventricular epicardial biopsies obtained from patients undergoing cardiovascular interventions and divided into three categories: Referent control, hypertension without a heart failure designation (HTN (-) HFpEF), and hypertension with heart failure (HTN (+) HFpEF). Biopsies were evaluated for cardiac myocyte cross-sectional area (CSA) and collagen volume fraction. Primary fibroblast cultures were assessed for differences in proliferation and protein expression of collagen I, Membrane Type 1-Matrix Metalloproteinase (MT1-MMP), and α smooth muscle actin (αSMA). Biopsies from HTN (-) HFpEF and HTN (+) HFpEF exhibited increases in myocyte CSA over referent control although only HTN (+) HFpEF exhibited significant increases in fibrillar collagen content. No significant changes in proliferation or αSMA was detected in HTN (-) HFpEF or HTN (+) HFpEF cultures versus referent control. Significant increases in production of collagen I was detected in HF (-) HFpEF fibroblasts, whereas significant decreases in MT1-MMP levels were measured in HTN (+) HFpEF cells. We conclude that epicardial biopsies provide a viable source for primary fibroblast cultures and that phenotypic differences are demonstrated by HTN (-) HFpEF and HTN (+) HFpEF cells versus referent control.


Asunto(s)
Biomarcadores/metabolismo , Fibroblastos/patología , Fibrosis/patología , Insuficiencia Cardíaca/patología , Ventrículos Cardíacos/patología , Hipertensión/fisiopatología , Miocardio/patología , Anciano , Estudios de Casos y Controles , Proliferación Celular , Células Cultivadas , Femenino , Fibroblastos/metabolismo , Fibrosis/metabolismo , Insuficiencia Cardíaca/metabolismo , Ventrículos Cardíacos/metabolismo , Humanos , Masculino , Persona de Mediana Edad , Miocardio/metabolismo , Pronóstico
12.
J Neurosci ; 30(13): 4693-706, 2010 Mar 31.
Artículo en Inglés | MEDLINE | ID: mdl-20357120

RESUMEN

A major barrier to regeneration of CNS axons is the presence of growth-inhibitory proteins associated with myelin and the glial scar. To identify chemical compounds with the ability to overcome the inhibition of regeneration, we screened a novel triazine library, based on the ability of compounds to increase neurite outgrowth from cerebellar neurons on inhibitory myelin substrates. The screen produced four "hit compounds," which act with nanomolar potency on several different neuronal types and on several distinct substrates relevant to glial inhibition. Moreover, the compounds selectively overcome inhibition rather than promote growth in general. The compounds do not affect neuronal cAMP levels, PKC activity, or EGFR (epidermal growth factor receptor) activation. Interestingly, one of the compounds alters microtubule dynamics and increases microtubule density in both fibroblasts and neurons. This same compound promotes regeneration of dorsal column axons after acute lesions and potentiates regeneration of optic nerve axons after nerve crush in vivo. These compounds should provide insight into the mechanisms through which glial-derived inhibitors of regeneration act, and could lead to the development of novel therapies for CNS injury.


Asunto(s)
Neuroglía/fisiología , Neuronas/efectos de los fármacos , Triazinas/farmacología , Animales , Axones/efectos de los fármacos , Axones/fisiología , Células Cultivadas , Cerebelo/citología , Corteza Cerebral/citología , AMP Cíclico/metabolismo , Receptores ErbB/metabolismo , Fibroblastos/efectos de los fármacos , Fibroblastos/fisiología , Ensayos Analíticos de Alto Rendimiento , Ratones , Ratones Endogámicos C57BL , Vaina de Mielina/fisiología , Compresión Nerviosa , Neuritas/efectos de los fármacos , Neuritas/fisiología , Neuronas/fisiología , Neuronas/ultraestructura , Nervio Óptico/citología , Proteína Quinasa C/metabolismo , Ratas , Ratas Sprague-Dawley , Regeneración , Médula Espinal/citología , Triazinas/química
13.
Ann Rheum Dis ; 69(6): 1220-6, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20410070

RESUMEN

OBJECTIVES: Reduced caveolin-1 levels in lung fibroblasts from patients with scleroderma and the lungs of bleomycin-treated mice promote collagen overexpression and lung fibrosis. This study was undertaken to determine whether caveolin-1 is deficient in leucocytes from bleomycin-treated mice and patients with scleroderma and to examine the consequences of this deficiency and its reversal. METHODS: Mice or cells received the caveolin-1 scaffolding domain (CSD) peptide to reverse the pathological effects of reduced caveolin-1 expression. In bleomycin-treated mice, the levels of caveolin-1 in leucocytes and the effect of CSD peptide on leucocyte accumulation in lung tissue were examined. To validate the results in human disease and to identify caveolin-1-regulated molecular mechanisms, monocytes and neutrophils were isolated from patients with scleroderma and control subjects and caveolin-1, extracellular signal-regulated protein kinase (ERK), c-Jun N-terminal kinase (JNK), p38, CXC chemokine receptor 4 (CXCR4) and matrix metalloproteinase 9 (MMP-9) expression/activation were evaluated. These parameters were also studied in monocytes treated with cytokines or CSD peptide. RESULTS: Leucocyte caveolin-1 is important in lung fibrosis. In bleomycin-treated mice, caveolin-1 expression was diminished in monocytes and CSD peptide inhibited leucocyte recruitment into the lungs. These observations are relevant to human disease. Monocytes and neutrophils from patients with scleroderma contained less caveolin-1 and more activated ERK, JNK and p38 than those from control subjects. Treatment with CSD peptide reversed ERK, JNK and p38 hyperactivation. Scleroderma monocytes also overexpressed CXCR4 and MMP-9, which was inhibited by the CSD peptide. Cytokine treatment of normal monocytes caused adoption of the scleroderma phenotype (low caveolin-1, high CXCR4 and MMP-9 and signalling molecule hyperactivation). CONCLUSIONS: Caveolin-1 downregulation in leucocytes contributes to fibrotic lung disease, highlighting caveolin-1 as a promising therapeutic target in scleroderma.


Asunto(s)
Caveolina 1/fisiología , Leucocitos/fisiología , Fibrosis Pulmonar/metabolismo , Esclerodermia Sistémica/metabolismo , Adulto , Anciano , Animales , Bleomicina , Caveolina 1/sangre , Caveolina 1/deficiencia , Células Cultivadas , Modelos Animales de Enfermedad , Femenino , Humanos , Leucocitos/metabolismo , Masculino , Ratones , Persona de Mediana Edad , Monocitos/efectos de los fármacos , Monocitos/fisiología , Infiltración Neutrófila/efectos de los fármacos , Fragmentos de Péptidos/farmacología , Fibrosis Pulmonar/inducido químicamente , Fibrosis Pulmonar/patología , Esclerodermia Sistémica/patología , Transducción de Señal/fisiología , Adulto Joven
14.
Dev Biol ; 315(2): 383-96, 2008 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-18261719

RESUMEN

Atrioventricular (AV) endocardium transforms into the cushion mesenchyme, the primordia of the valves and membranous septa, through epithelial-mesenchymal transformation (EMT). While bone morphogenetic protein (BMP)-2 is known to be critical for AV EMT, the role of BMP-2 in post-EMT AV valvulogenesis remains to be elucidated. To find BMP signaling loops, we first localized Type I BMP receptors (BMPRs), BMPR-1A (ALK3), -1B (ALK6) and ALK2 in AV cushion mesenchyme in stage-24 chick embryos. Based on the BMP receptor expression pattern, we examined the functional roles of BMP-2 and BMP signaling in post-EMT valvulogenesis by using stage-24 AV cushion mesenchymal cell aggregates cultured on 3D-collagen gels. Exogenous BMP-2 or constitutively active (ca) BMPR-1B (ALK6)-virus treatments induced migration of the mesenchymal cells into the collagen gels, whereas noggin, an antagonist of BMPs, or dominant-negative (dn) BMPR-1 B (ALK6)-virus treatments reduced cell migration from the mesenchymal cell aggregates. Exogenous BMP-2 or caBMPR-1B (ALK6) treatments significantly promoted expression of an extracellular matrix (ECM) protein, periostin, a known valvulogenic matrix maturation mediator, at both mRNA and protein levels, whereas periostin expression was repressed by adding noggin or dnBMPR-1B (ALK6)-virus to the culture. Moreover, transcripts of Twist and Id1, which have been implicated in cell migration in embryogenesis and activation of the periostin promoter, were induced by BMP-2 but repressed by noggin in cushion mesenchymal cell cultures. These data provide evidence that BMP-2 and BMP signaling induce biological processes involved in early AV valvulogenesis, i.e. mesenchymal cell migration and expression of periostin, indicating critical roles for BMP signaling in post-EMT AV cushion tissue maturation and differentiation.


Asunto(s)
Proteínas Morfogenéticas Óseas/fisiología , Moléculas de Adhesión Celular/metabolismo , Cojinetes Endocárdicos/embriología , Factor de Crecimiento Transformador beta/fisiología , Animales , Proteínas Aviares/genética , Proteínas Aviares/metabolismo , Secuencia de Bases , Proteína Morfogenética Ósea 2 , Receptores de Proteínas Morfogenéticas Óseas de Tipo 1/genética , Receptores de Proteínas Morfogenéticas Óseas de Tipo 1/metabolismo , Proteínas Morfogenéticas Óseas/farmacología , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/fisiología , Movimiento Celular/efectos de los fármacos , Movimiento Celular/fisiología , Embrión de Pollo , Cartilla de ADN/genética , Células Madre Embrionarias/citología , Células Madre Embrionarias/efectos de los fármacos , Células Madre Embrionarias/metabolismo , Cojinetes Endocárdicos/citología , Cojinetes Endocárdicos/efectos de los fármacos , Cojinetes Endocárdicos/metabolismo , Hibridación in Situ , Proteína 1 Inhibidora de la Diferenciación/genética , Proteína 1 Inhibidora de la Diferenciación/metabolismo , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/metabolismo , Modelos Biológicos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Transducción de Señal , Proteínas Smad/metabolismo , Factor de Crecimiento Transformador beta/farmacología , Proteína 1 Relacionada con Twist/genética , Proteína 1 Relacionada con Twist/metabolismo
15.
Dev Biol ; 316(2): 200-13, 2008 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-18313657

RESUMEN

Cardiac valve leaflets develop from rudimentary structures termed endocardial cushions. These pre-valve tissues arise from a complex interplay of signals between the myocardium and endocardium whereby secreted cues induce the endothelial cells to transform into migratory mesenchyme through an endothelial to mesenchymal transformation (EMT). Even though much is currently known regarding the initial EMT process, the mechanisms by which these undifferentiated cushion mesenchymal tissues are remodeled "post-EMT" into mature fibrous valve leaflets remains one of the major, unsolved questions in heart development. Expression analyses, presented in this report, demonstrate that periostin, a component of the extracellular matrix, is predominantly expressed in post-EMT valve tissues and their supporting apparatus from embryonic to adult life. Analyses of periostin gene targeted mice demonstrate that it is within these regions that significant defects are observed. Periostin null mice exhibit atrial septal defects, structural abnormalities of the AV valves and their supporting tensile apparatus, and aberrant differentiation of AV cushion mesenchyme. Rescue experiments further demonstrate that periostin functions as a hierarchical molecular switch that can promote the differentiation of mesenchymal cells into a fibroblastic lineage while repressing their transformation into other mesodermal cell lineages (e.g. myocytes). This is the first report of an extracellular matrix protein directly regulating post-EMT AV valve differentiation, a process foundational and indispensable for the morphogenesis of a cushion into a leaflet.


Asunto(s)
Nodo Atrioventricular/embriología , Moléculas de Adhesión Celular/genética , Regulación del Desarrollo de la Expresión Génica , Válvulas Cardíacas/embriología , Corazón/embriología , Corazón/fisiología , Animales , Nodo Atrioventricular/ultraestructura , Moléculas de Adhesión Celular/deficiencia , Desarrollo Embrionario , Válvulas Cardíacas/ultraestructura , Ratones , Ratones Noqueados , Microscopía de Fuerza Atómica
16.
Biochem Biophys Res Commun ; 380(1): 33-8, 2009 Feb 27.
Artículo en Inglés | MEDLINE | ID: mdl-19150341

RESUMEN

Modulation of angiogenesis is a promising approach for treating a wide variety of human diseases including ischemic heart disease and cancer. In this study, we show that ADAM-17 is an important regulator of several key steps during angiogenesis. Knocking down ADAM-17 expression using lentivirus-delivered siRNA in HUVECs inhibited cell proliferation and the ability of cells to form close contact in two-dimensional cultures. Similarly, ADAM-17 depletion inhibited the ability of HUVECs to form capillary-like networks on top of three-dimensional Matrigel as well as in co-culture with fibroblasts within a three-dimensional scaffold. In mechanistic studies, both baseline and VEGF-induced MMP-2 activation and Matrigel invasion were inhibited by ADAM-17 depletion. Based on our findings we propose that ADAM-17 is part of a novel pro-angiogenic pathway leading to MMP-2 activation and vessel formation.


Asunto(s)
Proteínas ADAM/fisiología , Células Endoteliales/fisiología , Neovascularización Fisiológica , Proteínas ADAM/genética , Proteína ADAM17 , Proliferación Celular , Células Endoteliales/citología , Células Endoteliales/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Silenciador del Gen , Humanos , Metaloproteinasa 2 de la Matriz/metabolismo , Neovascularización Fisiológica/genética , Cordón Umbilical/citología , Factor A de Crecimiento Endotelial Vascular/farmacología
18.
FASEB Bioadv ; 1(3): 167-179, 2019 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-31482149

RESUMEN

The Insulin-like growth factor (IGF) system plays an important role in variety cellular biological functions; we previously reported levels of IGF binding proteins (IGFBP) -3 and -5 are increased in dermal and pulmonary fibrosis associated with the prototypic fibrosing disease systemic sclerosis (SSc), induce extracellular matrix (ECM) production, and promote fibrosis. We sought to examine the effects of another member of the family, IGFBP-4, on ECM production and fibrosis using cell-based, ex vivo organ culture and in vivo mouse lung fibrosis models. IGFBP-4 mRNA levels were significantly decreased in pulmonary fibroblasts of patients with SSc. ECM components were significantly reduced by endogenous and exogenous IGFBP-4. IGFBP-4 also blocked TGFß-induced ECM production, and inhibited ECM production ex vivo in human lung and skin in organ culture. In vivo, IGFBP-4 reduced bleomycin-induced collagen production and histologic evidence of fibrosis. Silencing IGFBP-4 expression to mimic levels observed in SSc lung fibroblasts resulted in increased ECM production. IGFBP-4 reduced mRNA and protein levels of the chemokine receptor CXCR4 and the pro-fibrotic factor CTGF. Further, CTGF silencing potentiated the anti-fibrotic effects of IGFBP-4. Reduced IGFBP-4 levels in SSc lung fibroblasts may contribute to the fibrotic phenotype via loss of IGFBP-4 anti-fibrotic activity.

19.
J Scleroderma Relat Disord ; 4(2): 127-136, 2019 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-35382388

RESUMEN

The potential value of mesenchymal stromal/stem cell therapy in treating skin fibrosis in scleroderma (systemic sclerosis) and of the caveolin-1 scaffolding domain peptide in treating lung, skin, and heart fibrosis is known. To understand how these observations may relate to differences between mesenchymal stromal/stem cells from healthy subjects and subjects with fibrosis, we have characterized the fibrogenic and adipogenic potential of adipose-derived mesenchymal stromal/stem cells from systemic sclerosis patients, from mice with fibrotic lung and skin disease induced by systemic bleomycin treatment, and from healthy controls. Early passage systemic sclerosis adipose-derived mesenchymal stromal/stem cells have a profibrotic/anti-adipogenic phenotype compared to healthy adipose-derived mesenchymal stromal/stem cells (low caveolin-1, high α-smooth muscle actin, high HSP47, low pAKT, low capacity for adipogenic differentiation). This phenotype is mimicked by treating healthy adipose-derived mesenchymal stromal/stem cells with transforming growth factor beta or caveolin-1 small interfering RNA and is reversed in systemic sclerosis adipose-derived mesenchymal stromal/stem cells by treatment with caveolin-1 scaffolding domain peptide, but not scrambled caveolin-1 scaffolding domain peptide. Similar results were obtained with adipose-derived mesenchymal stromal/stem cells from systemic sclerosis patients and from bleomycin-treated mice, indicating the central role of caveolin-1 in mesenchymal stromal/stem cell differentiation in fibrotic disease.

20.
PLoS One ; 13(12): e0207844, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30576317

RESUMEN

Dysregulation of the renin-angiotensin system leads to systemic hypertension and maladaptive fibrosis in various organs. We showed recently that myocardial fibrosis and the loss of cardiac function in mice with transverse aortic constriction (TAC) could be averted by treatment with the caveolin-1 scaffolding domain (CSD) peptide. Here, we used angiotensin II (AngII) infusion (2.1 mg/kg/day for 2 wk) in mice as a second model to confirm and extend our observations on the beneficial effects of CSD on heart and kidney disease. AngII caused cardiac hypertrophy (increased heart weight to body weight ratio (HW/BW) and cardiomyocyte cross-sectional area); fibrosis in heart and kidney (increased levels of collagen I and heat shock protein-47 (HSP47)); and vascular leakage (increased levels of IgG in heart and kidney). Echocardiograms of AngII-infused mice showed increased left ventricular posterior wall thickness (pWTh) and isovolumic relaxation time (IVRT), and decreased ejection fraction (EF), stroke volume (SV), and cardiac output (CO). CSD treatment (i.p. injections, 50 µg/mouse/day) of AngII-infused mice significantly suppressed all of these pathological changes in fibrosis, hypertrophy, vascular leakage, and ventricular function. AngII infusion increased ß1 and ß3 integrin levels and activated Pyk2 in both heart and kidney. These changes were also suppressed by CSD. Finally, bone marrow cell (BMC) isolated from AngII-infused mice showed hyper-migration toward SDF1. When AngII-infused mice were treated with CSD, BMC migration was reduced to the basal level observed in cells from control mice. Importantly, CSD did not affect the AngII-induced increase in blood pressure (BP), indicating that the beneficial effects of CSD were not mediated via normalization of BP. These results strongly indicate that CSD suppresses AngII-induced pathological changes in mice, suggesting that CSD can be developed as a treatment for patients with hypertension and pressure overload-induced heart failure.


Asunto(s)
Angiotensina II/administración & dosificación , Caveolina 1/administración & dosificación , Corazón/efectos de los fármacos , Riñón/efectos de los fármacos , Riñón/patología , Miocardio/patología , Fragmentos de Péptidos/administración & dosificación , Angiotensina II/fisiología , Angiotensinas/antagonistas & inhibidores , Animales , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/fisiología , Permeabilidad Capilar/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Fibrosis/etiología , Fibrosis/patología , Fibrosis/prevención & control , Hipertrofia Ventricular Izquierda/etiología , Hipertrofia Ventricular Izquierda/patología , Hipertrofia Ventricular Izquierda/prevención & control , Masculino , Ratones , Ratones Endogámicos C57BL , Sistema Renina-Angiotensina/efectos de los fármacos , Sistema Renina-Angiotensina/fisiología , Transducción de Señal/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA