Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 302
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
PLoS Pathog ; 15(11): e1008014, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31703116

RESUMEN

Clostridium perfringens epsilon toxin (ETX) is responsible for causing the economically devastating disease, enterotoxaemia, in livestock. It is well accepted that ETX causes blood brain barrier (BBB) permeability, however the mechanisms involved in this process are not well understood. Using in vivo and in vitro methods, we determined that ETX causes BBB permeability in mice by increasing caveolae-dependent transcytosis in brain endothelial cells. When mice are intravenously injected with ETX, robust ETX binding is observed in the microvasculature of the central nervous system (CNS) with limited to no binding observed in the vasculature of peripheral organs, indicating that ETX specifically targets CNS endothelial cells. ETX binding to CNS microvasculature is dependent on MAL expression, as ETX binding to CNS microvasculature of MAL-deficient mice was not detected. ETX treatment also induces extravasation of molecular tracers including 376Da fluorescein salt, 60kDA serum albumin, 70kDa dextran, and 155kDA IgG. Importantly, ETX-induced BBB permeability requires expression of both MAL and caveolin-1, as mice deficient in MAL or caveolin-1 did not exhibit ETX-induced BBB permeability. Examination of primary murine brain endothelial cells revealed an increase in caveolae in ETX-treated cells, resulting in dynamin and lipid raft-dependent vacuolation without cell death. ETX-treatment also results in a rapid loss of EEA1 positive early endosomes and accumulation of large, RAB7-positive late endosomes and multivesicular bodies. Based on these results, we hypothesize that ETX binds to MAL on the apical surface of brain endothelial cells, causing recruitment of caveolin-1, triggering caveolae formation and internalization. Internalized caveolae fuse with early endosomes which traffic to late endosomes and multivesicular bodies. We believe that these multivesicular bodies fuse basally, releasing their contents into the brain parenchyma.


Asunto(s)
Toxinas Bacterianas/farmacología , Barrera Hematoencefálica/fisiopatología , Encéfalo/fisiopatología , Caveolina 1/fisiología , Permeabilidad de la Membrana Celular/fisiología , Proteínas Proteolipídicas Asociadas a Mielina y Linfocito/fisiología , Transcitosis/efectos de los fármacos , Animales , Barrera Hematoencefálica/efectos de los fármacos , Encéfalo/efectos de los fármacos , Caveolas/efectos de los fármacos , Caveolas/metabolismo , Permeabilidad de la Membrana Celular/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
2.
Arterioscler Thromb Vasc Biol ; 40(6): 1510-1522, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32349535

RESUMEN

OBJECTIVE: Endothelial Cav-1 (caveolin-1) expression plays a relevant role during atherogenesis by controlling NO production, vascular inflammation, LDL (low-density lipoprotein) transcytosis, and extracellular matrix remodeling. Additional studies have identified cholesterol-rich membrane domains as important regulators of autophagy by recruiting ATGs (autophagy-related proteins) to the plasma membrane. Here, we investigate how the expression of Cav-1 in the aortic endothelium influences autophagy and whether enhanced autophagy contributes to the atheroprotective phenotype observed in Cav-1-deficient mice. Approach and Results: To analyze the impact of Cav-1 deficiency on regulation of autophagy in the aortic endothelium during the progression of atherosclerosis, we fed Ldlr-/- and Cav-1-/-Ldlr-/- mice a Western diet and assessed autophagy in the vasculature. We observe that the absence of Cav-1 promotes autophagy activation in athero-prone areas of the aortic endothelium by enhancing autophagic flux. Mechanistically, we found that Cav-1 interacts with the ATG5-ATG12 complex and influences the cellular localization of autophagosome components in lipid rafts, which controls the autophagosome formation and autophagic flux. Pharmacological inhibition of autophagy attenuates the atheroprotection observed in Cav-1-/- mice by increasing endothelial inflammation and macrophage recruitment, identifying a novel molecular mechanism by which Cav-1 deficiency protects against the progression of atherosclerosis. CONCLUSIONS: These results identify Cav-1 as a relevant regulator of autophagy in the aortic endothelium and demonstrate that pharmacological suppression of autophagic flux in Cav-1-deficient mice attenuates the atheroprotection observed in Cav-1-/- mice. Additionally, these findings suggest that activation of endothelial autophagy by blocking Cav-1 might provide a potential therapeutic strategy for cardiovascular diseases including atherosclerosis.


Asunto(s)
Aterosclerosis/prevención & control , Autofagia/fisiología , Caveolina 1/deficiencia , Endotelio Vascular/fisiopatología , Vasculitis/prevención & control , Adenina/análogos & derivados , Adenina/farmacología , Animales , Aorta/patología , Aorta/fisiopatología , Aorta/ultraestructura , Aterosclerosis/etiología , Autofagia/efectos de los fármacos , Caveolina 1/análisis , Caveolina 1/fisiología , Dieta Occidental , Células Endoteliales/química , Células Endoteliales/fisiología , Células Endoteliales/ultraestructura , Endotelio Vascular/química , Endotelio Vascular/ultraestructura , Femenino , Humanos , Masculino , Microdominios de Membrana/química , Microdominios de Membrana/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Células 3T3 NIH , Receptores de LDL/deficiencia
3.
Dev Biol ; 445(2): 271-279, 2019 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-30476483

RESUMEN

Local transplantation of stem cells has therapeutic effects on skin damage but cannot provide satisfactory wound healing. Studies on the mechanisms underlying the therapeutic effects of stem cells on skin wound healing will be needed. Hence, in the present study, we explored the role of Caveolin-1 in epidermal stem cells (EpiSCs) in the modulation of wound healing. We first isolated EpiSCs from mouse skin tissues and established stable EpiSCs with overexpression of Caveolin-1 using a lentiviral construct. We then evaluated the epidermal growth factor (EGF)-induced cell proliferation ability using cell counting Kit-8 (CCK-8) assay and assessed EpiSC pluripotency by examining Nanog mRNA levels in EpiSCs. Furthermore, we treated mice with skin burn injury using EpiSCs with overexpression of Caveolin-1. Histological examinations were conducted to evaluate re-epithelialization, wound scores, cell proliferation and capillary density in wounds. We found that overexpression of Caveolin-1 in EpiSCs promoted EGF-induced cell proliferation ability and increased wound closure in a mouse model of skin burn injury. Histological evaluation demonstrated that overexpression of Caveolin-1 in EpiSCs promoted re-epithelialization in wounds, enhanced cellularity, and increased vasculature, as well as increased wound scores. Taken together, our results suggested that Caveolin-1 expression in the EpiSCs play a critical role in the regulation of EpiSC proliferation ability and alteration of EpiSC proliferation ability may be an effective approach in promoting EpiSC-based therapy in skin wound healing.


Asunto(s)
Caveolina 1/fisiología , Cicatrización de Heridas/fisiología , Animales , Quemaduras/genética , Quemaduras/patología , Quemaduras/fisiopatología , Caveolina 1/antagonistas & inhibidores , Caveolina 1/genética , Proliferación Celular/genética , Proliferación Celular/fisiología , Células Epidérmicas/patología , Células Epidérmicas/fisiología , Femenino , Expresión Génica , Técnicas de Silenciamiento del Gen , Masculino , Ratones , Ratones Endogámicos BALB C , Neovascularización Fisiológica/genética , Ratas , Repitelización/genética , Repitelización/fisiología , Células Madre/patología , Células Madre/fisiología , Regulación hacia Arriba , Cicatrización de Heridas/genética
4.
Circulation ; 140(3): 225-239, 2019 07 16.
Artículo en Inglés | MEDLINE | ID: mdl-31154825

RESUMEN

BACKGROUND: Atherosclerosis is driven by synergistic interactions between pathological, biomechanical, inflammatory, and lipid metabolic factors. Our previous studies demonstrated that absence of caveolin-1 (Cav1)/caveolae in hyperlipidemic mice strongly inhibits atherosclerosis, which was attributed to activation of endothelial nitric oxide (NO) synthase (eNOS) and increased production of NO and reduced inflammation and low-density lipoprotein trafficking. However, the contribution of eNOS activation and NO production in the athero-protection of Cav1 and the exact mechanisms by which Cav1/caveolae control the pathogenesis of diet-induced atherosclerosis are still not clear. METHODS: Triple-knockout mouse lacking expression of eNOS, Cav1, and Ldlr were generated to explore the role of NO production in Cav1-dependent athero-protective function. The effects of Cav1 on lipid trafficking, extracellular matrix remodeling, and vascular inflammation were studied both in vitro and in vivo with a mouse model of diet-induced atherosclerosis. The expression of Cav1 and distribution of caveolae regulated by flow were analyzed by immunofluorescence staining and transmission electron microscopy. RESULTS: We found that absence of Cav1 significantly suppressed atherogenesis in Ldlr-/-eNOS-/- mice, demonstrating that athero-suppression is independent of increased NO production. Instead, we find that the absence of Cav1/caveolae inhibited low-density lipoprotein transport across the endothelium and proatherogenic fibronectin deposition and disturbed flow-mediated endothelial cell inflammation. Consistent with the idea that Cav1/caveolae may play a role in early flow-dependent inflammatory priming, distinct patterns of Cav1 expression and caveolae distribution were observed in athero-prone and athero-resistant areas of the aortic arch even in wild-type mice. CONCLUSIONS: These findings support a role for Cav1/caveolae as a central regulator of atherosclerosis that links biomechanical, metabolic, and inflammatory pathways independently of endothelial eNOS activation and NO production.


Asunto(s)
Aterosclerosis/metabolismo , Caveolina 1/fisiología , Endotelio Vascular/metabolismo , Lipoproteínas LDL/metabolismo , Óxido Nítrico Sintasa de Tipo III/metabolismo , Transcitosis/fisiología , Animales , Aterosclerosis/patología , Aterosclerosis/prevención & control , Células Cultivadas , Perros , Endotelio Vascular/patología , Activación Enzimática/fisiología , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos
5.
Biochem Biophys Res Commun ; 525(2): 313-318, 2020 04 30.
Artículo en Inglés | MEDLINE | ID: mdl-32087973

RESUMEN

For achieving efficient cancer treatment, it is important to elucidate the mechanism responsible for the accumulation of nanoparticles in tumor tissue. Recent studies suggest that nanoparticles are not delivered merely through gaps between tumor endothelial cells. We previously reported that the maturation of the vascular structure by the vascular endothelial cell growth factor receptor 2 (VEGFR2) using a previously developed siRNA delivery technology (RGD-MEND) significantly enhanced the accumulation of nanoparticles in types of cancers that area vessel-rich (renal cell carcinoma). This result was completely inconsistent with the generally accepted theory of the enhanced permeability and retention (EPR) effect. We hypothesized that a caveolin-1 (Cav1)-mediated transcellular route would be involved with the penetration of nanoparticles into tumor vasculature. To reveal the exact mechanism responsible for this enhancement, we observed the delivery of long-circulating liposomes (LPs) after Cav1 was co-suppressed by RGD-MEND with VEGFR2. The enhanced delivery of LPs by siRNA against VEGFR2 (siVEGFR2) was accompanied by the elevated expression of the Cav1 protein. In addition, Cav1 knockdown by siRNA against Cav1 (siCav1) canceled the enhanced delivery of LPs by siVEGFR2. The injection of siCav1 had no effect on the formation of alpha smooth muscle actin or vascular endothelial cell adhesion molecules. These results suggest that a Cav1-induced transcellular route and not a paracellular route, at least partially, contributes to the accumulation of nanoparticles in tumors.


Asunto(s)
Caveolina 1/fisiología , Liposomas/metabolismo , Neoplasias/metabolismo , Transcitosis , Animales , Carcinoma de Células Renales/irrigación sanguínea , Carcinoma de Células Renales/metabolismo , Caveolina 1/efectos de los fármacos , Humanos , Nanopartículas/metabolismo , Neoplasias/irrigación sanguínea , ARN Interferente Pequeño/farmacología , Receptor 2 de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores
6.
Exp Dermatol ; 29(2): 177-183, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31769542

RESUMEN

Expression of caveolin-1 (Cav-1) is an important pathophysiological factor in acne. Cav-1 strongly interacts with such well-recognized etiopathogenic factors such as hyperseborrhea, follicular hyperkeratinization and pathogenicity of Cutibacterium acnes. Cav-1 is a strong negative regulator of transforming growth factor beta (TGF-ß) expression. It acts as a critical determinant of autophagy, which is significantly induced in acne lesions through C. acnes and by absorption of fatty acids. Cav-1 also demonstrates different correlations with the development of innate immunity. We propose that normalization of Cav-1 expression can serve as a target in anti-acne therapy.


Asunto(s)
Acné Vulgar/metabolismo , Caveolina 1/fisiología , Células Epiteliales/fisiología , Acné Vulgar/tratamiento farmacológico , Acné Vulgar/fisiopatología , Adipocitos/metabolismo , Autofagia , Caveolina 1/metabolismo , Ácidos Grasos/metabolismo , Interacciones Huésped-Patógeno , Humanos , Inmunidad Innata , Glándulas Sebáceas/patología , Glándulas Sebáceas/fisiopatología
7.
FASEB J ; 33(6): 7545-7554, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30894019

RESUMEN

Interventions that preserve motor neurons or restore functional motor neuroplasticity may extend longevity in amyotrophic lateral sclerosis (ALS). Delivery of neurotrophins may potentially revive degenerating motor neurons, yet this approach is dependent on the proper subcellular localization of neurotrophin receptor (NTR) to plasmalemmal signaling microdomains, termed membrane/lipid rafts (MLRs). We previously showed that overexpression of synapsin-driven caveolin-1 (Cav-1) (SynCav1) increases MLR localization of NTR [e.g., receptor tyrosine kinase B (TrkB)], promotes hippocampal synaptic and neuroplasticity, and significantly improves learning and memory in aged mice. The present study crossed a SynCav1 transgene-positive (SynCav1+) mouse with the mutant human superoxide dismutase glycine to alanine point mutation at amino acid 93 (hSOD1G93A) mouse model of ALS. When compared with hSOD1G93A, hSOD1G93A/SynCav1+ mice exhibited greater body weight and longer survival as well as better motor function. Microscopic analyses of hSOD1G93A/SynCav1+ spinal cords revealed preserved spinal cord α-motor neurons and preserved mitochondrial morphology. Moreover, hSOD1G93A/SynCav1+ spinal cords contained more MLRs (cholera toxin subunit B positive) and MLR-associated TrkB and Cav-1 protein expression. These findings demonstrate that SynCav1 delays disease progression in a mouse model of ALS, potentially by preserving or restoring NTR expression and localization to MLRs.-Sawada, A., Wang, S., Jian, M., Leem, J., Wackerbarth, J., Egawa, J., Schilling, J. M., Platoshyn, O., Zemljic-Harpf, A., Roth, D. M., Patel, H. H., Patel, P. M., Marsala, M., Head, B. P. Neuron-targeted caveolin-1 improves neuromuscular function and extends survival in SOD1G93A mice.


Asunto(s)
Caveolina 1/fisiología , Músculo Esquelético/fisiología , Fenómenos Fisiológicos del Sistema Nervioso , Superóxido Dismutasa-1/genética , Animales , Peso Corporal , Caveolina 1/metabolismo , Estimulación Eléctrica , Humanos , Longevidad , Masculino , Ratones , Ratones Transgénicos , Neuronas Motoras/citología , Tasa de Supervivencia
8.
Arterioscler Thromb Vasc Biol ; 39(4): 754-764, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30786746

RESUMEN

Objective- Arteriovenous fistulae (AVF) are the most common access created for hemodialysis; however, many AVF fail to mature and require repeated intervention, suggesting a need to improve AVF maturation. Eph-B4 (ephrin type-B receptor 4) is the embryonic venous determinant that is functional in adult veins and can regulate AVF maturation. Cav-1 (caveolin-1) is the major scaffolding protein of caveolae-a distinct microdomain that serves as a mechanosensor at the endothelial cell membrane. We hypothesized that Cav-1 function is critical for Eph-B4-mediated AVF maturation. Approach and Results- In a mouse aortocaval fistula model, both Cav-1 mRNA and protein were increased in the AVF compared with control veins. Cav-1 KO (knockout) mice showed increased fistula wall thickening ( P=0.0005) and outward remodeling ( P<0.0001), with increased eNOS (endothelial NO synthase) activity compared with WT (wild type) mice. Ephrin-B2/Fc inhibited AVF outward remodeling in WT mice but not in Cav-1 KO mice and was maintained in Cav-1 RC (Cav-1 endothelial reconstituted) mice (WT, P=0.0001; Cav-1 KO, P=0.7552; Cav-1 RC, P=0.0002). Cavtratin-a Cav-1 scaffolding domain peptide-decreased AVF wall thickness in WT mice and in Eph-B4 het mice compared with vehicle alone (WT, P=0.0235; Eph-B4 het, P=0.0431); cavtratin also increased AVF patency (day 42) in WT mice ( P=0.0275). Conclusions- Endothelial Cav-1 mediates Eph-B4-mediated AVF maturation. The Eph-B4-Cav-1 axis regulates adaptive remodeling during venous adaptation to the fistula environment. Manipulation of Cav-1 function may be a translational strategy to enhance AVF patency.


Asunto(s)
Derivación Arteriovenosa Quirúrgica , Caveolina 1/fisiología , Receptor EphB4/fisiología , Transducción de Señal/fisiología , Vena Cava Inferior/fisiología , Animales , Aorta Abdominal/cirugía , Caveolas/metabolismo , Caveolina 1/biosíntesis , Caveolina 1/deficiencia , Caveolina 1/genética , Caveolina 1/farmacología , Células Cultivadas , Evaluación Preclínica de Medicamentos , Hemorreología , Humanos , Pulmón/citología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo III/antagonistas & inhibidores , Óxido Nítrico Sintasa de Tipo III/fisiología , Fragmentos de Péptidos/farmacología , Remodelación Vascular/fisiología , Vena Cava Inferior/cirugía
9.
Proc Natl Acad Sci U S A ; 114(40): 10737-10742, 2017 10 03.
Artículo en Inglés | MEDLINE | ID: mdl-28923916

RESUMEN

Ocular neovascularization is a devastating pathology of numerous ocular diseases and is a major cause of blindness. Caveolin-1 (Cav-1) plays important roles in the vascular system. However, little is known regarding its function and mechanisms in ocular neovascularization. Here, using comprehensive model systems and a cell permeable peptide of Cav-1, cavtratin, we show that Cav-1 is a critical player in ocular neovascularization. The genetic deletion of Cav-1 exacerbated and cavtratin administration inhibited choroidal and retinal neovascularization. Importantly, combined administration of cavtratin and anti-VEGF-A inhibited neovascularization more effectively than monotherapy, suggesting the existence of other pathways inhibited by cavtratin in addition to VEGF-A. Indeed, we found that cavtratin suppressed multiple critical components of pathological angiogenesis, including inflammation, permeability, PDGF-B and endothelial nitric oxide synthase expression (eNOS). Mechanistically, we show that cavtratin inhibits CNV and the survival and migration of microglia and macrophages via JNK. Together, our data demonstrate the unique advantages of cavtratin in antiangiogenic therapy to treat neovascular diseases.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Anticuerpos Monoclonales/farmacología , Caveolina 1/fisiología , Neovascularización Coroidal/prevención & control , MAP Quinasa Quinasa 4/metabolismo , Fragmentos de Péptidos/farmacología , Neovascularización Retiniana/prevención & control , Animales , Caveolina 1/farmacología , Neovascularización Coroidal/metabolismo , Neovascularización Coroidal/patología , Quimioterapia Combinada , Humanos , Ratones Noqueados , Neovascularización Retiniana/metabolismo , Neovascularización Retiniana/patología , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores
10.
Cell Commun Signal ; 17(1): 37, 2019 04 17.
Artículo en Inglés | MEDLINE | ID: mdl-30995923

RESUMEN

BACKGROUND: We previously showed that caveolin-1 (cav-1), an integral membrane protein, is required for the synthesis of matrix proteins by glomerular mesangial cells (MC). In a previous study to understand how cav-1 is involved in regulating matrix production, we had identified significant upregulation of the antifibrotic protein follistatin in cav-1 knockout MC. Follistatin inhibits the profibrotic effects of several members of the transforming growth factor beta superfamily, in particular the activins. Here, we characterize the molecular mechanism through which cav-1 regulates the expression of follistatin. METHODS: Kidneys from cav-1 wild type and knockout (KO) mice were analyzed and primary cultures of MC from cav-1 wild-type and KO mice were utilized. FST promoter deletion constructs were generated to determine the region of the promoter important for mediating FST upregulation in cav-1 KO MC. siRNA-mediated down-regulation and overexpression of Sp1 in conjunction with luciferase activity assays, immunoprecipitation, western blotting and ChiP was used to assess the role of Sp1 in transcriptionally regulating FST expression. Pharmacologic kinase inhibitors and specific siRNA were used to determine the post-translational mechanism through which cav-1 affects Sp1 activity. RESULTS: Our results establish that follistatin upregulation occurs at the transcript level. We identified Sp1 as the critical transcription factor regulating activation of the FST promoter in cav-1 KO MC through binding to a region within 123 bp of the transcription start site. We further determined that the lack of cav-1 increases Sp1 nuclear levels and transcriptional activity. This occurred through increased phosphoinositide 3-kinase (PI3K) activity and downstream protein kinase C (PKC) zeta-mediated phosphorylation and activation of Sp1. CONCLUSIONS: These findings shed light on the transcriptional mechanism by which cav-1 represses the expression of a major antifibrotic protein, and can inform the development of novel antifibrotic treatment strategies.


Asunto(s)
Caveolina 1/fisiología , Folistatina/genética , Regulación de la Expresión Génica , Células Mesangiales/patología , Factor de Transcripción Sp1/metabolismo , Animales , Caveolina 1/genética , Caveolina 1/metabolismo , Núcleo Celular/metabolismo , Células Cultivadas , Fibrosis , Células Mesangiales/metabolismo , Ratones , Ratones Noqueados , Fosfatidilinositol 3-Quinasas/metabolismo , Regiones Promotoras Genéticas , Proteína Quinasa C/metabolismo , Transcripción Genética
11.
Exp Mol Pathol ; 111: 104315, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31629729

RESUMEN

Caveolin is a structural protein of flask-shaped invaginations of the plasma membrane termed as caveolae and is widely expressed on the endothelial cells, smooth muscle cells and fibroblasts in the different parts of the body including the lung tissues. The expression of caveolin-1 in the lung tissues is important to prevent the fibrogenic actions of TGF-ß1 in lung fibrosis of different etiology including idiopathic pulmonary fibrosis, systemic sclerosis-associated interstitial lung disease and allergen-induced airway remodeling. Caveolin-1-mediated internalization and degradation of TGF-ß1 receptors may possibly account for the decreased actions of TGF-ß1. Studies have shown that the deficiency of caveolin-1 is very important in inducing lung fibrosis and its upregulation is reported to prevent lung fibrosis. The biological actions of caveolin-1 involve signaling pathways including JNK signaling, IL-4, STAT-3, miR199a-5p, CXCR4+ and CXCL12. The present review discusses the key role of caveolin and associated signaling pathways in the pathogenesis of lung fibrosis of different etiology.


Asunto(s)
Caveolina 1/fisiología , Fibrosis Pulmonar/etiología , Remodelación de las Vías Aéreas (Respiratorias)/fisiología , Animales , Citocinas/metabolismo , Epigénesis Genética , Humanos , Fibrosis Pulmonar Idiopática/etiología , Fibrosis Pulmonar Idiopática/patología , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Fibrosis Pulmonar/patología , Factores de Transcripción STAT/metabolismo , Transducción de Señal
12.
Blood ; 127(15): 1930-9, 2016 Apr 14.
Artículo en Inglés | MEDLINE | ID: mdl-26837700

RESUMEN

Caveolin-1 (Cav-1) is a key organizer of membrane specializations and a scaffold protein that regulates signaling in multiple cell types. We found increased Cav-1 expression in human and murine T cells after allogeneic hematopoietic cell transplantation. Indeed, Cav-1(-/-)donor T cells caused less severe acute graft-versus-host disease (GVHD) and yielded higher numbers of regulatory T cells (Tregs) compared with controls. Depletion of Tregs from the graft abrogated this protective effect. Correspondingly, Treg frequencies increased when Cav-1(-/-)T cells were exposed to transforming growth factor-ß/T-cell receptor (TCR)/CD28 activation or alloantigen stimulation in vitro compared with wild-type T cells. Mechanistically, we found that the phosphorylation of Cav-1 is dispensable for the control of T-cell fate by using a nonphosphorylatable Cav-1 (Y14F/Y14F) point-mutation variant. Moreover, the close proximity of lymphocyte-specific protein tyrosine kinase (Lck) to the TCR induced by TCR-activation was reduced in Cav-1(-/-)T cells. Therefore, less TCR/Lck clustering results in suboptimal activation of the downstream signaling events, which correlates with the preferential development into a Treg phenotype. Overall, we report a novel role for Cav-1 in TCR/Lck spatial distribution upon TCR triggering, which controls T-cell fate toward a regulatory phenotype. This alteration translated into a significant increase in the frequency of Tregs and reduced GVHD in vivo.


Asunto(s)
Caveolina 1/metabolismo , Caveolina 1/fisiología , Regulación de la Expresión Génica , Receptores de Antígenos de Linfocitos T/metabolismo , Linfocitos T/citología , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Antígenos CD28/metabolismo , Linfocitos T CD4-Positivos/citología , Caveolina 1/genética , Diferenciación Celular , Factores de Transcripción Forkhead/metabolismo , Enfermedad Injerto contra Huésped/inmunología , Trasplante de Células Madre Hematopoyéticas , Humanos , Activación de Linfocitos/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Fosforilación , Estudios Prospectivos , Transducción de Señal , Linfocitos T Reguladores/citología , Factor de Crecimiento Transformador beta/metabolismo , Trasplante Homólogo
13.
J Surg Res ; 232: 171-178, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30463715

RESUMEN

BACKGROUND: Myocardial infarction (MI) is a major etiology for the development of heart failure. We have previously shown that high molecular weight polyethylene glycol (PEG) can protect cardiac myocytes from hypoxia-reoxygenation injury in vitro. In this study, we investigated the potential protective effects of 15-20 kD PEG postinfarction without reperfusion. METHODS: One milliliter of PEG 15-20 was delivered intravenously following permanent left anterior descending ligation in adult male rats with phosphate buffer saline (PBS) as control (n = 9 in each group). Echocardiography was performed at baseline and at 8 wk post-MI. Left ventricles (LVs) were harvested to quantify fibrosis, apoptosis, cell survival signaling, regulation of ß-adrenergic signaling, and caveolin (Cav) expression. RESULTS: The PEG group had significant recovery of LV function at 8 wk compared with the PBS group. There was less LV fibrosis in both the infarct and remote territory. Cell survival signaling was upregulated in the PEG group with increased Akt and ERK phosphorylation. PEG inhibited apoptosis as measured by terminal deoxynucleotidyl transferase [TdT]-mediated dUTP nick-end labeling positive nuclei and caspase-3 activity. There was maintenance of Cav-1, Cav-2, and Cav-3 expression following PEG treatment versus a decline in the PBS group. Negative regulators of ß-adrenergic signaling, G protein-coupled receptor kinase-2, and ß-arrestin 1 and 2 were all upregulated in PBS-treated samples compared to normal control; however, PEG treatment led to decreased expression. CONCLUSIONS: These data suggest that PEG 15-20 may have significant protective effects post-MI even in the setting of no acute reperfusion. Upregulation of Cav expression appears to be a key mechanism for the beneficial effects of PEG on ventricular remodeling and function.


Asunto(s)
Infarto del Miocardio/fisiopatología , Polietilenglicoles/farmacología , Remodelación Ventricular/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Caspasa 3/metabolismo , Caveolina 1/análisis , Caveolina 1/fisiología , Masculino , Peso Molecular , Ratas , Ratas Sprague-Dawley , Receptores Adrenérgicos beta/fisiología , Transducción de Señal/efectos de los fármacos , Función Ventricular Izquierda
14.
Cell Mol Biol (Noisy-le-grand) ; 64(1): 40-46, 2018 Jan 31.
Artículo en Inglés | MEDLINE | ID: mdl-29412792

RESUMEN

Our study aims to access the influence of caveolin1 (CAV1) on ß cell expression profiles. We knocked down the expression of CAV1 in both NIT-1 cells and islets isolated from C57BL/6J mice using an RNA interference technique, which was realized by the transfer of an shRNA vector targeting CAV1 mRNA into NIT-1 cells or islets through latent virus infection. First, we identified the change in gene expression profiles in islets, in which the CAV1 expression level was down-regulated, as ascertained by mouse gene expression microarray, and the results showed that pathways related to ß cell proliferation and pancreatic secretion functions were significantly influenced. The results of MTT demonstrated that the knockdown of CAV1 expression in NIT-1 cells promoted proliferation. The protein array results showed that pro-apoptotic cytokines were down-regulated in the NIT-1 cell line with CAV1 knockdown. These findings suggest that CAV1 might be involved in apoptosis and proliferation regulation in ß cells, and therefore could be a potential target for the development of novel therapies for diabetes mellitus.


Asunto(s)
Caveolina 1/fisiología , Células Secretoras de Insulina/metabolismo , Animales , Apoptosis/genética , Autofagia/genética , Caveolina 1/genética , Línea Celular , Proliferación Celular , Células Secretoras de Insulina/citología , Islotes Pancreáticos/metabolismo , Ratones Endogámicos C57BL , Interferencia de ARN , Transcriptoma
15.
J Biochem Mol Toxicol ; 32(10): e22202, 2018 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-30088837

RESUMEN

Anoikis resistance is considered to be an essential prerequisite of tumor metastasis and which is an important mechanism in the metastatic process of gastric cancer (GC). Caveolin-1 (CAV-1), a protein component of caveolae, has been reported to regulate several cancer cell behaviors including anoikis resistance. However, the role of CAV-1 in the acquisition of anoikis resistance in GC cells has never been explored. In this study, we investigated the promoting effect of CAV-1 on anchorage-independent growth and anoikis resistance, and the involvement of the related signaling pathways in such process in SGC-7901 cells. The results showed that CAV-1 could promote anchorage-independent growth and anoikis resistance in detached SGC-7901 cells, which was associated with the activation of Src-dependent epidermal growth factor receptor-integrin ß signaling as well as the phosphorylation of PI3K/Akt and MEK/ERK signaling pathways. The data from this study might contribute to the in-depth understanding of the metastatic mechanism for GC.


Asunto(s)
Anoicis/fisiología , Caveolina 1/fisiología , Integrina beta1/metabolismo , Proteínas Proto-Oncogénicas pp60(c-src)/metabolismo , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patología , Adhesión Celular , Línea Celular Tumoral , Receptores ErbB/metabolismo , Humanos , Sistema de Señalización de MAP Quinasas , Metástasis de la Neoplasia , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal
16.
Acta Pharmacol Sin ; 39(5): 669-682, 2018 May.
Artículo en Inglés | MEDLINE | ID: mdl-29595191

RESUMEN

Reactive nitrogen species (RNS) play important roles in mediating cerebral ischemia-reperfusion injury. RNS activate multiple signaling pathways and participate in different cellular events in cerebral ischemia-reperfusion injury. Recent studies have indicated that caveolin-1 and matrix metalloproteinase (MMP) are important signaling molecules in the pathological process of ischemic brain injury. During cerebral ischemia-reperfusion, the production of nitric oxide (NO) and peroxynitrite (ONOO-), two representative RNS, down-regulates the expression of caveolin-1 (Cav-1) and, in turn, further activates nitric oxide synthase (NOS) to promote RNS generation. The increased RNS further induce MMP activation and mediate disruption of the blood-brain barrier (BBB), aggravating the brain damage in cerebral ischemia-reperfusion injury. Therefore, the feedback interaction among RNS/Cav-1/MMPs provides an amplified mechanism for aggravating ischemic brain damage during cerebral ischemia-reperfusion injury. Targeting the RNS/Cav-1/MMP pathway could be a promising therapeutic strategy for protecting against cerebral ischemia-reperfusion injury. In this mini-review article, we highlight the important role of the RNS/Cav-1/MMP signaling cascades in ischemic stroke injury and review the current progress of studies seeking therapeutic compounds targeting the RNS/Cav-1/MMP signaling cascades to attenuate cerebral ischemia-reperfusion injury. Several representative natural compounds, including calycosin-7-O-ß-D-glucoside, baicalin, Momordica charantia polysaccharide (MCP), chlorogenic acid, lutein and lycopene, have shown potential for targeting the RNS/Cav-1/MMP signaling pathway to protect the brain in ischemic stroke. Therefore, the RNS/Cav-1/MMP pathway is an important therapeutic target in ischemic stroke treatment.


Asunto(s)
Isquemia Encefálica/prevención & control , Fármacos Neuroprotectores/uso terapéutico , Daño por Reperfusión/prevención & control , Transducción de Señal/efectos de los fármacos , Accidente Cerebrovascular/prevención & control , Animales , Isquemia Encefálica/fisiopatología , Caveolina 1/metabolismo , Caveolina 1/fisiología , Humanos , Metaloproteinasas de la Matriz/metabolismo , Metaloproteinasas de la Matriz/fisiología , Momordica charantia/química , Especies de Nitrógeno Reactivo/metabolismo , Especies de Nitrógeno Reactivo/fisiología , Accidente Cerebrovascular/fisiopatología
17.
Endocr J ; 65(9): 893-901, 2018 Sep 27.
Artículo en Inglés | MEDLINE | ID: mdl-29877208

RESUMEN

Hashimoto's thyroiditis (HT) is considered a T helper-type 1 (Th1) cytokine-dominant autoimmune thyroid disease. Caveolin-1 (Cav-1), a part of the thyroxisome multiprotein complex, is localized at the apical pole of thyrocytes and is indispensable for synthesis of thyroid hormones and modulation of oxidative stress in order to avoid cell damage and apoptosis. Reduced autophagy induces thyroid follicular cells (TFC) apoptosis by activating reactive oxygen species (ROS) in HT patients. Nevertheless, whether Cav-1 has roles in the regulation of autophagy remains largely unclear. In this study, we examined Th1 cytokines and Cav-1 expression in HT thyroid tissues, determined the effects of interleukin-1beta (IL-1ß) and interferon-gamma (IFN-γ) on Cav-1 and autophagy activity in TFC, and investigated the association between Cav-1 and autophagy activity in vitro. Our results indicate that higher levels of IL-1ß and IFN-γ and lower levels of Cav-1 were expressed in thyroid tissues of HT patients than in those of normal controls. Cav-1 mRNA and protein levels were significantly decreased in TFC exposed to IL-1ß and IFN-γ, accompanied by decreased expression of autophagy-related protein LC3B-II. Interestingly, small interfering RNA (siRNA)-mediated Cav-1 knockdown in TFC reduced LC3B-II protein expression. Taken together, these results suggest that lack of Cav-1 expression inhibited autophagy activity in TFC exposed to Th1 cytokines (IL-1ß and IFN-γ), which might be a novel pathogenetic mechanism of HT.


Asunto(s)
Autofagia/genética , Caveolina 1/fisiología , Enfermedad de Hashimoto/genética , Células Epiteliales Tiroideas/fisiología , Autofagia/efectos de los fármacos , Estudios de Casos y Controles , Caveolina 1/genética , Células Cultivadas , Técnicas de Silenciamiento del Gen , Enfermedad de Hashimoto/metabolismo , Humanos , Interferón gamma/farmacología , Interleucina-1beta/farmacología , Células TH1/metabolismo , Células TH1/fisiología , Células Epiteliales Tiroideas/efectos de los fármacos , Células Epiteliales Tiroideas/metabolismo
18.
Curr Diab Rep ; 17(3): 19, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-28283950

RESUMEN

PURPOSE OF REVIEW: Diabetic nephropathy, a major microvascular complication of diabetes and the most common cause of end-stage renal disease, is characterized by prominent accumulation of extracellular matrix. The membrane microdomains caveolae, and their integral protein caveolin-1, play critical roles in the regulation of signal transduction. In this review we discuss current knowledge of the contribution of caveolin-1/caveolae to profibrotic signaling and the pathogenesis of diabetic kidney disease, and assess its potential as a therapeutic target. RECENT FINDINGS: Caveolin (cav)-1 is key to facilitating profibrotic signal transduction induced by several stimuli known to be pathogenic in diabetic nephropathy, including the most prominent factors hyperglycemia and angiotensin II. Phosphorylation of cav-1 on Y14 is an important regulator of these responses. In vivo studies support a pathogenic role for caveolae in the progression of diabetic nephropathy. Targeting caveolin-1/caveolae would enable inhibition of multiple profibrotic pathways, representing a novel and potentially potent therapeutic option for diabetic nephropathy.


Asunto(s)
Caveolina 1/fisiología , Nefropatías Diabéticas/etiología , Animales , Caveolas/fisiología , Caveolina 1/antagonistas & inhibidores , Nefropatías Diabéticas/tratamiento farmacológico , Nefropatías Diabéticas/fisiopatología , Matriz Extracelular/metabolismo , Humanos , Estrés Oxidativo , Transducción de Señal/fisiología
19.
World J Surg ; 41(10): 2625-2630, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28560511

RESUMEN

BACKGROUND: Caveolin-1 (CAV-1), the main scaffold protein in caveolae, is frequently deregulated in human cancer. Of importance, this protein has been described to show tumor suppressor or oncogenic properties depending on the cell type and the stage of the disease. In fact, its role in colorectal cancer (CRC) remains to be fully clarified due to discrepancies in the literature. METHODS: We analyzed CAV-1 by western blot in a set of early-stage CRC patients with paired tumor tissue and normal colonic mucosa available. CAV-1 mRNA and expression levels of miR-124, 133 and 802 were quantified by real-time PCR. RESULTS: We found CAV-1 strongly downregulated in 76.2% of tumor samples and associated with the subgroup of elderly patients (p = 0.027). We observed by real-time PCR a lack of correlation between CAV-1 mRNA and protein levels in some cases with CAV-1 downregulated by western blot, and miR-124 deregulation was identified as a potential contributing alteration to decrease CAV-1 protein expression. CONCLUSION: CAV-1 is commonly downregulated in patients with primary CRC, which suggests its tumor suppressor role in early stages of this disease. Moreover, based on our findings, the previous discrepancies observed in different studies to date could be due to a complex posttranscriptional CAV-1 regulation.


Asunto(s)
Caveolina 1/fisiología , Neoplasias Colorrectales/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Caveolina 1/análisis , Caveolina 1/genética , Neoplasias Colorrectales/patología , Regulación hacia Abajo , Femenino , Humanos , Masculino , MicroARNs/fisiología , Persona de Mediana Edad , Estadificación de Neoplasias , ARN Mensajero/análisis , Reacción en Cadena en Tiempo Real de la Polimerasa
20.
J Am Soc Nephrol ; 27(4): 1066-75, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26264854

RESUMEN

Nephrotic syndrome is a CKD defined by proteinuria with subsequent hypoalbuminemia, hyperlipidemia, and edema caused by impaired renal glomerular filtration barrier function. We previously identified mutations in epithelial membrane protein 2 (EMP2) as a monogenic cause of this disease. Here, we generated an emp2-knockout zebrafish model using transcription activator-like effector nuclease-based genome editing. We found that loss of emp2 in zebrafish upregulated caveolin-1 (cav1), a major component of caveolae, in embryos and adult mesonephric glomeruli and exacerbated podocyte injury. This phenotype was partially rescued by glucocorticoids. Furthermore, overexpression of cav1 in zebrafish podocytes was sufficient to induce the same phenotype observed in emp2 homozygous mutants, which was also treatable with glucocorticoids. Similarly, knockdown of EMP2 in cultured human podocytes resulted in increased CAV1 expression and decreased podocyte survival in the presence of puromycin aminonucleoside, whereas glucocorticoid treatment ameliorated this phenotype. Taken together, we have established excessive CAV1 as a mediator of the predisposition to podocyte injury because of loss of EMP2, suggesting CAV1 could be a novel therapeutic target in nephrotic syndrome and podocyte injury.


Asunto(s)
Caveolina 1/fisiología , Glicoproteínas de Membrana/fisiología , Podocitos/fisiología , Regulación hacia Arriba , Animales , Humanos , Síndrome Nefrótico/etiología , Pez Cebra
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA