Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 199
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Cardiovasc Diabetol ; 23(1): 138, 2024 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-38664801

RESUMEN

BACKGROUND: Neutral cholesterol ester hydrolase 1 (NCEH1) plays a critical role in the regulation of cholesterol ester metabolism. Deficiency of NCHE1 accelerated atherosclerotic lesion formation in mice. Nonetheless, the role of NCEH1 in endothelial dysfunction associated with diabetes has not been explored. The present study sought to investigate whether NCEH1 improved endothelial function in diabetes, and the underlying mechanisms were explored. METHODS: The expression and activity of NCEH1 were determined in obese mice with high-fat diet (HFD) feeding, high glucose (HG)-induced mouse aortae or primary endothelial cells (ECs). Endothelium-dependent relaxation (EDR) in aortae response to acetylcholine (Ach) was measured. RESULTS: Results showed that the expression and activity of NCEH1 were lower in HFD-induced mouse aortae, HG-exposed mouse aortae ex vivo, and HG-incubated primary ECs. HG exposure reduced EDR in mouse aortae, which was exaggerated by endothelial-specific deficiency of NCEH1, whereas NCEH1 overexpression restored the impaired EDR. Similar results were observed in HFD mice. Mechanically, NCEH1 ameliorated the disrupted EDR by dissociating endothelial nitric oxide synthase (eNOS) from caveolin-1 (Cav-1), leading to eNOS activation and nitric oxide (NO) release. Moreover, interaction of NCEH1 with the E3 ubiquitin-protein ligase ZNRF1 led to the degradation of Cav-1 through the ubiquitination pathway. Silencing Cav-1 and upregulating ZNRF1 were sufficient to improve EDR of diabetic aortas, while overexpression of Cav-1 and downregulation of ZNRF1 abolished the effects of NCEH1 on endothelial function in diabetes. Thus, NCEH1 preserves endothelial function through increasing NO bioavailability secondary to the disruption of the Cav-1/eNOS complex in the endothelium of diabetic mice, depending on ZNRF1-induced ubiquitination of Cav-1. CONCLUSIONS: NCEH1 may be a promising candidate for the prevention and treatment of vascular complications of diabetes.


Asunto(s)
Caveolina 1 , Dieta Alta en Grasa , Células Endoteliales , Endotelio Vascular , Ratones Endogámicos C57BL , Óxido Nítrico Sintasa de Tipo III , Vasodilatación , Animales , Masculino , Ratones , Aorta/enzimología , Aorta/fisiopatología , Aorta/metabolismo , Aorta/efectos de los fármacos , Aorta/patología , Caveolina 1/metabolismo , Caveolina 1/deficiencia , Caveolina 1/genética , Células Cultivadas , Diabetes Mellitus Experimental/enzimología , Diabetes Mellitus Experimental/fisiopatología , Células Endoteliales/enzimología , Células Endoteliales/metabolismo , Células Endoteliales/efectos de los fármacos , Endotelio Vascular/fisiopatología , Endotelio Vascular/metabolismo , Endotelio Vascular/enzimología , Endotelio Vascular/efectos de los fármacos , Ratones Noqueados , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo III/metabolismo , Obesidad/enzimología , Obesidad/fisiopatología , Obesidad/metabolismo , Transducción de Señal , Esterol Esterasa/metabolismo , Esterol Esterasa/genética , Ubiquitinación , Vasodilatación/efectos de los fármacos
2.
Tuberculosis (Edinb) ; 147: 102493, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38547568

RESUMEN

Tuberculosis, caused by Mycobacterium tuberculosis, remains one of the deadliest infections in humans. Because Mycobacterium bovis Bacillus Calmette-Guérin (BCG) share genetic similarities with Mycobacterium tuberculosis, it is often used as a model to elucidate the molecular mechanisms of more severe tuberculosis infection. Caveolin-1 has been implied in many physiological processes and diseases, but it's role in mycobacterial infections has barely been studied. We isolated macrophages from Wildtype or Caveolin-1 deficient mice and analyzed hallmarks of infection, such as internalization, induction of autophagy and apoptosis. For in vivo assays we intravenously injected mice with BCG and investigated tissues for bacterial load with colony-forming unit assays, bioactive lipids with mass spectrometry and changes of protein expressions by Western blotting. Our results revealed that Caveolin-1 was important for early killing of BCG infection in vivo and in vitro, controlled acid sphingomyelinase (Asm)-dependent ceramide formation, apoptosis and inflammatory cytokines upon infection with BCG. In accordance, Caveolin-1 deficient mice and macrophages showed higher bacterial burdens in the livers. The findings indicate that Caveolin-1 plays a role in infection of mice and murine macrophages with BCG, by controlling cellular apoptosis and inflammatory host response. These clues might be useful in the fight against tuberculosis.


Asunto(s)
Apoptosis , Caveolina 1 , Macrófagos , Ratones Endogámicos C57BL , Ratones Noqueados , Mycobacterium bovis , Esfingomielina Fosfodiesterasa , Tuberculosis , Animales , Caveolina 1/metabolismo , Caveolina 1/deficiencia , Caveolina 1/genética , Mycobacterium bovis/patogenicidad , Macrófagos/microbiología , Macrófagos/metabolismo , Tuberculosis/microbiología , Tuberculosis/inmunología , Tuberculosis/metabolismo , Tuberculosis/patología , Esfingomielina Fosfodiesterasa/metabolismo , Esfingomielina Fosfodiesterasa/deficiencia , Autofagia , Interacciones Huésped-Patógeno , Modelos Animales de Enfermedad , Carga Bacteriana , Citocinas/metabolismo , Ceramidas/metabolismo , Hígado/microbiología , Hígado/metabolismo , Hígado/patología , Células Cultivadas , Ratones , Mediadores de Inflamación/metabolismo , Factores de Tiempo
3.
J Cardiovasc Pharmacol ; 78(2): 175-183, 2021 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-34554674

RESUMEN

ABSTRACT: Atrial fibrillation (AF) is a common arrhythmia in the clinic. Ablation failure and recurrence after cardioversion have become medical problems worldwide. An important pathological feature of AF is atrial fibrosis, which increases susceptibility to AF. As an important target of fibrosis signal integration, the signal transducer and activator of transcription 3 (STAT3) signaling pathway plays an important role in fibrosis. Caveolin-1 (CAV1), a cell membrane protein, is involved in a variety of the biological functions of cells. However, the role of CAV1 in atrial fibrosis remains unclear. In this study, Masson's trichrome staining was used to detect the degree of atrial fibrosis, and the expression of CAV1 in the human atrium was evaluated by immunohistochemistry. To further study the role of CAV1, its expression in cultured rat atrial fibroblasts was silenced using siRNAs. Atrial fibroblasts were treated with angiotensin II to observe the effects on CAV1 and the transforming growth factor-ß1 and STAT3 signaling pathways. We also detected the effects of CAV1 scaffolding domain (CSD) peptide on fibrosis through the addition of exogenous CSD peptide. The results showed that CAV1 expression decreased with the aggravation of atrial fibrosis and that this effect increased the incidence of AF. The depletion of CAV1 induced excessive extracellular matrix deposition by activating the STAT3 and transforming growth factor-ß1/SMAD2 signaling pathways, and this effect was exacerbated by stimulation with angiotensin II and improved by CSD peptide. These data suggested that CAV1 not only plays a critical role in fibrosis progression but also provides a target for the treatment of atrial fibrosis and AF.


Asunto(s)
Fibrilación Atrial/metabolismo , Remodelación Atrial , Caveolina 1/deficiencia , Fibroblastos/metabolismo , Atrios Cardíacos/metabolismo , Frecuencia Cardíaca , Factor de Transcripción STAT3/metabolismo , Adulto , Anciano , Animales , Fibrilación Atrial/genética , Fibrilación Atrial/patología , Fibrilación Atrial/fisiopatología , Caveolina 1/genética , Células Cultivadas , Matriz Extracelular/metabolismo , Matriz Extracelular/patología , Femenino , Fibroblastos/patología , Fibrosis , Atrios Cardíacos/patología , Atrios Cardíacos/fisiopatología , Humanos , Masculino , Persona de Mediana Edad , Miofibroblastos/metabolismo , Miofibroblastos/patología , Ratas Sprague-Dawley , Factor de Transcripción STAT3/genética , Transducción de Señal
4.
Front Immunol ; 12: 713808, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34434195

RESUMEN

Polarization of hepatic macrophages plays a crucial role in the injury and repair processes of acute and chronic liver diseases. However, the underlying molecular mechanisms remain elusive. Caveolin-1 (Cav1) is the structural protein of caveolae, the invaginations of the plasma membrane. It has distinct functions in regulating hepatitis, cirrhosis, and hepatocarcinogenesis. Given the increasing number of cases of liver cancer, nonalcoholic steatohepatitis, and non-alcoholic fatty liver disease worldwide, investigations on the role of Cav1 in liver diseases are warranted. In this study, we aimed to investigate the role of Cav1 in the pathogenesis of acute liver injury. Wild-type (WT) and Cav1 knockout (KO) mice (Cav1tm1Mls) were injected with carbon tetrachloride (CCl4). Cav1 KO mice showed significantly reduced degeneration, necrosis, and apoptosis of hepatocytes and decreased level of alanine transaminase (ALT) compared to WT mice. Moreover, Cav1 was required for the recruitment of hepatic macrophages. The analysis of the mRNA levels of CD86, tumor necrosis factor (TNF), and interleukin (IL)-6, as well as the protein expression of inducible nitric oxide synthase (iNOS), indicated that Cav1 deficiency inhibited the polarization of hepatic macrophages towards the M1 phenotype in the injured liver. Consistent with in vivo results, the expressions of CD86, TNF, IL-6, and iNOS were significantly downregulated in Cav1 KO macrophages. Also, fluorescence-activated cell sorting (FACS) analysis showed that the proportion of M1 macrophages was significantly decreased in the liver tissues obtained from Cav1 KO mice following CCl4 treatment. In summary, our results showed that Cav1 deficiency protected mice against CCl4-induced acute liver injury by regulating polarization of hepatic macrophages. We provided direct genetic evidence that Cav1 expressed in hepatic macrophages contributed to the pathogenesis of acute liver injury by regulating the polarization of hepatic macrophages towards the M1 phenotype. These findings suggest that Cav1 expressed in macrophages may represent a potential therapeutic target for acute liver injury.


Asunto(s)
Tetracloruro de Carbono/efectos adversos , Caveolina 1/deficiencia , Enfermedad Hepática Inducida por Sustancias y Drogas/etiología , Enfermedad Hepática Inducida por Sustancias y Drogas/metabolismo , Macrófagos/inmunología , Macrófagos/metabolismo , Animales , Apoptosis/genética , Apoptosis/inmunología , Biomarcadores , Enfermedad Hepática Inducida por Sustancias y Drogas/patología , Citocinas/metabolismo , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades , Femenino , Hepatocitos/metabolismo , Hepatocitos/patología , Activación de Macrófagos/inmunología , Macrófagos/patología , Masculino , Ratones , Óxido Nítrico/metabolismo
5.
Theranostics ; 11(13): 6154-6172, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33995651

RESUMEN

SH2 domain containing tyrosine phosphatase 2 (Shp2; PTPN11) regulates several intracellular pathways downstream of multiple growth factor receptors. Our studies implicate that Shp2 interacts with Caveolin-1 (Cav-1) protein in retinal ganglion cells (RGCs) and negatively regulates BDNF/TrkB signaling. This study aimed to investigate the mechanisms underlying the protective effects of shp2 silencing in the RGCs in glaucomatous conditions. Methods: Shp2 was silenced in the Cav-1 deficient mice and the age matched wildtype littermates using adeno-associated viral (AAV) constructs. Shp2 expression modulation was performed in an acute and a chronic mouse model of experimental glaucoma. AAV2 expressing Shp2 eGFP-shRNA under a strong synthetic CAG promoter was administered intravitreally in the animals' eyes. The contralateral eye received AAV-eGFP-scramble-shRNA as control. Animals with Shp2 downregulation were subjected to either microbead injections or acute ocular hypertension experimental paradigm. Changes in inner retinal function were evaluated by measuring positive scotopic threshold response (pSTR) while structural and biochemical alterations were evaluated through H&E staining, western blotting and immunohistochemical analysis of the retinal tissues. Results: A greater loss of pSTR amplitudes was observed in the WT mice compared to Cav-1-/- retinas in both the models. Silencing of Shp2 phosphatase imparted protection against inner retinal function loss in chronic glaucoma model in WT mice. The functional rescue also translated to structural preservation of ganglion cell layer in the chronic glaucoma condition in WT mice which was not evident in Cav-1-/- mice retinas. Conclusions: This study indicates that protective effects of Shp2 ablation under chronic experimental glaucoma conditions are dependent on Cav-1 in the retina, suggesting in vivo interactions between the two proteins.


Asunto(s)
Caveolina 1/fisiología , Terapia Genética , Vectores Genéticos/uso terapéutico , Glaucoma/terapia , Proteína Tirosina Fosfatasa no Receptora Tipo 11/antagonistas & inhibidores , Retina/patología , alfa-Globulinas/genética , Animales , Apoptosis , Factor Neurotrófico Derivado del Encéfalo/fisiología , Caveolina 1/deficiencia , Caveolina 1/genética , ADN Complementario/genética , Dependovirus/genética , Quinasa 1 de Adhesión Focal/fisiología , Técnicas de Silenciamiento del Gen , Genes Reporteros , Genes Sintéticos , Glaucoma/metabolismo , Glaucoma/patología , Integrina beta1/fisiología , Presión Intraocular , Inyecciones Intravítreas , Glicoproteínas de Membrana/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Regiones Promotoras Genéticas , Proteína Tirosina Fosfatasa no Receptora Tipo 11/biosíntesis , Proteína Tirosina Fosfatasa no Receptora Tipo 11/genética , Proteínas Tirosina Quinasas/fisiología , Regulación hacia Arriba
6.
Mol Brain ; 14(1): 53, 2021 03 16.
Artículo en Inglés | MEDLINE | ID: mdl-33726791

RESUMEN

In addition to providing structural support, caveolin-1 (Cav1), a component of lipid rafts, including caveolae, in the plasma membrane, is involved in various cellular mechanisms, including signal transduction. Although pre-synaptic membrane dynamics and trafficking are essential cellular processes during synaptic vesicle exocytosis/synaptic transmission and synaptic vesicle endocytosis/synaptic retrieval, little is known about the involvement of Cav1 in synaptic vesicle dynamics. Here we demonstrate that synaptic vesicle exocytosis is significantly impaired in Cav1-knockdown (Cav1-KD) neurons. Specifically, the size of the synaptic recycled vesicle pool is modestly decreased in Cav1-KD synapses and the kinetics of synaptic vesicle endocytosis are somewhat slowed. Notably, neurons rescued by triple mutants of Cav1 lacking palmitoylation sites mutants show impairments in both synaptic transmission and retrieval. Collectively, our findings implicate Cav1 in activity-driven synaptic vesicle dynamics-both exocytosis and endocytosis-and demonstrate that palmitoylation of Cav1 is important for this activity.


Asunto(s)
Caveolina 1/deficiencia , Hipocampo/citología , Proteínas del Tejido Nervioso/deficiencia , Neuronas/fisiología , Transmisión Sináptica/fisiología , Animales , Caveolina 1/antagonistas & inhibidores , Caveolina 1/genética , Caveolina 1/fisiología , Células Cultivadas , Exocitosis/fisiología , Microdominios de Membrana , Mutación , Proteínas del Tejido Nervioso/antagonistas & inhibidores , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/fisiología , Ácido Palmítico/metabolismo , Terminales Presinápticos/química , Terminales Presinápticos/fisiología , Procesamiento Proteico-Postraduccional , Interferencia de ARN , ARN Interferente Pequeño/genética , Ratas , Ratas Sprague-Dawley
7.
FEBS Lett ; 595(4): 532-547, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33314143

RESUMEN

Integrin-mediated adhesion regulates cellular responses to changes in the mechanical and biochemical properties of the extracellular matrix. Cell-matrix adhesion regulates caveolar endocytosis, dependent on caveolin 1 (Cav1) Tyr14 phosphorylation (pY14Cav1), to control anchorage-dependent signaling. We find that cell-matrix adhesion regulates pY14Cav1 levels in mouse fibroblasts. Biochemical fractionation reveals endogenous pY14Cav1 to be present in caveolae and focal adhesions (FA). Adhesion does not affect caveolar pY14Cav1, supporting its regulation at FA, in which PF-228-mediated inhibition of focal adhesion kinase (FAK) disrupts. Cell adhesion on 2D polyacrylamide matrices of increasing stiffness stimulates Cav1 phosphorylation, which is comparable to the phosphorylation of FAK. Inhibition of FAK across varying stiffnesses shows it regulates pY14Cav1 more prominently at higher stiffness. Taken together, these studies reveal the presence of FAK-pY14Cav1 crosstalk at FA, which is regulated by cell-matrix adhesion.


Asunto(s)
Caveolina 1/genética , Matriz Extracelular/metabolismo , Fibroblastos/metabolismo , Quinasa 1 de Adhesión Focal/genética , Procesamiento Proteico-Postraduccional , Tirosina/metabolismo , Animales , Caveolas/efectos de los fármacos , Caveolas/metabolismo , Caveolas/ultraestructura , Caveolina 1/deficiencia , Adhesión Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Embrión de Mamíferos , Endocitosis/efectos de los fármacos , Matriz Extracelular/efectos de los fármacos , Matriz Extracelular/ultraestructura , Fibroblastos/efectos de los fármacos , Fibroblastos/ultraestructura , Quinasa 1 de Adhesión Focal/metabolismo , Adhesiones Focales/efectos de los fármacos , Adhesiones Focales/metabolismo , Adhesiones Focales/ultraestructura , Mecanotransducción Celular , Ratones , Ratones Noqueados , Fosforilación/efectos de los fármacos , Cultivo Primario de Células , Inhibidores de Proteínas Quinasas/farmacología , Quinolonas/farmacología , Sulfonas/farmacología
8.
Invest Ophthalmol Vis Sci ; 61(12): 19, 2020 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-33079993

RESUMEN

Purpose: The immune-privileged environment and complex organization of retinal tissue support the retina's essential role in visual function, yet confound inquiries into cell-specific inflammatory effects that lead to dysfunction and degeneration. Caveolin-1 (Cav1) is an integral membrane protein expressed in several retinal cell types and is implicated in immune regulation. However, whether Cav1 promotes or inhibits inflammatory processes in the retina (as well as in other tissues) remains unclear. Previously, we showed that global-Cav1 depletion resulted in reduced retinal inflammatory cytokine production but paradoxically elevated retinal immune cell infiltration. We hypothesized that these disparate responses are the result of differential cell-specific Cav1 functions in the retina. Methods: We used Cre/lox technology to deplete Cav1 specifically in the neural retinal (NR) compartment to clarify the role NR-specific Cav1 (NR-Cav1) in the retinal immune response to intravitreal inflammatory challenge induced by activation of Toll-like receptor-4 (TLR4). We used multiplex protein suspension array and flow cytometry to evaluate innate immune activation. Additionally, we used bioinformatics assessment of differentially expressed membrane-associated proteins to infer relationships between NR-Cav1 and immune response pathways. Results: NR-Cav1 depletion, which primarily affects Müller glia Cav1 expression, significantly altered immune response pathway regulators, decreased retinal inflammatory cytokine production, and reduced retinal immune cell infiltration in response to LPS-stimulated inflammatory induction. Conclusions: Cav1 expression in the NR compartment promotes the innate TLR4-mediated retinal tissue immune response. Additionally, we have identified novel potential immune modulators differentially expressed with NR-Cav1 depletion. This study further clarifies the role of NR-Cav1 in retinal inflammation.


Asunto(s)
Caveolina 1/fisiología , Inflamación/inducido químicamente , Lipopolisacáridos/toxicidad , Retina/metabolismo , Retinitis/inducido químicamente , Animales , Western Blotting , Caveolina 1/deficiencia , Citocinas/metabolismo , Sinergismo Farmacológico , Electrorretinografía , Citometría de Flujo , Inmunohistoquímica , Inflamación/metabolismo , Inflamación/patología , Inyecciones Intravítreas , Espectrometría de Masas , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía Confocal , Nistagmo Optoquinético/fisiología , Proteómica , Retinitis/metabolismo , Retinitis/patología , Salmonella typhimurium , Receptor Toll-Like 4/metabolismo
9.
Am J Physiol Cell Physiol ; 319(5): C933-C944, 2020 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-32936699

RESUMEN

Caveolin-1 (Cav-1) is a scaffolding protein and a major component of caveolae/lipid rafts. Previous reports have shown that endothelial dysfunction in Cav-1-deficient (Cav-1-/-) mice is mediated by elevated oxidative stress through endothelial nitric oxide synthase (eNOS) uncoupling and increased NADPH oxidase. Oxidant stress is the net balance of oxidant generation and scavenging, and the role of Cav-1 as a regulator of antioxidant enzymes in vascular tissue is poorly understood. Extracellular SOD (SOD3) is a copper (Cu)-containing enzyme that is secreted from vascular smooth muscle cells/fibroblasts and subsequently binds to the endothelial cells surface, where it scavenges extracellular [Formula: see text] and preserves endothelial function. SOD3 activity is dependent on Cu, supplied by the Cu transporter ATP7A, but whether Cav-1 regulates the ATP7A-SOD3 axis and its role in oxidative stress-mediated vascular dysfunction has not been studied. Here we show that the activity of SOD3, but not SOD1, was significantly decreased in Cav-1-/- vessels, which was rescued by re-expression of Cav-1 or Cu supplementation. Loss of Cav-1 reduced ATP7A protein, but not mRNA, and this was mediated by ubiquitination of ATP7A and proteasomal degradation. ATP7A bound to Cav-1 and was colocalized with SOD3 in caveolae/lipid rafts or perinucleus in vascular tissues or cells. Impaired endothelium-dependent vasorelaxation in Cav-1-/- mice was rescued by gene transfer of SOD3 or by ATP7A-overexpressing transgenic mice. These data reveal an unexpected role of Cav-1 in stabilizing ATP7A protein expression by preventing its ubiquitination and proteasomal degradation, thereby increasing SOD3 activity, which in turn protects against vascular oxidative stress-mediated endothelial dysfunction.


Asunto(s)
Caveolina 1/genética , ATPasas Transportadoras de Cobre/genética , Células Endoteliales/metabolismo , Superóxido Dismutasa-1/genética , Superóxido Dismutasa/genética , Animales , Aorta/citología , Aorta/metabolismo , Caveolina 1/deficiencia , Cobre/farmacología , Proteínas Transportadoras de Cobre/genética , Proteínas Transportadoras de Cobre/metabolismo , ATPasas Transportadoras de Cobre/metabolismo , Células Endoteliales/citología , Células Endoteliales/efectos de los fármacos , Fibroblastos/citología , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Regulación de la Expresión Génica , Masculino , Arterias Mesentéricas/citología , Arterias Mesentéricas/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Chaperonas Moleculares/genética , Chaperonas Moleculares/metabolismo , Estrés Oxidativo , Cultivo Primario de Células , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteolisis , Transducción de Señal , Superóxido Dismutasa/metabolismo , Superóxido Dismutasa-1/metabolismo , Ubiquitinación/efectos de los fármacos , Vasodilatación/efectos de los fármacos
10.
Int J Mol Sci ; 21(15)2020 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-32718046

RESUMEN

Elevated free fatty acids (FFAs) impair beta cell function and reduce beta cell mass as a consequence of the lipotoxicity that occurs in type 2 diabetes (T2D). We previously reported that the membrane protein caveolin-1 (CAV1) sensitizes to palmitate-induced apoptosis in the beta pancreatic cell line MIN6. Thus, our hypothesis was that CAV1 knock-out (CAV1 KO) mice subjected to a high fat diet (HFD) should suffer less damage to beta cells than wild type (WT) mice. Here, we evaluated the in vivo response of beta cells in the pancreatic islets of 8-week-old C57Bl/6J CAV1 KO mice subjected to a control diet (CD, 14% kcal fat) or a HFD (60% kcal fat) for 12 weeks. We observed that CAV1 KO mice were resistant to weight gain when on HFD, although they had high serum cholesterol and FFA levels, impaired glucose tolerance and were insulin resistant. Some of these alterations were also observed in mice on CD. Interestingly, KO mice fed with HFD showed an adaptive response of the pancreatic beta cells and exhibited a significant decrease in beta cell apoptosis in their islets compared to WT mice. These in vivo results suggest that although the CAV1 KO mice are metabolically unhealthy, they adapt better to a HFD than WT mice. To shed light on the possible signaling pathway(s) involved, MIN6 murine beta cells expressing (MIN6 CAV) or not expressing (MIN6 Mock) CAV1 were incubated with the saturated fatty acid palmitate in the presence of mitogen-activated protein kinase inhibitors. Western blot analysis revealed that CAV1 enhanced palmitate-induced JNK, p38 and ERK phosphorylation in MIN6 CAV1 cells. Moreover, all the MAPK inhibitors partially restored MIN6 viability, but the effect was most notable with the ERK inhibitor. In conclusion, our results suggest that CAV1 KO mice adapted better to a HFD despite their altered metabolic state and that this may at least in part be due to reduced beta cell damage. Moreover, they indicate that the ability of CAV1 to increase sensitivity to FFAs may be mediated by MAPK and particularly ERK activation.


Asunto(s)
Caveolina 1/deficiencia , Dieta Alta en Grasa/efectos adversos , Resistencia a la Insulina , Células Secretoras de Insulina/metabolismo , Sistema de Señalización de MAP Quinasas , Animales , Caveolina 1/metabolismo , Muerte Celular/efectos de los fármacos , Muerte Celular/genética , Células Secretoras de Insulina/patología , Masculino , Ratones , Ratones Noqueados
11.
Biochem Pharmacol ; 180: 114153, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32679126

RESUMEN

Caveolin-1 (Cav-1) is a multifunctional protein and critical for the production of nitric oxide (NO) in intestinal physiological and pathological conditions, but its role in the inflammatory bowel disease(IBD) are still controversial. In this study we tested the hypothesis that Cav-1 could be an important cellular defense against IBD through inhibiting nitrosative stress and mucosal barrier damage. Male wild-type mice and Cav-1 knockout (Cav-1-/-) mice were subjected to 3% dextran sodium sulfate(DSS) for 7d to establish the experimental colitis model. A representative iNOS inhibitor (1400 W) was adopted to suppress the activity of iNOS in parallel group. Body weight and disease activity index were monitored. The colon tissues were evaluated through histological analysis. We found Cav-1 was down-regulated in the colon tissue and accompanied with the increase of iNOS and NO levels after DSS administration. Cav-1-/- mice were greatly increased susceptibility to DSS-induced colitis with the more weight loss and higher disease score than WT mice. Ablation of Cav-1gene significantly resulted in RNS overproduction, tight junctions impaired and inflammation elevated, which aggravated the severity of the intestinal damages. Furthermore, pharmacologic inhibition of iNOS by 1400 W significantly attenuated DSS-induced colitis in both WT and Cav-1-/- groups. Our results revealed an important role of Cav-1 in preventing intestinal nitrosative stress and mucosal barrier damage in the development of DSS-induced colitis.


Asunto(s)
Caveolina 1/deficiencia , Colitis/inducido químicamente , Colitis/metabolismo , Sulfato de Dextran/toxicidad , Mucosa Intestinal/metabolismo , Estrés Nitrosativo/fisiología , Animales , Colitis/prevención & control , Mucosa Intestinal/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Estrés Nitrosativo/efectos de los fármacos
12.
J Endocrinol ; 247(1): 25-38, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32668406

RESUMEN

Here we have investigated the role of the protein caveolin 1 (Cav1) and caveolae in the secretion of the white adipocyte hormone adiponectin. Using mouse primary subcutaneous adipocytes genetically depleted of Cav1, we show that the adiponectin secretion, stimulated either adrenergically or by insulin, is abrogated while basal (unstimulated) release of adiponectin is elevated. Adiponectin secretion is similarly affected in wildtype mouse and human adipocytes where the caveolae structure was chemically disrupted. The altered ex vivo secretion in adipocytes isolated from Cav1 null mice is accompanied by lowered serum levels of the high-molecular weight (HMW) form of adiponectin, whereas the total concentration of adiponectin is unaltered. Interestingly, levels of HMW adiponectin are maintained in adipose tissue from Cav1-depleted mice, signifying that a secretory defect is present. The gene expression of key regulatory proteins known to be involved in cAMP/adrenergically triggered adiponectin exocytosis (the beta-3-adrenergic receptor and exchange protein directly activated by cAMP) remains intact in Cav1 null adipocytes. Microscopy and fractionation studies indicate that adiponectin vesicles do not co-localise with Cav1 but that some vesicles are associated with a specific fraction of caveolae. Our studies propose that Cav1 has an important role in secretion of HMW adiponectin, even though adiponectin-containing vesicles are not obviously associated with this protein. We suggest that Cav1, and/or the caveolae domain, is essential for the organisation of signalling pathways involved in the regulation of HMW adiponectin exocytosis, a function that is disrupted in Cav1/caveolae-depleted adipocytes.


Asunto(s)
Adipocitos Blancos/metabolismo , Adiponectina/metabolismo , Caveolina 1/fisiología , Adiponectina/sangre , Adiponectina/genética , Adulto , Anciano , Animales , Caveolina 1/deficiencia , Membrana Celular/química , Dieta , Exocitosis/fisiología , Femenino , Expresión Génica/fisiología , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Persona de Mediana Edad , Obesidad/etiología , Obesidad/metabolismo
13.
Psychopharmacology (Berl) ; 237(9): 2673-2684, 2020 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-32488350

RESUMEN

RATIONALE: Caveolin-1 (CAV1) is a structural protein critical for spatial organization of neuronal signaling molecules. Whether CAV1 is required for long-lasting neuronal plasticity remains unknown. OBJECTIVE AND METHODS: We sought to examine the effects of CAV1 knockout (KO) on functional plasticity and hypothesized that CAV1 deficiency would impact drug-induced long-term plasticity in the nucleus accumbens (NAc). We first examined cell morphology of NAc medium spiny neurons in a striatal/cortical co-culture system before moving in vivo to study effects of CAV1 KO on cocaine-induced plasticity. Whole-cell patch-clamp recordings were performed to determine effects of chronic cocaine (15 mg/kg) on medium spiny neuron excitability. To test for deficits in behavioral plasticity, we examined the effect of CAV1 KO on locomotor sensitization. RESULTS: Disruption of CAV1 expression leads to baseline differences in medium spiny neuron (MSN) structural morphology, such that MSNs derived from CAV1 KO animals have increased dendritic arborization when cultured with cortical neurons. The effect was dependent on phospholipase C and cell-type intrinsic loss of CAV1. Slice recordings of nucleus accumbens shell MSNs revealed that CAV1 deficiency produces a loss of neuronal plasticity. Specifically, cocaine-induced firing rate depression was absent in CAV1 KO animals, whereas baseline electrophysiological properties were similar. This was reflected by a loss of cocaine-mediated behavioral sensitization in CAV1 KO animals, with unaffected baseline locomotor responsiveness. CONCLUSIONS: This study highlights a critical role for nucleus accumbens CAV1 in plasticity related to the administration of drugs of abuse.


Asunto(s)
Caveolina 1/deficiencia , Plasticidad Neuronal/fisiología , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Núcleo Accumbens/efectos de los fármacos , Núcleo Accumbens/metabolismo , Animales , Caveolina 1/genética , Cocaína/farmacología , Técnicas de Cocultivo , Inhibidores de Captación de Dopamina/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neuritas/metabolismo , Plasticidad Neuronal/efectos de los fármacos , Técnicas de Placa-Clamp
14.
Mol Neurobiol ; 57(9): 3759-3784, 2020 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-32578008

RESUMEN

Retinal ganglion cell degeneration is a characteristic feature of glaucoma, and accordingly, protection of these cells constitutes a major therapeutic objective in the disease. Here, we demonstrate the key influence of caveolin (Cav) in regulating the inner retinal homeostasis in two models of experimentally elevated intraocular pressure (IOP). Two groups of Cav-1-/- and wild-type mice were used in the study. Animals were subjected to experimentally induced chronic and acutely elevated IOP and any changes in their retinal function were assessed by positive scotopic threshold response recordings. TUNEL and cleaved caspase-3 assays were performed to evaluate apoptotic changes in the retina while Brn3a immunostaining was used as a marker to assess and quantify ganglion cell layer (GCL) changes. H&E staining was carried out on retinal sections to evaluate histological differences in retinal laminar structure. Cav-1 ablation partially protected the inner retinal function in both chronic and acute models of elevated IOP. The protective effects of Cav-1 loss were also evident histologically by reduced loss of GCL density in both models. The phenotypic protection in Cav-1-/- glaucoma mice paralleled with increased TrkB phosphorylation and reduced endoplasmic reticulum stress markers and apoptotic activation in the inner retinas. This study corroborated previous findings of enhanced Shp2 phosphorylation in a chronic glaucoma model and established a novel role of Cav-1 in mediating activation of this phosphatase in the inner retina in vivo. Collectively, these findings highlight the critical involvement of Cav-1 regulatory mechanisms in ganglion cells in response to increased IOP, implicating Cav-1 as a potential therapeutic target in glaucoma.


Asunto(s)
Apoptosis , Caveolina 1/metabolismo , Glaucoma/patología , Neuroprotección , Retina/lesiones , Retina/patología , Animales , Caveolina 1/deficiencia , Enfermedad Crónica , Modelos Animales de Enfermedad , Estrés del Retículo Endoplásmico , Glaucoma/fisiopatología , Presión Intraocular , Ratones Endogámicos C57BL , Ratones Noqueados , Fosforilación , Proteína Tirosina Fosfatasa no Receptora Tipo 11/metabolismo , Proteómica , Receptor trkB/metabolismo , Retina/fisiopatología , Transducción de Señal
15.
Oxid Med Cell Longev ; 2020: 1868764, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32377291

RESUMEN

A more comprehensive understanding of the complexity of pancreatic cancer pathobiology, especially, and understanding of the role of the tumor microenvironment (TME) in disease progression should pave the way for therapies to improve patient response rates. Previous studies reported that caveolin-1 (Cav-1) has both tumor-promoting and tumor-suppressive functions. However, the function of Cav-1 in the pancreatic cancer microenvironment remains largely unexplored. Here, we show that coinjection of Cav-1-silenced pancreatic stellate cells (PSCs) with pancreatic cancer cells increased tumor growth. To comprehensively characterize paracrine communication between pancreatic cancer cells and PSCs, PSCs were cultured with pancreatic cancer cell conditioned medium (CM) containing cytokines. We reveal that Cav-1-silenced PSCs facilitated the growth of pancreatic cancer cells via enhanced paracrine shh/MMP2/bFGF/IL-6 signaling. Specifically, Cav-1-silenced PSCs exhibited increased shh expression, which heterotypically activated the shh signaling pathway in pancreatic cancer cells. Moreover, Cav-1-deficient PSCs accumulated ROS to enhance the shh pathway and angiogenesis in pancreatic cancer cells. In addition, overexpression of Nrf2 reversed the effects of Cav-1 knockdown on PSCs, increasing ROS production and enhancing paracrine shh/MMP2/bFGF/IL-6 signaling. Together, our findings show that stromal Cav-1 may mediate different mechanisms in the complex interaction between cancer cells and their microenvironment though Nrf2-induced shh signaling activation during pancreatic cancer progression.


Asunto(s)
Caveolina 1/metabolismo , Proteínas Hedgehog/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , Neoplasias Pancreáticas/metabolismo , Células Estrelladas Pancreáticas/metabolismo , Animales , Caveolina 1/deficiencia , Línea Celular Tumoral , Xenoinjertos , Células Endoteliales de la Vena Umbilical Humana , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Neovascularización Patológica/metabolismo , Neovascularización Patológica/patología , Neoplasias Pancreáticas/irrigación sanguínea , Neoplasias Pancreáticas/patología , Células Estrelladas Pancreáticas/patología
16.
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
17.
Cell Death Dis ; 11(4): 228, 2020 04 09.
Artículo en Inglés | MEDLINE | ID: mdl-32273493

RESUMEN

The integral membrane protein caveolin-1 (CAV1) plays a central role in radioresistance-mediating tumor-stroma interactions of advanced prostate cancer (PCa). Among the tumor-stroma, endothelial cells (EC) evolved as critical determinants of the radiation response. CAV1 deficiency in angiogenic EC was already shown to account for increased apoptosis rates of irradiated EC. This study explores the potential impact of differential CAV1 levels in EC on the acid sphingomyelinase (ASMase)/ceramide pathway as a key player in the regulation of EC apoptosis upon irradiation and cancer cell radioresistance. Enhanced apoptosis sensitivity of CAV1-deficient EC was associated with increased ASMase activity, ceramide generation, formation of large lipid platforms, and finally an altered p38 mitogen-activated protein kinase (MAPK)/heat-shock protein 27 (HSP27)/AKT (protein kinase B, PKB) signaling. CAV1-deficient EC increased the growth delay of LNCaP and PC3 PCa cells upon radiation treatment in direct 3D spheroid co-cultures. Exogenous C6 and C16 ceramide treatment in parallel increased the growth delay of PCa spheroids and induced PCa cell apoptosis. Analysis of the respective ceramide species in PCa cells with increased CAV1 levels like those typically found in radio-resistant advanced prostate tumors further revealed an upregulation of unsaturated C24:1 ceramide that might scavenge the effects of EC-derived apoptosis-inducing C16 ceramide. Higher ASMase as well as ceramide levels could be confirmed by immunohistochemistry in human advanced prostate cancer specimen bearing characteristic CAV1 tumor-stroma alterations. Conclusively, CAV1 critically regulates the generation of ceramide-dependent (re-)organization of the plasma membrane that in turn affects the radiation response of EC and adjacent PCa cells. Understanding the CAV1-dependent crosstalk between tumor cells and the host-derived tumor microvasculature and its impact on radiosensitivity may allow to define a rational strategy for overcoming tumor radiation resistance improving clinical outcomes by targeting CAV1.


Asunto(s)
Caveolina 1/metabolismo , Ceramidas/metabolismo , Células Endoteliales/efectos de la radiación , Neoplasias de la Próstata/patología , Neoplasias de la Próstata/radioterapia , Esfingomielina Fosfodiesterasa/metabolismo , Células del Estroma/patología , Caveolina 1/biosíntesis , Caveolina 1/deficiencia , Comunicación Celular/fisiología , Comunicación Celular/efectos de la radiación , Línea Celular Tumoral , Ceramidas/biosíntesis , Ceramidas/farmacología , Células Endoteliales/metabolismo , Células Endoteliales/patología , Humanos , Sistema de Señalización de MAP Quinasas/efectos de la radiación , Masculino , Células PC-3 , Neoplasias de la Próstata/metabolismo , Tolerancia a Radiación , Transducción de Señal , Células del Estroma/metabolismo , Microambiente Tumoral
18.
Cell Death Dis ; 11(2): 104, 2020 02 06.
Artículo en Inglés | MEDLINE | ID: mdl-32029710

RESUMEN

Caveolin-1 (CAV1) is a crucial regulator of lipid accumulation and metabolism. Previous studies have shown that global Cav1 deficiency affects lipid metabolism and hepatic steatosis. We aimed to analyze the consequences of hepatocyte-specific Cav1 knockout under healthy conditions and upon non-alcoholic fatty liver disease (NAFLD) development. Male and female hepatocyte-specific Cav1 knockout (HepCAV1ko) mice were fed a methionine/choline (MCD) deficient diet for 4 weeks. MCD feeding caused severe hepatic steatosis and slight fibrosis. In addition, liver function parameters, i.e., ALT, AST, and GLDH, were elevated, while cholesterol and glucose level were reduced upon MCD feeding. These differences were not affected by hepatocyte-specific Cav1 knockout. Microarray analysis showed strong differences in gene expression profiles of livers from HepCAV1ko mice compared those of global Cav1 knockout animals. Pathway enrichment analysis identified that metabolic alterations were sex-dimorphically regulated by hepatocyte-specific CAV1. In male HepCAV1ko mice, metabolic pathways were suppressed in NAFLD, whereas in female knockout mice induced. Moreover, gender-specific transcription profiles were modulated in healthy animals. In conclusion, our results demonstrate that hepatocyte-specific Cav1 knockout significantly altered gene profiles, did not affect liver steatosis and fibrosis in NAFLD and that gender had severe impact on gene expression patterns in healthy and diseased hepatocyte-specific Cav1 knockout mice.


Asunto(s)
Caveolina 1/metabolismo , Metabolismo Energético , Hepatocitos/metabolismo , Hígado/metabolismo , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Animales , Biomarcadores/sangre , Caveolina 1/deficiencia , Caveolina 1/genética , Células Cultivadas , Bases de Datos Genéticas , Modelos Animales de Enfermedad , Metabolismo Energético/genética , Femenino , Hepatocitos/patología , Hígado/patología , Cirrosis Hepática/genética , Cirrosis Hepática/metabolismo , Cirrosis Hepática/patología , Masculino , Ratones Noqueados , Enfermedad del Hígado Graso no Alcohólico/genética , Enfermedad del Hígado Graso no Alcohólico/patología , Caracteres Sexuales , Transcriptoma
19.
Biomolecules ; 9(10)2019 10 18.
Artículo en Inglés | MEDLINE | ID: mdl-31635212

RESUMEN

Caveolae-associated protein caveolin-1 (Cav-1) plays key roles in cellular processes such as mechanosensing, receptor coupling to signaling pathways, cell growth, apoptosis, and cancer. In 1321N1 astrocytoma cells Cav-1 interacts with the P2Y2 receptor (P2Y2R) to modulate its downstream signaling. P2Y2R and its signaling machinery also mediate pro-survival actions after mechanical injury. This study determines if Cav-1 knockdown (KD) affects P2Y2R signaling and its pro-survival actions in the 1321N1 astrocytoma cells mechanical injury model system. KD of Cav-1 decreased its expression in 1321N1 cells devoid of or expressing hHAP2Y2R by ~88% and ~85%, respectively. Cav-1 KD had no significant impact on P2Y2R expression. Post-injury densitometric analysis of pERK1/2 and Akt activities in Cav-1-positive 1321N1 cells (devoid of or expressing a hHAP2Y2R) revealed a P2Y2R-dependent temporal increase in both kinases. These temporal increases in pERK1/2 and pAkt were significantly decreased in Cav-1 KD 1321N1 (devoid of or expressing a hHAP2Y2R). Cav-1 KD led to an ~2.0-fold and ~2.4-fold decrease in the magnitude of the hHAP2Y2R-mediated pERK1/2 and pAkt kinases' activity, respectively. These early-onset hHAP2Y2R-mediated signaling responses in Cav-1-expressing and Cav-1 KD 1321N1 correlated with changes in cell viability (via a resazurin-based method) and apoptosis (via caspase-9 expression). In Cav-1-positive 1321N1 cells, expression of hHAP2Y2R led to a significant increase in cell viability and decreased apoptotic (caspase-9) activity after mechanical injury. In contrast, hHAP2Y2R-elicited changes in viability and apoptotic (caspase-9) activity were decreased after mechanical injury in Cav-1 KD 1321N1 cells expressing hHAP2Y2R. These findings support the importance of Cav-1 in modulating P2Y2R signaling during mechanical injury and its protective actions in a human astrocytoma cell line, whilst shedding light on potential new venues for brain injury or trauma interventions.


Asunto(s)
Astrocitoma/metabolismo , Caveolina 1/metabolismo , Receptores Purinérgicos P2Y2/metabolismo , Transducción de Señal , Estrés Mecánico , Astrocitoma/patología , Caspasa 9/metabolismo , Caveolina 1/deficiencia , Caveolina 1/aislamiento & purificación , Supervivencia Celular , Humanos , Células Tumorales Cultivadas
20.
Invest Ophthalmol Vis Sci ; 60(12): 4002-4007, 2019 09 03.
Artículo en Inglés | MEDLINE | ID: mdl-31560766

RESUMEN

Purpose: This study aims to investigate the pharmacologic consequence of genetic deletion of nitric oxide synthase 3 (NOS3) in caveolin 1 (Cav1)-/- mice (double knockout [DKO]) in response to a nitric oxide (NO) donor and two NOS inhibitors. Methods: NO donor sodium nitroprusside (SNP; 10-40 mg/mL), NOS inhibitor L-NG-nitroarginine methyl ester (L-NAME; 10-200 µM), and cavtratin (10-75 µM ) was administered topically to the eye while the contralateral eyes were vehicle controls. Intraocular pressure (IOP) was measured in both eyes by tonometry. Cyclic guanosine monophosphate (cGMP) level in outflow tissue was measured by ELISA assay. Protein expression were analyzed by western blot. Results: Inducible NOS (iNOS) expression significantly increased in the DKO mice compared with the wild type (WT), Cav1 knockout (Cav1 KO), and NOS3 KO mice. In contrast to WT, Cav1 KO and NOS3 KO mice, SNP concentration of up to 30 mg/mL did not significantly affect IOP in DKO mice. However, higher concentration (40 mg/mL) SNP significantly reduced IOP by 14% (n = 8, P < 0.01). Similarly, only 200 µM L-NAME produced a significant increase in IOP (n = 10, P < 0.05). Cavtratin did not significantly change IOP in DKO and NOS3 KO mice. cGMP activity in DKO mice was significantly lower than Cav1 KO mice (n = 4, P < 0.05). Conclusions: In conclusion, our results demonstrated that genetic deletion of NOS3 in Cav1 deficient mice resulted in reduced sensitivity to the NO donor SNP and the two NOS inhibitors possibly due to compromised NOS and cGMP activity.


Asunto(s)
Caveolina 1/deficiencia , Inhibidores Enzimáticos/farmacología , Eliminación de Gen , Presión Intraocular/efectos de los fármacos , Donantes de Óxido Nítrico/farmacología , Óxido Nítrico Sintasa de Tipo III/genética , Administración Oftálmica , Animales , Western Blotting , Caveolina 1/metabolismo , Caveolina 1/farmacología , GMP Cíclico/metabolismo , Ensayo de Inmunoadsorción Enzimática , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , NG-Nitroarginina Metil Éster/farmacología , Óxido Nítrico Sintasa de Tipo III/metabolismo , Nitroprusiato/farmacología , Fragmentos de Péptidos/farmacología , Tonometría Ocular
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...