Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 302
Filtrar
1.
J Zhejiang Univ Sci B ; 22(10): 818-838, 2021 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-34636186

RESUMO

OBJECTIVES: Cardiac hypertrophy and fibrosis are major pathological manifestations observed in left ventricular remodeling induced by angiotensin II (AngII). Low-intensity pulsed ultrasound (LIPUS) has been reported to ameliorate cardiac dysfunction and myocardial fibrosis in myocardial infarction (MI) through mechano-transduction and its downstream pathways. In this study, we aimed to investigate whether LIPUS could exert a protective effect by ameliorating AngII-induced cardiac hypertrophy and fibrosis and if so, to further elucidate the underlying molecular mechanisms. METHODS: We used AngII to mimic animal and cell culture models of cardiac hypertrophy and fibrosis. LIPUS irradiation was applied in vivo for 20 min every 2 d from one week before mini-pump implantation to four weeks after mini-pump implantation, and in vitro for 20 min on each of two occasions 6 h apart. Cardiac hypertrophy and fibrosis levels were then evaluated by echocardiographic, histopathological, and molecular biological methods. RESULTS: Our results showed that LIPUS could ameliorate left ventricular remodeling in vivo and cardiac fibrosis in vitro by reducing AngII-induced release of inflammatory cytokines, but the protective effects on cardiac hypertrophy were limited in vitro. Given that LIPUS increased the expression of caveolin-1 in response to mechanical stimulation, we inhibited caveolin-1 activity with pyrazolopyrimidine 2 (pp2) in vivo and in vitro. LIPUS-induced downregulation of inflammation was reversed and the anti-fibrotic effects of LIPUS were absent. CONCLUSIONS: These results indicated that LIPUS could ameliorate AngII-induced cardiac fibrosis by alleviating inflammation via a caveolin-1-dependent pathway, providing new insights for the development of novel therapeutic apparatus in clinical practice.


Assuntos
Cardiomegalia/terapia , Caveolina 1/fisiologia , Inflamação/prevenção & controle , Miocárdio/patologia , Ondas Ultrassônicas , Angiotensina II/farmacologia , Animais , Cardiomegalia/induzido quimicamente , Células Cultivadas , Fibrose , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/efeitos dos fármacos
2.
Life Sci ; 285: 119939, 2021 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-34506836

RESUMO

AIMS: Nitric oxide synthases (NOSs) are key enzymes regulating vascular function. Previously, we reported that ß-adrenergic (ß-AR) overstimulation, a common feature of cardiovascular diseases, did not impair endothelium-dependent vasodilation, although it resulted in endothelial NOS (eNOS) uncoupling and reduced NO bioavailability. In addition to NO, neuronal NOS (nNOS) produces H2O2, which contributes to vasodilation. However, there is limited information regarding vascular ß-AR signaling and nNOS. In the present study, we assessed the possible role of nNOS-derived H2O2 and caveolins on endothelial vasodilation function following ß-AR overstimulation. MAIN METHODS: Male C57BL/6 wild-type and nNOS knockout mice (nNOS-/-) were treated with the ß-AR agonist isoproterenol (ISO, 15 mg·kg-1·day-1, s.c.) or vehicle (VHE) for seven days. Relaxation responses of aortic rings were evaluated using wire myograph and H2O2 by Amplex Red. KEY FINDINGS: Acetylcholine- or calcium ionophore A23187-induced endothelium-dependent relaxation was similar in aortic rings from VHE and ISO. However, this relaxation was significantly reduced in aortas from ISO compared to VHE when (1) caveolae were disrupted, (2) nNOS was pharmacologically inhibited or genetically suppressed and (3) H2O2 was scavenged. NOS-derived H2O2 production was higher in the aortas of ISO mice than in those of VHE mice. Aortas from ISO-treated mice showed increased expression of caveolin-1, nNOS and catalase, while caveolin-3 expression did not change. SIGNIFICANCE: The results suggest a role of caveolin-1 and the nNOS/H2O2 vasodilatory pathway in endothelium-dependent relaxation following ß-AR overstimulation and reinforce the protective role of nNOS in cardiovascular diseases associated with high adrenergic tone.


Assuntos
Caveolina 1/fisiologia , Óxido Nítrico Sintase Tipo I/fisiologia , Receptores Adrenérgicos alfa/metabolismo , Vasodilatação/fisiologia , Agonistas Adrenérgicos beta/farmacologia , Animais , Calcimicina/farmacologia , Ionóforos de Cálcio/farmacologia , Doenças Cardiovasculares/genética , Doenças Cardiovasculares/fisiopatologia , Caveolina 1/genética , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/fisiologia , Peróxido de Hidrogênio/metabolismo , Isoproterenol/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Óxido Nítrico Sintase Tipo I/genética , Vasodilatação/efeitos dos fármacos , Vasodilatação/genética
3.
Int J Biochem Cell Biol ; 137: 106027, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34157397

RESUMO

It has been reported that caveolin-1 (Cav-1) acts as a tumor promoter in hepatocellular carcinoma (HCC). Our previous studies showed that Cav-1 promoted mouse hepatocarcinoma cell adhesion to fibronectin by upregulating ß-galactoside α2,6-sialyltransferase I (ST6Gal-I) expression. However, the detailed mechanism by which Cav-1 regulates ST6Gal-I is not fully understood. In this study, we found that the expression levels of Cav-1 and ST6Gal-I were increased in HCC tissues and correlated with poor prognosis. Cav-1 upregulated ST6Gal-I expression to promote the migration and invasion of HCC cells by inducing epithelial-to-mesenchymal transition. Importantly, the binding of the transcription factor nuclear receptor 4A2/retinoid X receptor alpha (NR4A2/RXRα) to the -550/-200 region of the ST6GAL1 promoter was critical for Cav-1-induced ST6GAL1 gene expression. Furthermore, Cav-1 expression activated the phosphatidylinositol 3-kinase/protein kinase B/mammalian target of rapamycin (PI3K/AKT/mTOR) signaling pathway, followed by upregulation of NR4A2 expression and phosphorylation of RXRα, which facilitated the complex of NR4A2 and phosphorylated RXRα forming and binding to the ST6GAL1 promoter region to induce its transcription. Finally, in the diethylnitrosamine (DEN)-induced HCC murine model, the expression levels of NR4A2, p-RXRα, ST6Gal-I, and α2,6-linked sialic acid decreased in parallel in Cav-1-/- mice compared with Cav-1+/+ mice, which was consistent with the above in vitro results. These findings provide insight into the mechanism of ST6GAL1 gene transcription mediated by Cav-1, which may lead to the development of novel therapeutic strategies targeting metastasis in HCC.


Assuntos
Carcinoma Hepatocelular/patologia , Caveolina 1/fisiologia , Movimento Celular , Regulação Neoplásica da Expressão Gênica , Membro 2 do Grupo A da Subfamília 4 de Receptores Nucleares/metabolismo , Receptor X Retinoide alfa/metabolismo , Sialiltransferases/metabolismo , Animais , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/metabolismo , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Membro 2 do Grupo A da Subfamília 4 de Receptores Nucleares/genética , Receptor X Retinoide alfa/genética , Sialiltransferases/genética , Transdução de Sinais , Células Tumorais Cultivadas , beta-D-Galactosídeo alfa 2-6-Sialiltransferase
4.
Theranostics ; 11(13): 6154-6172, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33995651

RESUMO

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.


Assuntos
Caveolina 1/fisiologia , Terapia Genética , Vetores Genéticos/uso terapêutico , Glaucoma/terapia , Proteína Tirosina Fosfatase não Receptora Tipo 11/antagonistas & inibidores , Retina/patologia , alfa-Globulinas/genética , Animais , Apoptose , Fator Neurotrófico Derivado do Encéfalo/fisiologia , Caveolina 1/deficiência , Caveolina 1/genética , DNA Complementar/genética , Dependovirus/genética , Quinase 1 de Adesão Focal/fisiologia , Técnicas de Silenciamento de Genes , Genes Reporter , Genes Sintéticos , Glaucoma/metabolismo , Glaucoma/patologia , Integrina beta1/fisiologia , Pressão Intraocular , Injeções Intravítreas , Glicoproteínas de Membrana/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Regiões Promotoras Genéticas , Proteína Tirosina Fosfatase não Receptora Tipo 11/biossíntese , Proteína Tirosina Fosfatase não Receptora Tipo 11/genética , Proteínas Tirosina Quinases/fisiologia , Regulação para Cima
5.
Mol Brain ; 14(1): 53, 2021 03 16.
Artigo em Inglês | MEDLINE | ID: mdl-33726791

RESUMO

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.


Assuntos
Caveolina 1/deficiência , Hipocampo/citologia , Proteínas do Tecido Nervoso/deficiência , Neurônios/fisiologia , Transmissão Sináptica/fisiologia , Animais , Caveolina 1/antagonistas & inibidores , Caveolina 1/genética , Caveolina 1/fisiologia , Células Cultivadas , Exocitose/fisiologia , Microdomínios da Membrana , Mutação , Proteínas do Tecido Nervoso/antagonistas & inibidores , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/fisiologia , Ácido Palmítico/metabolismo , Terminações Pré-Sinápticas/química , Terminações Pré-Sinápticas/fisiologia , Processamento de Proteína Pós-Traducional , Interferência de RNA , RNA Interferente Pequeno/genética , Ratos , Ratos Sprague-Dawley
6.
Cancer Lett ; 506: 11-22, 2021 05 28.
Artigo em Inglês | MEDLINE | ID: mdl-33639203

RESUMO

The mechanisms underlying the hypoxic cancer cell-mediated differentiation of cancer-associated fibroblasts (CAFs) have not been elucidated yet. The present study showed that the hypoxic head and neck squamous cell carcinoma (HNSCC) cells promoted CAF-like differentiation through secreting TGF-ß and small extracellular vesicles (sEVs) that contain enhanced levels of miR-192/215 family miRNAs. Caveolin-1 (CAV1), which is a target gene of miR-192/215, inhibited the TGF-ß/SMAD signaling and promoted CAF-like differentiation of the fibroblasts. Restoring the levels of CAV1 inhibited the hypoxic sEV- and TGF-ß-induced CAF-like differentiation. The enhanced levels of miR-192/215 encapsulated in the HNSCC tissue-derived sEVs (but not serum-derived sEVs) indicated hypoxic and aggressive cancer stroma. miR-215 in the tumor tissue-derived sEVs (but not circulating sEVs) was correlated with poor overall survival of patients with HNSCC. This study demonstrated that sEVs function as a "courier" to deliver miRNAs from the cancer cells to the fibroblasts, which promotes the remodeling of the hypoxic tumor microenvironment, and that cancer tissue-derived sEV could potentially serve as a source of biomarker.


Assuntos
Fibroblastos Associados a Câncer/citologia , Vesículas Extracelulares/fisiologia , Neoplasias de Cabeça e Pescoço/patologia , MicroRNAs/fisiologia , Carcinoma de Células Escamosas de Cabeça e Pescoço/patologia , Hipóxia Tumoral/fisiologia , Caveolina 1/fisiologia , Diferenciação Celular , Linhagem Celular Tumoral , Progressão da Doença , Humanos
7.
Cancer Lett ; 505: 1-12, 2021 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-33610729

RESUMO

The aim of this work was to investigate whether Caveolin-1 (Cav-1), a membrane scaffolding protein widely implicated in cancer, may play a role in radiation response in rhabdomyosarcoma (RMS), a pediatric soft tissue tumor. For this purpose, we employed human RD cells in which Cav-1 expression was stably increased via gene transfection. After radiation treatment, we observed that Cav-1 limited cell cycle arrest in the G2/M phase and enhanced resistance to cell senescence and apoptosis via reduction of p21Cip1/Waf1, p16INK4a and Caspase-3 cleavage. After radiotherapy, Cav-1-mediated cell radioresistance was characterized by low accumulation of H2AX foci, as confirmed by Comet assay, marked neutralization of reactive oxygen species (ROS) and enhanced DNA repair via activation of ATM, Ku70/80 complex and DNA-PK. We found that Cav-1-overexpressing RD cells, already under basal conditions, had higher glutathione (GSH) content and greater catalase expression, which conferred protection against acute treatment with hydrogen peroxide. Furthermore, pre-treatment of Cav-1-overexpressing cells with PP2 or LY294002 compounds restored the sensitivity to radiation treatment, indicating a role for Src-kinases and Akt pathways in Cav-1-mediated radioresistance. These findings were confirmed using radioresistant RD and RH30 lines generated by hypofractionated radiotherapy protocol, which showed marked increase of Cav-1, catalase and Akt, and sensitivity to PP2 and LY294002 treatment. In conclusion, these data suggest that concerted activity of Cav-1 and catalase, in cooperation with activation of Src-kinase and Akt pathways, may represent a network of vital mechanisms that allow irradiated RMS cells to evade cell death induced by oxidative stress and DNA damage.


Assuntos
Caveolina 1/fisiologia , Reparo do DNA , Estresse Oxidativo , Tolerância a Radiação , Rabdomiossarcoma/radioterapia , Apoptose , Linhagem Celular Tumoral , Humanos , Proteínas Proto-Oncogênicas c-akt/fisiologia , Espécies Reativas de Oxigênio/metabolismo , Rabdomiossarcoma/metabolismo , Rabdomiossarcoma/patologia , Quinases da Família src/fisiologia
8.
Int J Radiat Biol ; 97(3): 356-366, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33416428

RESUMO

PURPOSE: High LET including alpha radiation-based approaches have been proved as a promising mode for cancer therapy owing to their biophysical and radiobiological advantages compared to photon beams. Studies pertaining to effect of α-radiation on cancer cells are limited to cytotoxic high doses. MATERIALS AND METHODS: In this study, human lung adenocarcinoma (A549) cells were α-irradiated using 241Am α-irradiator and effects of low dose of alpha radiation on these cells was studied under in vitro and in vivo conditions. RESULTS: Clonogenic and other assays showed increased cellular proliferation at lower doses (1.36 and 6.8 cGy) but killing at higher doses (13.6-54.4 cGy). Further studies at low dose of alpha (1.36 cGy) showed increased TGF-ß1 in the conditioned medium (CM) at early time point (24 h) but CM replacement did not affect the clonogenic survival. In these cells, increased phosphorylation of connexin 43 was correlated with decrease in gap-junction communication observed by dye transfer co-culture experiment. A decrease in caveolin-1 but increase in survivin expression was observed in low dose α-irradiated cells. An increase in cyclinD1 and decrease in Bcl-2, the target proteins of survivin, was observed in these cells. Low dose α-irradiated cancer cells transplanted in SCID mice showed significantly higher tumor volume, which was accompanied with an increased fraction of mitotic and PCNA/Ki67 positive cells in these tumor tissues. CONCLUSIONS: Taken together, our results suggest an increase in proliferation and tumor volume at in vitro and in vivo levels, respectively, when A549 cells were irradiated with low dose of α-radiation. These findings may be relevant for a better understanding of radiobiological processes during high LET-based cancer radiotherapy.


Assuntos
Partículas alfa/uso terapêutico , Caveolina 1/fisiologia , Conexina 43/fisiologia , Neoplasias Pulmonares/radioterapia , Survivina/fisiologia , Animais , Caveolina 1/análise , Linhagem Celular Tumoral , Proliferação de Células/efeitos da radiação , Conexina 43/análise , Humanos , Neoplasias Pulmonares/patologia , Camundongos , Transdução de Sinais/fisiologia , Survivina/análise
9.
Invest Ophthalmol Vis Sci ; 61(12): 19, 2020 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-33079993

RESUMO

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.


Assuntos
Caveolina 1/fisiologia , Inflamação/induzido quimicamente , Lipopolissacarídeos/toxicidade , Retina/metabolismo , Retinite/induzido quimicamente , Animais , Western Blotting , Caveolina 1/deficiência , Citocinas/metabolismo , Sinergismo Farmacológico , Eletrorretinografia , Citometria de Fluxo , Imuno-Histoquímica , Inflamação/metabolismo , Inflamação/patologia , Injeções Intravítreas , Espectrometria de Massas , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microscopia Confocal , Nistagmo Optocinético/fisiologia , Proteômica , Retinite/metabolismo , Retinite/patologia , Salmonella typhimurium , Receptor 4 Toll-Like/metabolismo
10.
J Cell Biol ; 219(11)2020 11 02.
Artigo em Inglês | MEDLINE | ID: mdl-33053168

RESUMO

The composition and physical properties of the extracellular matrix (ECM) critically influence tumor progression, but the molecular mechanisms underlying ECM layering are poorly understood. Tumor-stroma interaction critically depends on cell communication mediated by exosomes, small vesicles generated within multivesicular bodies (MVBs). We show that caveolin-1 (Cav1) centrally regulates exosome biogenesis and exosomal protein cargo sorting through the control of cholesterol content at the endosomal compartment/MVBs. Quantitative proteomics profiling revealed that Cav1 is required for exosomal sorting of ECM protein cargo subsets, including Tenascin-C (TnC), and for fibroblast-derived exosomes to efficiently deposit ECM and promote tumor invasion. Cav1-driven exosomal ECM deposition not only promotes local stromal remodeling but also the generation of distant ECM-enriched stromal niches in vivo. Cav1 acts as a cholesterol rheostat in MVBs, determining sorting of ECM components into specific exosome pools and thus ECM deposition. This supports a model by which Cav1 is a central regulatory hub for tumor-stroma interactions through a novel exosome-dependent ECM deposition mechanism.


Assuntos
Caveolina 1/fisiologia , Exossomos/metabolismo , Matriz Extracelular/metabolismo , Fibroblastos/metabolismo , Corpos Multivesiculares/metabolismo , Proteoma/metabolismo , Tenascina/fisiologia , Animais , Fibroblastos/citologia , Camundongos , Camundongos Knockout
11.
Cell Death Dis ; 11(8): 647, 2020 08 03.
Artigo em Inglês | MEDLINE | ID: mdl-32811813

RESUMO

Despite their emerging relevance to fully understand disease pathogenesis, we have as yet a poor understanding as to how biomechanical signals are integrated with specific biochemical pathways to determine cell behaviour. Mesothelial-to-mesenchymal transition (MMT) markers colocalized with TGF-ß1-dependent signaling and yes-associated protein (YAP) activation across biopsies from different pathologies exhibiting peritoneal fibrosis, supporting mechanotransduction as a central driving component of these class of fibrotic lesions and its crosstalk with specific signaling pathways. Transcriptome and proteome profiling of the response of mesothelial cells (MCs) to linear cyclic stretch revealed molecular changes compatible with bona fide MMT, which (i) overlapped with established YAP target gene subsets, and were largely dependent on endogenous TGF-ß1 signaling. Importantly, TGF-ß1 blockade blunts the transcriptional upregulation of these gene signatures, but not the mechanical activation and nuclear translocation of YAP per se. We studied the role therein of caveolin-1 (CAV1), a plasma membrane mechanotransducer. Exposure of CAV1-deficient MCs to cyclic stretch led to a robust upregulation of MMT-related gene programs, which was blunted upon TGF-ß1 inhibition. Conversely, CAV1 depletion enhanced both TGF-ß1 and TGFBRI expression, whereas its re-expression blunted mechanical stretching-induced MMT. CAV1 genetic deficiency exacerbated MMT and adhesion formation in an experimental murine model of peritoneal ischaemic buttons. Taken together, these results support that CAV1-YAP/TAZ fine-tune the fibrotic response through the modulation of MMT, onto which TGF-ß1-dependent signaling coordinately converges. Our findings reveal a cooperation between biomechanical and biochemical signals in the triggering of MMT, representing a novel potential opportunity to intervene mechanically induced disorders coursing with peritoneal fibrosis, such as post-surgical adhesions.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Caveolina 1/metabolismo , Fibrose Peritoneal/metabolismo , Fatores de Transcrição/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/fisiologia , Animais , Caveolina 1/fisiologia , Caveolinas/metabolismo , Modelos Animais de Doenças , Células Epiteliais/metabolismo , Transição Epitelial-Mesenquimal/genética , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Diálise Peritoneal/métodos , Fibrose Peritoneal/genética , Fibrose Peritoneal/patologia , Peritônio/metabolismo , Transdução de Sinais/efeitos dos fármacos , Proteína Smad3/metabolismo , Aderências Teciduais/metabolismo , Fatores de Transcrição/fisiologia , Fator de Crescimento Transformador beta1/metabolismo , Proteínas de Sinalização YAP
12.
Cell Death Dis ; 11(8): 648, 2020 08 03.
Artigo em Inglês | MEDLINE | ID: mdl-32811828

RESUMO

Caveolin-1 (CAV1), is a broadly expressed, membrane-associated scaffolding protein that acts both, as a tumor suppressor and a promoter of metastasis, depending on the type of cancer and stage. CAV1 is downregulated in human tumors, tumor cell lines and oncogene-transformed cells. The tumor suppressor activity of CAV1 is generally associated with its presence at the plasma membrane, where it participates, together with cavins, in the formation of caveolae and also has been suggested to interact with and inhibit a wide variety of proteins through interactions mediated by the scaffolding domain. However, a pool of CAV1 is also located at the endoplasmic reticulum (ER), modulating the secretory pathway in a manner dependent on serine-80 (S80) phosphorylation. In melanoma cells, CAV1 expression suppresses tumor formation, but the protein is largely absent from the plasma membrane and does not form caveolae. Perturbations to the function of the ER are emerging as a central driver of cancer, highlighting the activation of the unfolded protein response (UPR), a central pathway involved in stress mitigation. Here we provide evidence indicating that the expression of CAV1 represses the activation of the UPR in vitro and in solid tumors, reflected in the attenuation of PERK and IRE1α signaling. These effects correlated with increased susceptibility of cells to ER stress and hypoxia. Interestingly, the tumor suppressor activity of CAV1 was abrogated by site-directed mutagenesis of S80, correlating with a reduced ability to repress the UPR. We conclude that the tumor suppression by CAV1 involves the attenuation of the UPR, and identified S80 as essential in this context. This suggests that intracellular CAV1 regulates cancer through alternative signaling outputs.


Assuntos
Caveolina 1/metabolismo , Resposta a Proteínas não Dobradas/fisiologia , Animais , Caveolina 1/fisiologia , Linhagem Celular Tumoral , Membrana Celular/metabolismo , Retículo Endoplasmático/metabolismo , Estresse do Retículo Endoplasmático/fisiologia , Endorribonucleases/metabolismo , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fosforilação , Proteínas Serina-Treonina Quinases/metabolismo , Transdução de Sinais/efeitos dos fármacos , eIF-2 Quinase/metabolismo
13.
Sci Rep ; 10(1): 12816, 2020 07 30.
Artigo em Inglês | MEDLINE | ID: mdl-32733088

RESUMO

Caveolin-1 is the main structural protein of caveolae, small membrane invaginations involved in signal transduction and mechanoprotection. Here, we generated cav1-KO zebrafish lacking Cav1 and caveolae, and investigated the impact of this loss on adult heart function and response to cryoinjury. We found that cardiac function was impaired in adult cav1-KO fish, which showed a significantly decreased ejection fraction and heart rate. Using atomic force microscopy, we detected an increase in the stiffness of epicardial cells and cells of the cortical zone lacking Cav1/caveolae. This loss of cardiac elasticity might explain the decreased cardiac contraction and function. Surprisingly, cav1-KO mutants were able to regenerate their heart after a cryoinjury but showed a transient decrease in cardiomyocyte proliferation.


Assuntos
Fenômenos Fisiológicos Cardiovasculares/genética , Cavéolas , Caveolina 1/genética , Caveolina 1/fisiologia , Elasticidade , Deleção de Genes , Miócitos Cardíacos/patologia , Miócitos Cardíacos/fisiologia , Peixe-Zebra , Animais , Transdução de Sinais/fisiologia
14.
JCI Insight ; 5(19)2020 10 02.
Artigo em Inglês | MEDLINE | ID: mdl-32841217

RESUMO

Increased metabolism distinguishes myofibroblasts or fibrotic lung fibroblasts (fLfs) from the normal lung fibroblasts (nLfs). The mechanism of metabolic activation in fLfs has not been fully elucidated. Furthermore, the antifibrogenic effects of caveolin-1 scaffolding domain peptide CSP/CSP7 involving metabolic reprogramming in fLfs are unclear. We therefore analyzed lactate and succinate levels, as well as the expression of glycolytic enzymes and hypoxia inducible factor-1α (HIF-1α). Lactate and succinate levels, as well as the basal expression of glycolytic enzymes and HIF-1α, were increased in fLfs. These changes were reversed following restoration of p53 or its transcriptional target microRNA-34a (miR-34a) expression in fLfs. Conversely, inhibition of basal p53 or miR-34a increased glucose metabolism, glycolytic enzymes, and HIF-1α in nLfs. Treatment of fLfs or mice having bleomycin- or Ad-TGF-ß1-induced lung fibrosis with CSP/CSP7 reduced the expression of glycolytic enzymes and HIF-1α. Furthermore, inhibition of p53 or miR-34a abrogated CSP/CSP7-mediated restoration of glycolytic flux in fLfs in vitro and in mice with pulmonary fibrosis and lacking p53 or miR-34a expression in fibroblasts in vivo. Our data indicate that dysregulation of glucose metabolism in fLfs is causally linked to loss of basal expression of p53 and miR-34a. Treatment with CSP/CSP7 constrains aberrant glucose metabolism through restoration of p53 and miR-34a.


Assuntos
Caveolina 1/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Glucose/metabolismo , MicroRNAs/metabolismo , Fragmentos de Peptídeos/farmacologia , Fibrose Pulmonar/prevenção & controle , Fator de Crescimento Transformador beta1/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Animais , Caveolina 1/fisiologia , Feminino , Glicólise , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , MicroRNAs/genética , Fragmentos de Peptídeos/fisiologia , Fibrose Pulmonar/induzido quimicamente , Fibrose Pulmonar/metabolismo , Fibrose Pulmonar/patologia , Fator de Crescimento Transformador beta1/genética , Proteína Supressora de Tumor p53/genética
15.
J Endocrinol ; 247(1): 25-38, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32668406

RESUMO

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.


Assuntos
Adipócitos Brancos/metabolismo , Adiponectina/metabolismo , Caveolina 1/fisiologia , Adiponectina/sangue , Adiponectina/genética , Adulto , Idoso , Animais , Caveolina 1/deficiência , Membrana Celular/química , Dieta , Exocitose/fisiologia , Feminino , Expressão Gênica/fisiologia , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Pessoa de Meia-Idade , Obesidade/etiologia , Obesidade/metabolismo
16.
Placenta ; 97: 1-5, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32501218

RESUMO

Although many pregnant women have been infected by coronavirus, the presence of intrauterine vertical transmission has not been conclusively reported yet. What prevents this highly contagious virus from reaching the fetus? Is it only the presence of a strong placental barrier, or is it the natural absence of the some receptor that the viruses use for transmission? We, therefore, need to comprehensively understand the mechanism of action of the mammalian epithelial barriers located in two different organs with functional similarity. The barriers selected as potential targets by SARS-CoV-2 are the alveolo-capillary barrier (ACB), and the syncytio-capillary barrier (SCB). Caveolae are omega-shaped structures located on the cell membrane. They consist of caveolin-1 protein (Cav-1) and are involved in the internalisation of some viruses. By activating leukocytes and nuclear factor-κB, Cav-1 initiates inflammatory reactions. The presence of more than one Cav-1 binding sites on coronavirus is an important finding supporting the possible relationship between SARS-CoV-2-mediated lung injury. While the ACB cells express Cav-1 there is no caveolin expression in syncytiotrophoblasts. In this short review, we will try to explain our hypothesis that the lack of caveolin expression in the SCB is one of the most important physiological mechanisms that prevents vertical transmission of SARS-CoV-2. Since the physiological Cav-1 deficiency appears to prevent acute cell damage treatment algorithms could potentially be developed to block this pathway in the non-pregnant population affected by SARS-CoV-2.


Assuntos
Betacoronavirus/fisiologia , Infecções por Coronavirus/prevenção & controle , Infecções por Coronavirus/transmissão , Doenças Fetais/prevenção & controle , Transmissão Vertical de Doenças Infecciosas/prevenção & controle , Troca Materno-Fetal/imunologia , Pandemias/prevenção & controle , Pneumonia Viral/prevenção & controle , Pneumonia Viral/transmissão , Betacoronavirus/imunologia , COVID-19 , Caveolina 1/fisiologia , Infecções por Coronavirus/imunologia , Epitélio/fisiologia , Epitélio/virologia , Feminino , Doenças Fetais/imunologia , Doenças Fetais/virologia , Células Gigantes/fisiologia , Células Gigantes/virologia , Humanos , Imunidade Inata/fisiologia , Pneumonia Viral/imunologia , Gravidez , Fatores de Risco , SARS-CoV-2 , Internalização do Vírus
17.
Arterioscler Thromb Vasc Biol ; 40(6): 1510-1522, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32349535

RESUMO

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.


Assuntos
Aterosclerose/prevenção & controle , Autofagia/fisiologia , Caveolina 1/deficiência , Endotélio Vascular/fisiopatologia , Vasculite/prevenção & controle , Adenina/análogos & derivados , Adenina/farmacologia , Animais , Aorta/patologia , Aorta/fisiopatologia , Aorta/ultraestrutura , Aterosclerose/etiologia , Autofagia/efeitos dos fármacos , Caveolina 1/análise , Caveolina 1/fisiologia , Dieta Ocidental , Células Endoteliais/química , Células Endoteliais/fisiologia , Células Endoteliais/ultraestrutura , Endotélio Vascular/química , Endotélio Vascular/ultraestrutura , Feminino , Humanos , Masculino , Microdomínios da Membrana/química , Microdomínios da Membrana/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Células NIH 3T3 , Receptores de LDL/deficiência
18.
Cell Reprogram ; 22(4): 189-197, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32315545

RESUMO

Since no study has explored whether exercise could improve impaired proliferation, migration, and angiogenesis of endothelial progenitor cells (EPCs) in animal models or humans with type 2 diabetes, we aimed to explore the effect of different models of exercise on EPC function and expression of caveolin-1, PI3K, and AKT in mice with type 2 diabetes. Male db/db mice (age: 8 weeks) with type 2 diabetes were subjected to aerobic training (AT), resistance training (RT), or combined aerobic and resistance training (AT+RT) 3 or 4 days/week. Mice in the control group remained sedentary with no specific training requirement. Bone marrow-derived EPCs were isolated, and the protein concentrations of caveolin-1, Pi3k, and AKT, and EPC function, were identified in the 1st, 4th, 8th, and 12th weeks of the intervention. Greater increases in proliferation, migration, and angiogenesis were observed in the AT, RT, and AT+RT groups than in the control group. AT+RT was more effective than AT or RT in improving the migratory and angiogenesis function of EPCs in mice with type 2 diabetes and achieved maximum improvement after 8 weeks of intervention. Western blot analysis showed that caveolin-1, p-PI3k, and p-Akt levels were obviously increased in the AT, RT, and AT+RT groups compared with the control group. The expression level of these proteins in the AT+RT group was higher than that in the AT and RT groups. AT+RT may be a helpful reference when choosing exercise methods for the prevention of diabetes-related cardiovascular diseases.


Assuntos
Diabetes Mellitus Tipo 2/metabolismo , Células Progenitoras Endoteliais/fisiologia , Condicionamento Físico Animal , Animais , Caveolina 1/fisiologia , Movimento Celular , Proliferação de Células , Modelos Animais de Doenças , Masculino , Camundongos , Neovascularização Fisiológica , Fosfatidilinositol 3-Quinases/fisiologia , Condicionamento Físico Animal/métodos , Proteínas Proto-Oncogênicas c-akt/fisiologia , Transdução de Sinais
20.
Biochem Biophys Res Commun ; 525(2): 313-318, 2020 04 30.
Artigo em Inglês | MEDLINE | ID: mdl-32087973

RESUMO

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.


Assuntos
Caveolina 1/fisiologia , Lipossomos/metabolismo , Neoplasias/metabolismo , Transcitose , Animais , Carcinoma de Células Renais/irrigação sanguínea , Carcinoma de Células Renais/metabolismo , Caveolina 1/efeitos dos fármacos , Humanos , Nanopartículas/metabolismo , Neoplasias/irrigação sanguínea , RNA Interferente Pequeno/farmacologia , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/antagonistas & inibidores
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA