Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 339
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Cell ; 144(3): 402-13, 2011 Feb 04.
Artigo em Inglês | MEDLINE | ID: mdl-21295700

RESUMO

The functions of caveolae, the characteristic plasma membrane invaginations, remain debated. Their abundance in cells experiencing mechanical stress led us to investigate their role in membrane-mediated mechanical response. Acute mechanical stress induced by osmotic swelling or by uniaxial stretching results in a rapid disappearance of caveolae, in a reduced caveolin/Cavin1 interaction, and in an increase of free caveolins at the plasma membrane. Tether-pulling force measurements in cells and in plasma membrane spheres demonstrate that caveola flattening and disassembly is the primary actin- and ATP-independent cell response that buffers membrane tension surges during mechanical stress. Conversely, stress release leads to complete caveola reassembly in an actin- and ATP-dependent process. The absence of a functional caveola reservoir in myotubes from muscular dystrophic patients enhanced membrane fragility under mechanical stress. Our findings support a new role for caveolae as a physiological membrane reservoir that quickly accommodates sudden and acute mechanical stresses.


Assuntos
Cavéolas/fisiologia , Células Endoteliais/citologia , Células Musculares/fisiologia , Actinas/fisiologia , Trifosfato de Adenosina/fisiologia , Animais , Cavéolas/ultraestrutura , Linhagem Celular , Células Endoteliais/fisiologia , Humanos , Camundongos , Células Musculares/citologia , Estresse Mecânico
2.
Nat Rev Mol Cell Biol ; 14(2): 98-112, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23340574

RESUMO

Caveolae are submicroscopic, plasma membrane pits that are abundant in many mammalian cell types. The past few years have seen a quantum leap in our understanding of the formation, dynamics and functions of these enigmatic structures. Caveolae have now emerged as vital plasma membrane sensors that can respond to plasma membrane stresses and remodel the extracellular environment. Caveolae at the plasma membrane can be removed by endocytosis to regulate their surface density or can be disassembled and their structural components degraded. Coat proteins, called cavins, work together with caveolins to regulate the formation of caveolae but also have the potential to dynamically transmit signals that originate in caveolae to various cellular destinations. The importance of caveolae as protective elements in the plasma membrane, and as membrane organizers and sensors, is highlighted by links between caveolae dysfunction and human diseases, including muscular dystrophies and cancer.


Assuntos
Cavéolas/fisiologia , Membrana Celular/química , Membrana Celular/metabolismo , Animais , Cavéolas/química , Cavéolas/metabolismo , Caveolinas/química , Caveolinas/genética , Caveolinas/metabolismo , Caveolinas/fisiologia , Citoproteção/genética , Citoproteção/fisiologia , Endocitose/genética , Endocitose/fisiologia , Humanos , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Proteínas de Membrana/fisiologia , Modelos Biológicos , Transdução de Sinais/genética , Transdução de Sinais/fisiologia
3.
Nat Rev Mol Cell Biol ; 11(10): 688-99, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20861879

RESUMO

Ten years ago, we wrote a Review on lipid rafts and signalling in the launch issue of Nature Reviews Molecular Cell Biology. At the time, this field was suffering from ambiguous methodology and imprecise nomenclature. Now, new techniques are deepening our insight into the dynamics of membrane organization. Here, we discuss how the field has matured and present an evolving model in which membranes are occupied by fluctuating nanoscale assemblies of sphingolipids, cholesterol and proteins that can be stabilized into platforms that are important in signalling, viral infection and membrane trafficking.


Assuntos
Membrana Celular/fisiologia , Microdomínios da Membrana/fisiologia , Biofísica/métodos , Cavéolas/fisiologia , Cavéolas/ultraestrutura , Membrana Celular/ultraestrutura , Detergentes , Humanos , Complexo Principal de Histocompatibilidade , Espectrometria de Massas/métodos , Microdomínios da Membrana/genética , Microdomínios da Membrana/ultraestrutura , Transdução de Sinais , Solubilidade , Espectrometria de Fluorescência/métodos , Linfócitos T/imunologia , Linfócitos T/fisiologia
4.
Biol Cell ; 113(8): 329-343, 2021 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-33826772

RESUMO

Inside living cells, the remodelling of membrane tubules by actomyosin networks is crucial for processes such as intracellular trafficking or organelle reshaping. In this review, we first present various in vivo situations in which actin affects membrane tubule remodelling, then we recall some results on force production by actin dynamics and on membrane tubules physics. Finally, we show that our knowledge of the underlying mechanisms by which actomyosin dynamics affect tubule morphology has recently been moved forward. This is thanks to in vitro experiments that mimic cellular membranes and actin dynamics and allow deciphering the physics of tubule remodelling in biochemically controlled conditions, and shed new light on tubule shape regulation.


Assuntos
Citoesqueleto de Actina , Membrana Celular , Células Eucarióticas , Citoesqueleto de Actina/fisiologia , Citoesqueleto de Actina/ultraestrutura , Actinas/metabolismo , Cavéolas/fisiologia , Membrana Celular/fisiologia , Membrana Celular/ultraestrutura , Vesículas Revestidas por Clatrina/fisiologia , Endocitose/fisiologia , Células Eucarióticas/fisiologia , Células Eucarióticas/ultraestrutura , Transporte Proteico
5.
Exp Dermatol ; 29(2): 136-148, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31845391

RESUMO

Caveolae are flask-shaped invaginations of the cell membrane rich in cholesterol and sphingomyelin, with caveolin proteins acting as their primary structural components that allow compartmentalization and orchestration of various signalling molecules. In this review, we discuss how pleiotropic functions of caveolin-1 (Cav1) and its intricate roles in numerous cellular functions including lipid trafficking, signalling, cell migration and proliferation, as well as cellular senescence, infection and inflammation, are integral for normal development and functioning of skin and its appendages. We then examine how disruption of the homeostatic levels of Cav1 can lead to development of various cutaneous pathophysiologies including skin cancers, cutaneous fibroses, psoriasis, alopecia, age-related changes in skin and aberrant wound healing and propose how levels of Cav1 may have theragnostic value in skin physiology/pathophysiology.


Assuntos
Cavéolas/fisiologia , Caveolina 1/metabolismo , Neoplasias Cutâneas/metabolismo , Fenômenos Fisiológicos da Pele , Pele/metabolismo , Infecções Bacterianas/metabolismo , Movimento Celular , Proliferação de Células , Senescência Celular , Fibrose/metabolismo , Cabelo/metabolismo , Humanos , Inflamação/metabolismo , Metabolismo dos Lipídeos , Psoríase/metabolismo , Transdução de Sinais , Pele/patologia , Cicatrização
6.
Proc Natl Acad Sci U S A ; 114(20): E4010-E4019, 2017 05 16.
Artigo em Inglês | MEDLINE | ID: mdl-28461495

RESUMO

The fibroblast growth factor (FGF) homologous factor FGF13, a noncanonical FGF, has been best characterized as a voltage-gated Na+ channel auxiliary subunit. Other cellular functions have been suggested, but not explored. In inducible, cardiac-specific Fgf13 knockout mice, we found-even in the context of the expected reduction in Na+ channel current-an unanticipated protection from the maladaptive hypertrophic response to pressure overload. To uncover the underlying mechanisms, we searched for components of the FGF13 interactome in cardiomyocytes and discovered the complete set of the cavin family of caveolar coat proteins. Detailed biochemical investigations showed that FGF13 acts as a negative regulator of caveolae abundance in cardiomyocytes by controlling the relative distribution of cavin 1 between the sarcolemma and cytosol. In cardiac-specific Fgf13 knockout mice, cavin 1 redistribution to the sarcolemma stabilized the caveolar structural protein caveolin 3. The consequent increase in caveolae density afforded protection against pressure overload-induced cardiac dysfunction by two mechanisms: (i) enhancing cardioprotective signaling pathways enriched in caveolae, and (ii) increasing the caveolar membrane reserve available to buffer membrane tension. Thus, our results uncover unexpected roles for a FGF homologous factor and establish FGF13 as a regulator of caveolae-mediated mechanoprotection and adaptive hypertrophic signaling.


Assuntos
Cardiomegalia/metabolismo , Cavéolas/fisiologia , Caveolinas/metabolismo , Fatores de Crescimento de Fibroblastos/metabolismo , Miócitos Cardíacos/fisiologia , Animais , Cardiomegalia/etiologia , Cardiomegalia/patologia , Modelos Animais de Doenças , Feminino , Fatores de Crescimento de Fibroblastos/genética , Fibrose , Masculino , Microdomínios da Membrana/metabolismo , Camundongos Knockout , Miocárdio/patologia , Miócitos Cardíacos/ultraestrutura , Pressão , Sarcolema/fisiologia , Sarcolema/ultraestrutura
7.
Proc Natl Acad Sci U S A ; 113(28): 7834-9, 2016 07 12.
Artigo em Inglês | MEDLINE | ID: mdl-27342861

RESUMO

Lipid membrane curvature plays important roles in various physiological phenomena. Curvature-regulated dynamic membrane remodeling is achieved by the interaction between lipids and proteins. So far, several membrane sensing/sculpting proteins, such as Bin/amphiphysin/Rvs (BAR) proteins, are reported, but there remains the possibility of the existence of unidentified membrane-deforming proteins that have not been uncovered by sequence homology. To identify new lipid membrane deformation proteins, we applied liposome-based microscopic screening, using unbiased-darkfield microscopy. Using this method, we identified phospholipase Cß1 (PLCß1) as a new candidate. PLCß1 is well characterized as an enzyme catalyzing the hydrolysis of phosphatidylinositol-4,5-bisphosphate (PIP2). In addition to lipase activity, our results indicate that PLCß1 possessed the ability of membrane tubulation. Lipase domains and inositol phospholipids binding the pleckstrin homology (PH) domain of PLCß1 were not involved, but the C-terminal sequence was responsible for this tubulation activity. Computational modeling revealed that the C terminus displays the structural homology to the BAR domains, which is well known as a membrane sensing/sculpting domain. Overexpression of PLCß1 caused plasma membrane tubulation, whereas knockdown of the protein reduced the number of caveolae and induced the evagination of caveolin-rich membrane domains. Taken together, our results suggest a new function of PLCß1: plasma membrane remodeling, and in particular, caveolae formation.


Assuntos
Cavéolas/fisiologia , Fosfolipase C beta/metabolismo , Animais , Lipossomos , Camundongos , Camundongos Endogâmicos C57BL , Células Swiss 3T3
8.
Vet Res ; 49(1): 16, 2018 02 13.
Artigo em Inglês | MEDLINE | ID: mdl-29439726

RESUMO

Cell-penetrating peptide (CPP) is a promising cargo for delivering bioactive molecules. In this study, the N terminus of VP1 from chicken anemia virus, designated as CVP1, was found to carry enriched arginine residues with α-helix. By confocal imaging, flow cytometry and MTT assay, we identified CVP1 as a novel, safe and efficient CPP. CVP1-FITC peptide could entry different types of cells tested with dose dependence, but without cytotoxic effects. Compared with TAT-FITC peptide, the CVP1-FITC peptide showed much higher cell-penetrating activity. Moreover, CVP1 could successfully deliver ß-glycosidase, poly (I:C) and plasmid into HCT116 cells. Inhibitors and temperature sensitivity analysis further indicated that the cell-penetrating activity of CVP1 was based on ATP-dependent and caveolae-mediated endocytosis. All these data demonstrate that CVP1 has efficient cell-penetrating activity and great potential for developing a novel delivery vector.


Assuntos
Cavéolas/fisiologia , Peptídeos Penetradores de Células/administração & dosagem , Vírus da Anemia da Galinha/fisiologia , Animais , Cavéolas/virologia , Linhagem Celular , Galinhas , Cães , Sistemas de Liberação de Medicamentos/veterinária , Endocitose/fisiologia , Células HCT116 , Células HEK293 , Humanos , Células Madin Darby de Rim Canino
9.
Biophys J ; 113(5): 1047-1059, 2017 Sep 05.
Artigo em Inglês | MEDLINE | ID: mdl-28877488

RESUMO

Caveolae are signal transduction centers, yet their subcellular distribution and preservation in cardiac myocytes after cell isolation are not well documented. Here, we quantify caveolae located within 100 nm of the outer cell surface membrane in rabbit single-ventricular cardiomyocytes over 8 h post-isolation and relate this to the presence of caveolae in intact tissue. Hearts from New Zealand white rabbits were either chemically fixed by coronary perfusion or enzymatically digested to isolate ventricular myocytes, which were subsequently fixed at 0, 3, and 8 h post-isolation. In live cells, the patch-clamp technique was used to measure whole-cell plasma membrane capacitance, and in fixed cells, caveolae were quantified by transmission electron microscopy. Changes in cell-surface topology were assessed using scanning electron microscopy. In fixed ventricular myocardium, dual-axis electron tomography was used for three-dimensional reconstruction and analysis of caveolae in situ. The presence and distribution of surface-sarcolemmal caveolae in freshly isolated cells matches that of intact myocardium. With time, the number of surface-sarcolemmal caveolae decreases in isolated cardiomyocytes. This is associated with a gradual increase in whole-cell membrane capacitance. Concurrently, there is a significant increase in area, diameter, and circularity of sub-sarcolemmal mitochondria, indicative of swelling. In addition, electron tomography data from intact heart illustrate the regular presence of caveolae not only at the surface sarcolemma, but also on transverse-tubular membranes in ventricular myocardium. Thus, caveolae are dynamic structures, present both at surface-sarcolemmal and transverse-tubular membranes. After cell isolation, the number of surface-sarcolemmal caveolae decreases significantly within a time frame relevant for single-cell research. The concurrent increase in cell capacitance suggests that membrane incorporation of surface-sarcolemmal caveolae underlies this, but internalization and/or micro-vesicle loss to the extracellular space may also contribute. Given that much of the research into cardiac caveolae-dependent signaling utilizes isolated cells, and since caveolae-dependent pathways matter for a wide range of other study targets, analysis of isolated cell data should take the time post-isolation into account.


Assuntos
Cavéolas , Ventrículos do Coração/citologia , Miócitos Cardíacos/citologia , Animais , Cavéolas/fisiologia , Separação Celular , Células Cultivadas , Capacitância Elétrica , Tomografia com Microscopia Eletrônica , Imageamento Tridimensional , Microscopia Eletrônica de Varredura , Microscopia Eletrônica de Transmissão , Mitocôndrias/fisiologia , Modelos Biológicos , Miócitos Cardíacos/fisiologia , Técnicas de Patch-Clamp , Coelhos , Sarcolema/fisiologia , Propriedades de Superfície , Fixação de Tecidos
10.
Am J Physiol Cell Physiol ; 312(4): C459-C477, 2017 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-28122734

RESUMO

Caveolins (Cavs) are ~20 kDa scaffolding proteins that assemble as oligomeric complexes in lipid raft domains to form caveolae, flask-shaped plasma membrane (PM) invaginations. Caveolae ("little caves") require lipid-lipid, protein-lipid, and protein-protein interactions that can modulate the localization, conformational stability, ligand affinity, effector specificity, and other functions of proteins that are partners of Cavs. Cavs are assembled into small oligomers in the endoplasmic reticulum (ER), transported to the Golgi for assembly with cholesterol and other oligomers, and then exported to the PM as an intact coat complex. At the PM, cavins, ~50 kDa adapter proteins, oligomerize into an outer coat complex that remodels the membrane into caveolae. The structure of caveolae protects their contents (i.e., lipids and proteins) from degradation. Cellular changes, including signal transduction effects, can destabilize caveolae and produce cavin dissociation, restructuring of Cav oligomers, ubiquitination, internalization, and degradation. In this review, we provide a perspective of the life cycle (biogenesis, degradation), composition, and physiologic roles of Cavs and caveolae and identify unanswered questions regarding the roles of Cavs and cavins in caveolae and in regulating cell physiology.1.


Assuntos
Cavéolas/fisiologia , Caveolinas/metabolismo , Fenômenos Fisiológicos Celulares/fisiologia , Proteínas de Membrana/metabolismo , Transporte Proteico/fisiologia , Proteínas de Ligação a RNA/metabolismo , Animais , Humanos , Modelos Biológicos
11.
Curr Diab Rep ; 17(3): 19, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-28283950

RESUMO

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


Assuntos
Caveolina 1/fisiologia , Nefropatias Diabéticas/etiologia , Animais , Cavéolas/fisiologia , Caveolina 1/antagonistas & inibidores , Nefropatias Diabéticas/tratamento farmacológico , Nefropatias Diabéticas/fisiopatologia , Matriz Extracelular/metabolismo , Humanos , Estresse Oxidativo , Transdução de Sinais/fisiologia
12.
Birth Defects Res C Embryo Today ; 108(1): 45-64, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26991990

RESUMO

Caveolae, an almost ubiquitous, structural component of the plasma membrane, play a critical role in many functions essential for proper cell function, including membrane trafficking, signal transduction, extracellular matrix remodeling, and tissue regeneration. Three main types of caveolin proteins have been identified from caveolae since the discovery of caveolin-1 in the early 1990s. All three (Cav-1, Cav-2, and Cav-3) play crucial roles in mammalian physiology, and can effect pathogenesis in a wide range of human diseases. While many biological activities of caveolins have been uncovered since its discovery, their role and regulation in embryonic develop remain largely poorly understood, although there is increasing evidence that caveolins may be linked to lung and brain birth defects. Further investigations are clearly needed to decipher how caveolae/caveolins mediate cellular functions and activities of normal embryogenesis and how their perturbations contribute to developmental disorders.


Assuntos
Cavéolas/patologia , Cavéolas/fisiologia , Caveolina 1/metabolismo , Animais , Membrana Celular/metabolismo , Membrana Celular/fisiologia , Desenvolvimento Embrionário/fisiologia , Humanos , Transdução de Sinais/fisiologia
13.
J Physiol ; 594(20): 5941-5957, 2016 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-27121987

RESUMO

KEY POINTS: Hyperfibrinogenaemia (HFg) results in vascular remodelling, and fibrinogen (Fg) and amyloid ß (Aß) complex formation is a hallmark of Alzheimer's disease. However, the interconnection of these effects, their mechanisms and implications in cerebrovascular diseases are not known. Using a mouse model of HFg, we showed that at an elevated blood level, Fg increases cerebrovascular permeability via mainly caveolar protein transcytosis. This enhances deposition of Fg in subendothelial matrix and interstitium making the immobilized Fg a readily accessible substrate for binding Aß and cellular prion protein (PrPC ), the protein that is thought to have a greater effect on memory than Aß. We showed that enhanced formation of Fg-Aß and Fg-PrPC complexes are associated with reduction in short-term memory. The present study delineates a new mechanistic pathway for vasculo-neuronal dysfunctions found in inflammatory cardiovascular and cerebrovascular diseases associated with an elevated blood level of Fg. ABSTRACT: Many cardiovascular diseases are associated with inflammation and as such are accompanied by an increased blood level of fibrinogen (Fg). Besides its well-known prothrombotic effects Fg seems to have other destructive roles in developing microvascular dysfunction that include changes in vascular reactivity and permeability. Increased permeability of brain microvessels has the most profound effects as it may lead to cerebrovascular remodelling and result in memory reduction. The goal of the present study was to define mechanisms of cerebrovascular permeability and associated reduction in memory induced by elevated blood content of Fg. Genetically modified, transgenic hyperfibrinogenic (HFg) mice were used to study cerebrovascular transcellular and paracellular permeability in vivo. The extent of caveolar formation and the role of caveolin-1 signalling were evaluated by immunohistochemistry (IHC) and Western blot (WB) analysis in brain samples from experimental animals. Formation of Fg complexes with amyloid ß (Aß) and with cellular prion protein (PrPC ) were also assessed with IHC and WB analysis. Short-term memory of mice was assessed by novel object recognition and Y-maze tests. Caveolar protein transcytosis was found to have a prevailing role in overall increased cerebrovascular permeability in HFg mice. These results were associated with enhanced formation of caveolae. Increased formation of Fg-PrPC and Fg-Aß complexes were correlated with reduction in short-term memory in HFg mice. Using the model of hyperfibrinogenaemia, the present study shows a novel mechanistic pathway of inflammation-induced and Fg-mediated reduction in short-term memory.


Assuntos
Circulação Cerebrovascular/fisiologia , Transtornos Cerebrovasculares/metabolismo , Transtornos Cerebrovasculares/patologia , Fibrinogênio/metabolismo , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/metabolismo , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Cavéolas/metabolismo , Cavéolas/fisiologia , Caveolina 1/metabolismo , Memória de Curto Prazo/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Permeabilidade
14.
Am J Physiol Lung Cell Mol Physiol ; 310(11): L1078-87, 2016 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-27016585

RESUMO

Caveolae are stiff plasma membrane microdomains implicated in various cell response mechanisms like Ca(2+) signaling and mechanotransduction. Pulmonary arterial smooth muscle cells (PASMC) transduce mechanical stimuli into Ca(2+) increase via plasma membrane stretch-activated channels (SAC). This mechanotransduction process is modified in pulmonary hypertension (PH) during which stretch forces are increased by the increase in arterial blood pressure. We propose to investigate how caveolae are involved in the pathophysiology of PH and particularly in mechanotransduction. PASMC were freshly isolated from control rats (Ctrl rats) and rats suffering from PH induced by 3 wk of chronic hypoxia (CH rats). Using a caveolae disrupter (methyl-ß-cyclodextrin), we showed that SAC activity measured by patch-clamp, stretch-induced Ca(2+) increase measured with indo-1 probe and pulmonary arterial ring contraction to osmotic shock are enhanced in Ctrl rats when caveolae are disrupted. In CH rats, SAC activity, Ca(2+), and contraction responses to stretch are all higher compared with Ctrl rats. However, in contrast to Ctrl rats, caveolae disruption in CH-PASMC, reduces SAC activity, Ca(2+) responses to stretch and arterial contractions. Furthermore, by means of immunostainings and transmission electron microscopy, we observed that caveolae and caveolin-1 are expressed in PASMC from both Ctrl and CH rats and localize close to subplasmalemmal sarcoplasmic reticulum (ryanodine receptors) and mitochondria, thus facilitating Ca(2+) exchanges, particularly in CH. In conclusion, caveolae are implicated in mechanotransduction in Ctrl PASMC by buffering mechanical forces. In PH-PASMC, caveolae form a distinct Ca(2+) store facilitating Ca(2+) coupling between SAC and sarcoplasmic reticulum.


Assuntos
Cavéolas/fisiologia , Hipertensão Pulmonar/metabolismo , Mecanotransdução Celular , Animais , Sinalização do Cálcio , Caveolina 1/metabolismo , Células Cultivadas , Hipertensão Pulmonar/patologia , Masculino , Miócitos de Músculo Liso/metabolismo , Miócitos de Músculo Liso/patologia , Artéria Pulmonar/metabolismo , Artéria Pulmonar/patologia , Ratos Wistar
15.
J Physiol ; 593(20): 4561-74, 2015 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-26260249

RESUMO

Weightlessness induces the functional remodelling of arteries, but the changes to angiotensin II (Ang II)-elicited vasoconstriction and the underlying mechanism have never been reported. Caveolae are invaginations of the cell membrane crucial for the contraction of vascular smooth muscle cells, so we investigated the adaptation of Ang II-elicited vasoconstriction to simulated weightlessness and the role of caveolae in it. The 4 week hindlimb unweighted (HU) rat was used to simulate the effects of weightlessness. Ang II-elicited vasoconstriction was measured by isometric force recording. The morphology of caveolae was examined by transmission electron microscope. The binding of the angiotensin II type 1 receptor (AT1 ) and caveolin-1 (cav-1) was examined by coimmunoprecipitation and Western blot. We found that the maximal developing force (E(max)) of Ang II-elicited vasoconstriction was decreased in abdominal aorta by 30.6%, unchanged in thoracic aorta and increased in carotid artery by 17.9% after HU, while EC50 of the response was increased in all three arteries (P < 0.05). AT1 desensitization upon activation was significantly reduced by HU in all three arteries, as was the number of caveolae (P < 0.05). Furthermore, Ang II promoted the binding of AT1 and cav-1 significantly in control but not HU arteries. Both the number of caveolae and the binding of AT1 and cav-1 in HU arteries were restored by cholesterol pretreatment which also reinstated the change in EC50 as well as the level of AT1 desensitization. These results indicate that modified caveolae in vascular smooth muscle cells could interfere with the binding of AT1 and cav-1 mediating the adaptation of Ang II-elicited vasoconstriction to HU.


Assuntos
Angiotensina II/farmacologia , Aorta Abdominal/fisiologia , Aorta Torácica/fisiologia , Artérias Carótidas/fisiologia , Cavéolas/fisiologia , Vasoconstrição/efeitos dos fármacos , Animais , Aorta Abdominal/efeitos dos fármacos , Aorta Torácica/efeitos dos fármacos , Artérias Carótidas/efeitos dos fármacos , Caveolina 1/metabolismo , Colesterol/farmacologia , Membro Posterior , Elevação dos Membros Posteriores/fisiologia , Masculino , Músculo Liso Vascular/fisiologia , Miócitos de Músculo Liso/fisiologia , Ratos Sprague-Dawley , Receptor Tipo 1 de Angiotensina/metabolismo , Vasoconstrição/fisiologia , Ausência de Peso
16.
Biochem Biophys Res Commun ; 463(4): 928-33, 2015 Aug 07.
Artigo em Inglês | MEDLINE | ID: mdl-26071356

RESUMO

In bone cells, integrins on the cellular surface are the primary sensors of their mechanical environment. Although gravitational changes are known to affect the adhesion and functions of bone cells, whether integrins respond to hypergravity in osteoblasts remains unclear. In this work, we demonstrate that exposure to a hypergravitational environment (20 × g via centrifugation) resulted in the concentration of ß1, but not ß3, integrin on the cell membrane of osteoblast-like (MC3T3-E1) cells. Notably, the total expression of both integrins was unaffected by the hypergravitational environment. In addition, caveolin-dependent endocytosis was discovered to be involved in the regulation of the enrichment of ß1 integrin on the cell surface after stimulation by hypergravity. These findings could aid in the improvement of our understanding of the mechanisms underlying the effects of different gravitational forces on the human body.


Assuntos
Cavéolas/fisiologia , Endocitose , Hipergravidade , Integrina beta1/metabolismo , Osteoblastos/metabolismo , Células 3T3 , Animais , Sequência de Bases , Primers do DNA , Integrina beta3/metabolismo , Camundongos , Reação em Cadeia da Polimerase Via Transcriptase Reversa
17.
FASEB J ; 28(8): 3769-79, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24812087

RESUMO

Impaired adipogenesis renders an adipose tissue unable to expand, leading to lipotoxicity and conditions such as diabetes and cardiovascular disease. While factors important for adipogenesis have been studied extensively, those that set the limits of adipose tissue expansion remain undetermined. Feeding a Western-type diet to apolipoprotein E2 knock-in mice, a model of metabolic syndrome, produced 3 groups of equally obese mice: mice with normal glucose tolerance, hyperinsulinemic yet glucose-tolerant mice, and prediabetic mice with impaired glucose tolerance and reduced circulating insulin. Using proteomics, we compared subcutaneous adipose tissues from mice in these groups and found that the expression of PTRF (polymerase I and transcript release factor) associated selectively with their glucose tolerance status. Lentiviral and pharmacologically overexpressed PTRF, whose function is critical for caveola formation, compromised adipocyte differentiation of cultured 3T3-L1cells. In human adipose tissue, PTRF mRNA levels positively correlated with markers of lipolysis and cellular senescence. Furthermore, a negative relationship between telomere length and PTRF mRNA levels was observed in human subcutaneous fat. PTRF is associated with limited adipose tissue expansion underpinning the key role of caveolae in adipocyte regulation. Furthermore, PTRF may be a suitable adipocyte marker for predicting pathological obesity and inform clinical management.


Assuntos
Adipócitos/patologia , Adipogenia/fisiologia , Cavéolas/fisiologia , Dieta/efeitos adversos , Intolerância à Glucose/etiologia , Hiperinsulinismo/etiologia , Obesidade/etiologia , Estado Pré-Diabético/etiologia , Proteínas de Ligação a RNA/fisiologia , Gordura Subcutânea/metabolismo , Células 3T3-L1 , Adiponectina/sangue , Animais , Aorta/patologia , Apolipoproteína E2/genética , Senescência Celular , Feminino , Perfilação da Expressão Gênica , Técnicas de Introdução de Genes , Intolerância à Glucose/sangue , Intolerância à Glucose/patologia , Humanos , Hiperinsulinismo/sangue , Hiperinsulinismo/patologia , Resistência à Insulina , Lipólise , Fígado/química , Masculino , Proteínas de Membrana , Camundongos , Camundongos Endogâmicos C57BL , Obesidade/sangue , Obesidade/classificação , Obesidade/patologia , Estado Pré-Diabético/sangue , Estado Pré-Diabético/patologia , Gravidez , RNA Mensageiro/biossíntese , Proteínas Recombinantes de Fusão/metabolismo , Gordura Subcutânea/patologia , Encurtamento do Telômero , Triglicerídeos/metabolismo
18.
J Mol Cell Cardiol ; 76: 265-74, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25257915

RESUMO

Mechanical stretch of cardiac muscle modulates action potential propagation velocity, causing potentially arrhythmogenic conduction slowing. The mechanisms by which stretch alters cardiac conduction remain unknown, but previous studies suggest that stretch can affect the conformation of caveolae in myocytes and other cell types. We tested the hypothesis that slowing of action potential conduction due to cardiac myocyte stretch is dependent on caveolae. Cardiac action potential propagation velocities, measured by optical mapping in isolated mouse hearts and in micropatterned mouse cardiomyocyte cultures, decreased reversibly with volume loading or stretch, respectively (by 19±5% and 26±4%). Stretch-dependent conduction slowing was not altered by stretch-activated channel blockade with gadolinium or by GsMTx-4 peptide, but was inhibited when caveolae were disrupted via genetic deletion of caveolin-3 (Cav3 KO) or membrane cholesterol depletion by methyl-ß-cyclodextrin. In wild-type mouse hearts, stretch coincided with recruitment of caveolae to the sarcolemma, as observed by electron microscopy. In myocytes from wild-type but not Cav3 KO mice, stretch significantly increased cell membrane capacitance (by 98±64%), electrical time constant (by 285±149%), and lipid recruitment to the bilayer (by 84±39%). Recruitment of caveolae to the sarcolemma during physiologic cardiomyocyte stretch slows ventricular action potential propagation by increasing cell membrane capacitance.


Assuntos
Cavéolas/fisiologia , Sistema de Condução Cardíaco , Miócitos Cardíacos/fisiologia , Potenciais de Ação , Animais , Caveolina 3/genética , Caveolina 3/metabolismo , Células Cultivadas , Ventrículos do Coração/citologia , Mecanotransdução Celular , Camundongos Endogâmicos C57BL , Camundongos Knockout , Miócitos Cardíacos/ultraestrutura , Técnicas de Patch-Clamp , Sarcolema/metabolismo , Função Ventricular , Pressão Ventricular
19.
J Cell Sci ; 125(Pt 13): 3097-113, 2012 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-22454521

RESUMO

The biology of caveolin-1 (Cav1)/caveolae is intimately linked to actin dynamics and adhesion receptors. Caveolar domains are organized in hierarchical levels of complexity from curved or flattened caveolae to large, higher-order caveolar rosettes. We report that stress fibers controlled by Abl kinases and mDia1 determine the level of caveolar domain organization, which conditions the subsequent inward trafficking of caveolar domains induced upon loss of cell adhesion from the extracellular matrix. Abl-deficient cells have fewer stress fibers, a smaller pool of stress-fiber co-aligned Cav1 and increased clustering of Cav1/caveolae at the cell surface. Defective caveolar linkage to stress fibers prevents the formation of big caveolar rosettes upon loss of cell adhesion, correlating with a lack of inward trafficking. Live imaging of stress fibers and Cav1 showed that the actin-linked Cav1 pool loses its spatial organization in the absence of actin polymerization and is dragged and clustered by depolymerizing filaments. We identified mDia1 as the actin polymerization regulator downstream of Abl kinases that controls the stress-fiber-linked Cav1 pool. mDia1 knockdown results in Cav1/caveolae clustering and defective inward trafficking upon loss of cell adhesion. By contrast, cell elongation imposed by the excess of stress fibers induced by active mDia1 flattens caveolae. Furthermore, active mDia1 rescues the actin co-aligned Cav1 pool and Cav1 inward trafficking upon loss of adhesion in Abl-deficient cells. Thus, caveolar domain organization and trafficking are tightly coupled to adhesive and stress fiber regulatory pathways.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Cavéolas/metabolismo , Caveolina 1/metabolismo , Proteínas Tirosina Quinases/metabolismo , Actinas/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Cavéolas/fisiologia , Cavéolas/ultraestrutura , Caveolina 1/genética , Adesão Celular , Clonagem Molecular , Forminas , Técnicas de Silenciamento de Genes , Proteínas de Fluorescência Verde/metabolismo , Células HEK293 , Células HeLa , Humanos , Camundongos , Microscopia Eletrônica , Plasmídeos/genética , Plasmídeos/metabolismo , Polimerização , Estrutura Terciária de Proteína , Transporte Proteico , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Fibras de Estresse/metabolismo , Fibras de Estresse/fisiologia
20.
Anesthesiology ; 121(3): 538-48, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24821070

RESUMO

BACKGROUND: Caveolae are a nexus for protective signaling. Trafficking of caveolin to mitochondria is essential for adaptation to cellular stress though the trafficking mechanisms remain unknown. The authors hypothesized that G protein-coupled receptor/inhibitory G protein (Gi) activation leads to caveolin trafficking to mitochondria. METHODS: Mice were exposed to isoflurane or oxygen vehicle (30 min, ± 36 h pertussis toxin pretreatment, an irreversible Gi inhibitor). Caveolin trafficking, cardioprotective "survival kinase" signaling, mitochondrial function, and ultrastructure were assessed. RESULTS: Isoflurane increased cardiac caveolae (n = 8 per group; data presented as mean ± SD for Ctrl versus isoflurane; [caveolin-1: 1.78 ± 0.12 vs. 3.53 ± 0.77; P < 0.05]; [caveolin-3: 1.68 ± 0.29 vs. 2.67 ± 0.46; P < 0.05]) and mitochondrial caveolin levels (n = 16 per group; [caveolin-1: 0.87 ± 0.18 vs. 1.89 ± .19; P < 0.05]; [caveolin-3: 1.10 ± 0.29 vs. 2.26 ± 0.28; P < 0.05]), and caveolin-enriched mitochondria exhibited improved respiratory function (n = 4 per group; [state 3/complex I: 10.67 ± 1.54 vs. 37.6 ± 7.34; P < 0.05]; [state 3/complex II: 37.19 ± 4.61 vs. 71.48 ± 15.28; P < 0.05]). Isoflurane increased phosphorylation of survival kinases (n = 8 per group; [protein kinase B: 0.63 ± 0.20 vs. 1.47 ± 0.18; P < 0.05]; [glycogen synthase kinase 3ß: 1.23 ± 0.20 vs. 2.35 ± 0.20; P < 0.05]). The beneficial effects were blocked by pertussis toxin. CONCLUSIONS: Gi proteins are involved in trafficking caveolin to mitochondria to enhance stress resistance. Agents that target Gi activation and caveolin trafficking may be viable cardioprotective agents.


Assuntos
Caveolinas/metabolismo , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/fisiologia , Mitocôndrias/metabolismo , Animais , Cavéolas/efeitos dos fármacos , Cavéolas/fisiologia , Quinase 3 da Glicogênio Sintase/metabolismo , Glicogênio Sintase Quinase 3 beta , Isoflurano/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/ultraestrutura , Traumatismo por Reperfusão Miocárdica/prevenção & controle , Toxina Pertussis/farmacologia , Transporte Proteico , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA