Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 97
Filtrar
1.
Cell ; 150(4): 752-63, 2012 Aug 17.
Artigo em Inglês | MEDLINE | ID: mdl-22901807

RESUMO

Caveolin plays an essential role in the formation of characteristic surface pits, caveolae, which cover the surface of many animal cells. The fundamental principles of caveola formation are only slowly emerging. Here we show that caveolin expression in a prokaryotic host lacking any intracellular membrane system drives the formation of cytoplasmic vesicles containing polymeric caveolin. Vesicle formation is induced by expression of wild-type caveolins, but not caveolin mutants defective in caveola formation in mammalian systems. In addition, cryoelectron tomography shows that the induced membrane domains are equivalent in size and caveolin density to native caveolae and reveals a possible polyhedral arrangement of caveolin oligomers. The caveolin-induced vesicles or heterologous caveolae (h-caveolae) form by budding in from the cytoplasmic membrane, generating a membrane domain with distinct lipid composition. Periplasmic solutes are encapsulated in the budding h-caveola, and purified h-caveolae can be tailored to be targeted to specific cells of interest.


Assuntos
Cavéolas/metabolismo , Cavéolas/ultraestrutura , Caveolinas/metabolismo , Escherichia coli , Mamíferos/metabolismo , Animais , Linhagem Celular Tumoral , Membrana Celular/metabolismo , Membrana Celular/ultraestrutura , Humanos
2.
Proc Natl Acad Sci U S A ; 117(48): 30476-30487, 2020 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-33214152

RESUMO

None of the current superresolution microscopy techniques can reliably image the changes in endogenous protein nanoclustering dynamics associated with specific conformations in live cells. Single-domain nanobodies have been invaluable tools to isolate defined conformational states of proteins, and we reasoned that expressing these nanobodies coupled to single-molecule imaging-amenable tags could allow superresolution analysis of endogenous proteins in discrete conformational states. Here, we used anti-GFP nanobodies tagged with photoconvertible mEos expressed as intrabodies, as a proof-of-concept to perform single-particle tracking on a range of GFP proteins expressed in live cells, neurons, and small organisms. We next expressed highly specialized nanobodies that target conformation-specific endogenous ß2-adrenoreceptor (ß2-AR) in neurosecretory cells, unveiling real-time mobility behaviors of activated and inactivated endogenous conformers during agonist treatment in living cells. We showed that activated ß2-AR (Nb80) is highly immobile and organized in nanoclusters. The Gαs-GPCR complex detected with Nb37 displayed higher mobility with surprisingly similar nanoclustering dynamics to that of Nb80. Activated conformers are highly sensitive to dynamin inhibition, suggesting selective targeting for endocytosis. Inactivated ß2-AR (Nb60) molecules are also largely immobile but relatively less sensitive to endocytic blockade. Expression of single-domain nanobodies therefore provides a unique opportunity to capture highly transient changes in the dynamic nanoscale organization of endogenous proteins.


Assuntos
Modelos Moleculares , Conformação Proteica , Receptores Adrenérgicos beta 2/química , Imagem Individual de Molécula , Anticorpos de Domínio Único/química , Animais , Linhagem Celular , Endocitose , Imunofluorescência , Expressão Gênica , Genes Reporter , Humanos , Proteínas de Membrana/química , Proteínas de Membrana/metabolismo , Camundongos , Ligação Proteica , Receptores Adrenérgicos beta 2/genética , Receptores Adrenérgicos beta 2/metabolismo , Proteínas Recombinantes de Fusão , Imagem Individual de Molécula/métodos , Anticorpos de Domínio Único/metabolismo , Peixe-Zebra
3.
Genet Med ; 24(2): 332-343, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34906470

RESUMO

PURPOSE: In Mendelian disease diagnosis, variant analysis is a repetitive, error-prone, and time consuming process. To address this, we have developed the Mendelian Analysis Toolkit (MATK), a configurable, automated variant ranking program. METHODS: MATK aggregates variant information from multiple annotation sources and uses expert-designed rules with parameterized weights to produce a ranked list of potentially causal solutions. MATK performance was measured by a comparison between MATK-aided and human-domain expert analyses of 1060 families with inherited retinal degeneration (IRD), analyzed using an IRD-specific gene panel (589 individuals) and exome sequencing (471 families). RESULTS: When comparing MATK-assisted analysis with expert curation in both the IRD-specific gene panel and exome sequencing (1060 subjects), 97.3% of potential solutions found by experts were also identified by the MATK-assisted analysis (541 solutions identified with MATK of 556 solutions found by conventional analysis). Furthermore, MATK-assisted analysis identified 114 additional potential solutions from the 504 cases unsolved by conventional analysis. CONCLUSION: MATK expedites the process of identification of likely solving variants in Mendelian traits, and reduces variability stemming from human error and researcher bias. MATK facilitates data reanalysis to keep up with the constantly improving annotation sources and next-generation sequencing processing pipelines. The software is open source and available at https://gitlab.com/matthew_maher/mendelanalysis.


Assuntos
Degeneração Retiniana , Automação , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Degeneração Retiniana/diagnóstico , Degeneração Retiniana/genética , Software , Sequenciamento do Exoma
4.
Nature ; 537(7618): 107-111, 2016 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-27556945

RESUMO

An early step in intracellular transport is the selective recognition of a vesicle by its appropriate target membrane, a process regulated by Rab GTPases via the recruitment of tethering effectors. Membrane tethering confers higher selectivity and efficiency to membrane fusion than the pairing of SNAREs (soluble N-ethylmaleimide-sensitive factor attachment protein receptors) alone. Here we address the mechanism whereby a tethered vesicle comes closer towards its target membrane for fusion by reconstituting an endosomal asymmetric tethering machinery consisting of the dimeric coiled-coil protein EEA1 (refs 6, 7) recruited to phosphatidylinositol 3-phosphate membranes and binding vesicles harbouring Rab5. Surprisingly, structural analysis reveals that Rab5:GTP induces an allosteric conformational change in EEA1, from extended to flexible and collapsed. Through dynamic analysis by optical tweezers, we confirm that EEA1 captures a vesicle at a distance corresponding to its extended conformation, and directly measure its flexibility and the forces induced during the tethering reaction. Expression of engineered EEA1 variants defective in the conformational change induce prominent clusters of tethered vesicles in vivo. Our results suggest a new mechanism in which Rab5 induces a change in flexibility of EEA1, generating an entropic collapse force that pulls the captured vesicle towards the target membrane to initiate docking and fusion.


Assuntos
Endossomos/metabolismo , Entropia , Fusão de Membrana , Proteínas de Transporte Vesicular/química , Proteínas de Transporte Vesicular/metabolismo , Proteínas rab5 de Ligação ao GTP/metabolismo , Regulação Alostérica , Guanosina Trifosfato/metabolismo , Humanos , Pinças Ópticas , Fosfatos de Fosfatidilinositol/metabolismo , Maleabilidade , Ligação Proteica , Conformação Proteica , Proteínas SNARE/metabolismo , Proteínas de Transporte Vesicular/genética
5.
EMBO Rep ; 20(7): e47055, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-31267706

RESUMO

Most cells acquire cholesterol by endocytosis of circulating low-density lipoproteins (LDLs). After cholesteryl ester de-esterification in endosomes, free cholesterol is redistributed to intracellular membranes via unclear mechanisms. Our previous work suggested that the unconventional phospholipid lysobisphosphatidic acid (LBPA) may play a role in modulating the cholesterol flux through endosomes. In this study, we used the Prestwick library of FDA-approved compounds in a high-content, image-based screen of the endosomal lipids, lysobisphosphatidic acid and LDL-derived cholesterol. We report that thioperamide maleate, an inverse agonist of the histamine H3 receptor HRH3, increases highly selectively the levels of lysobisphosphatidic acid, without affecting any endosomal protein or function that we tested. Our data also show that thioperamide significantly reduces the endosome cholesterol overload in fibroblasts from patients with the cholesterol storage disorder Niemann-Pick type C (NPC), as well as in liver of Npc1-/- mice. We conclude that LBPA controls endosomal cholesterol mobilization and export to cellular destinations, perhaps by fluidifying or buffering cholesterol in endosomal membranes, and that thioperamide has repurposing potential for the treatment of NPC.


Assuntos
Colesterol/metabolismo , Endossomos/efeitos dos fármacos , Lisofosfolipídeos/metabolismo , Monoglicerídeos/metabolismo , Doença de Niemann-Pick Tipo C/metabolismo , Piperidinas/farmacologia , Animais , Células Cultivadas , Endossomos/metabolismo , Feminino , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Células HeLa , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C
6.
Nature ; 526(7574): 564-8, 2015 Oct 22.
Artigo em Inglês | MEDLINE | ID: mdl-26444236

RESUMO

The human kidney contains up to 2 million epithelial nephrons responsible for blood filtration. Regenerating the kidney requires the induction of the more than 20 distinct cell types required for excretion and the regulation of pH, and electrolyte and fluid balance. We have previously described the simultaneous induction of progenitors for both collecting duct and nephrons via the directed differentiation of human pluripotent stem cells. Paradoxically, although both are of intermediate mesoderm in origin, collecting duct and nephrons have distinct temporospatial origins. Here we identify the developmental mechanism regulating the preferential induction of collecting duct versus kidney mesenchyme progenitors. Using this knowledge, we have generated kidney organoids that contain nephrons associated with a collecting duct network surrounded by renal interstitium and endothelial cells. Within these organoids, individual nephrons segment into distal and proximal tubules, early loops of Henle, and glomeruli containing podocytes elaborating foot processes and undergoing vascularization. When transcription profiles of kidney organoids were compared to human fetal tissues, they showed highest congruence with first trimester human kidney. Furthermore, the proximal tubules endocytose dextran and differentially apoptose in response to cisplatin, a nephrotoxicant. Such kidney organoids represent powerful models of the human organ for future applications, including nephrotoxicity screening, disease modelling and as a source of cells for therapy.


Assuntos
Linhagem da Célula , Células-Tronco Pluripotentes Induzidas/citologia , Modelos Biológicos , Néfrons/citologia , Néfrons/embriologia , Organogênese , Organoides/citologia , Animais , Técnicas de Cocultura , Células Alimentadoras , Feto/anatomia & histologia , Feto/citologia , Feto/embriologia , Fibroblastos/citologia , Humanos , Túbulos Renais Coletores/citologia , Túbulos Renais Proximais/citologia , Túbulos Renais Proximais/embriologia , Túbulos Renais Proximais/fisiologia , Mesoderma/citologia , Camundongos , Néfrons/anatomia & histologia , Néfrons/fisiologia , Organoides/embriologia , Técnicas de Cultura de Tecidos
7.
Traffic ; 19(2): 105-110, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29159991

RESUMO

Visualization of scientific data is crucial not only for scientific discovery but also to communicate science and medicine to both experts and a general audience. Until recently, we have been limited to visualizing the three-dimensional (3D) world of biology in 2 dimensions. Renderings of 3D cells are still traditionally displayed using two-dimensional (2D) media, such as on a computer screen or paper. However, the advent of consumer grade virtual reality (VR) headsets such as Oculus Rift and HTC Vive means it is now possible to visualize and interact with scientific data in a 3D virtual world. In addition, new microscopic methods provide an unprecedented opportunity to obtain new 3D data sets. In this perspective article, we highlight how we have used cutting edge imaging techniques to build a 3D virtual model of a cell from serial block-face scanning electron microscope (SBEM) imaging data. This model allows scientists, students and members of the public to explore and interact with a "real" cell. Early testing of this immersive environment indicates a significant improvement in students' understanding of cellular processes and points to a new future of learning and public engagement. In addition, we speculate that VR can become a new tool for researchers studying cellular architecture and processes by populating VR models with molecular data.


Assuntos
Células/ultraestrutura , Compreensão/fisiologia , Software , Análise e Desempenho de Tarefas , Realidade Virtual , Humanos , Imageamento Tridimensional , Interface Usuário-Computador
8.
J Cell Mol Med ; 24(6): 3724-3738, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-32065471

RESUMO

In solid tumours, elevated interstitial fluid pressure (osmotic and hydrostatic pressure) is a barrier to drug delivery and correlates with poor prognosis. Glioblastoma (GBM) further experience compressive force when growing within a space limited by the skull. Caveolae are proposed to play mechanosensing roles, and caveola-forming proteins are overexpressed in GBM. We asked whether caveolae mediate the GBM response to osmotic pressure. We evaluated in vitro the influence of spontaneous or experimental down-regulation of caveola-forming proteins (caveolin-1, CAVIN1) on the proteolytic profile and invasiveness of GBM cells in response to osmotic pressure. In response to osmotic pressure, GBM cell lines expressing caveola-forming proteins up-regulated plasminogen activator (uPA) and/or matrix metalloproteinases (MMPs), some EMT markers and increased their in vitro invasion potential. Down-regulation of caveola-forming proteins impaired this response and prevented hyperosmolarity-induced mRNA expression of the water channel aquaporin 1. CRISPR ablation of caveola-forming proteins further lowered expression of matrix proteases and EMT markers in response to hydrostatic pressure, as a model of mechanical force. GBM respond to pressure by increasing matrix-degrading enzyme production, mesenchymal phenotype and invasion. Caveola-forming proteins mediate, at least in part, the pro-invasive response of GBM to pressure. This may represent a novel target in GBM treatment.


Assuntos
Neoplasias Encefálicas/metabolismo , Cavéolas/metabolismo , Caveolina 1/metabolismo , Glioblastoma/metabolismo , Pressão Hidrostática , Osmose , Aquaporina 1/genética , Aquaporina 1/metabolismo , Neoplasias Encefálicas/patologia , Neoplasias Encefálicas/ultraestrutura , Cavéolas/ultraestrutura , Linhagem Celular Tumoral , Matriz Extracelular/metabolismo , Regulação Neoplásica da Expressão Gênica , Glioblastoma/patologia , Glioblastoma/ultraestrutura , Humanos , Invasividade Neoplásica
9.
EMBO Rep ; 19(9)2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-30021837

RESUMO

Caveolae are plasma membrane invaginations involved in transport, signalling and mechanical membrane sensing in metazoans. Their formation depends upon multiple interactions between membrane-embedded caveolins, lipids and cytosolic cavin proteins. Of the four cavin family members, only cavin1 is strictly required for caveola formation. Here, we demonstrate that an eleven residue (undecad) repeat sequence (UC1) exclusive to cavin1 is essential for caveolar localization and promotes membrane remodelling through binding to phosphatidylserine. In the notochord of mechanically stimulated zebrafish embryos, the UC1 domain is required for caveolar stability and resistance to membrane stress. The number of undecad repeats in the cavin1 UC1 domain varies throughout evolution, and we find that an increased number also correlates with increased caveolar stability. Lastly, we show that the cavin1 UC1 domain induces dramatic remodelling of the plasma membrane when grafted into cavin2 suggesting an important role in membrane sculpting. Overall, our work defines a novel conserved cavin1 modular domain that controls caveolar assembly and stability.


Assuntos
Cavéolas/metabolismo , Proteínas de Membrana/metabolismo , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/metabolismo , Proteínas de Peixe-Zebra/metabolismo , Animais , Proteínas de Transporte/metabolismo , Membrana Celular/metabolismo , Análise Mutacional de DNA , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Células MCF-7 , Proteínas de Membrana/química , Proteínas de Membrana/genética , Notocorda/metabolismo , Células PC-3 , Proteínas de Ligação a Fosfato , Proteínas de Ligação a RNA/química , Estresse Mecânico , Peixe-Zebra , Proteínas de Peixe-Zebra/química , Proteínas de Peixe-Zebra/genética
10.
Proc Natl Acad Sci U S A ; 114(40): E8372-E8381, 2017 10 03.
Artigo em Inglês | MEDLINE | ID: mdl-28916735

RESUMO

The mammalian heart undergoes maturation during postnatal life to meet the increased functional requirements of an adult. However, the key drivers of this process remain poorly defined. We are currently unable to recapitulate postnatal maturation in human pluripotent stem cell-derived cardiomyocytes (hPSC-CMs), limiting their potential as a model system to discover regenerative therapeutics. Here, we provide a summary of our studies, where we developed a 96-well device for functional screening in human pluripotent stem cell-derived cardiac organoids (hCOs). Through interrogation of >10,000 organoids, we systematically optimize parameters, including extracellular matrix (ECM), metabolic substrate, and growth factor conditions, that enhance cardiac tissue viability, function, and maturation. Under optimized maturation conditions, functional and molecular characterization revealed that a switch to fatty acid metabolism was a central driver of cardiac maturation. Under these conditions, hPSC-CMs were refractory to mitogenic stimuli, and we found that key proliferation pathways including ß-catenin and Yes-associated protein 1 (YAP1) were repressed. This proliferative barrier imposed by fatty acid metabolism in hCOs could be rescued by simultaneous activation of both ß-catenin and YAP1 using genetic approaches or a small molecule activating both pathways. These studies highlight that human organoids coupled with higher-throughput screening platforms have the potential to rapidly expand our knowledge of human biology and potentially unlock therapeutic strategies.


Assuntos
Fatores Biológicos/metabolismo , Pontos de Checagem do Ciclo Celular , Miócitos Cardíacos/metabolismo , Organoides/metabolismo , Células-Tronco Pluripotentes/metabolismo , Regeneração/fisiologia , Adulto , Animais , Diferenciação Celular , Dano ao DNA , Humanos , Masculino , Miócitos Cardíacos/citologia , Organoides/citologia , Células-Tronco Pluripotentes/citologia , Ratos Sprague-Dawley
11.
J Biol Chem ; 292(34): 14292-14307, 2017 08 25.
Artigo em Inglês | MEDLINE | ID: mdl-28698382

RESUMO

Caveolae are bulb-shaped nanodomains of the plasma membrane that are enriched in cholesterol and sphingolipids. They have many physiological functions, including endocytic transport, mechanosensing, and regulation of membrane and lipid transport. Caveola formation relies on integral membrane proteins termed caveolins (Cavs) and the cavin family of peripheral proteins. Both protein families bind anionic phospholipids, but the precise roles of these lipids are unknown. Here, we studied the effects of phosphatidylserine (PtdSer), phosphatidylinositol 4-phosphate (PtdIns4P), and phosphatidylinositol 4,5-bisphosphate (PtdIns(4,5)P2) on caveolar formation and dynamics. Using live-cell, single-particle tracking of GFP-labeled Cav1 and ultrastructural analyses, we compared the effect of PtdSer disruption or phosphoinositide depletion with caveola disassembly caused by cavin1 loss. We found that PtdSer plays a crucial role in both caveola formation and stability. Sequestration or depletion of PtdSer decreased the number of detectable Cav1-GFP puncta and the number of caveolae visualized by electron microscopy. Under PtdSer-limiting conditions, the co-localization of Cav1 and cavin1 was diminished, and cavin1 degradation was increased. Using rapamycin-recruitable phosphatases, we also found that the acute depletion of PtdIns4P and PtdIns(4,5)P2 has minimal impact on caveola assembly but results in decreased lateral confinement. Finally, we show in a model of phospholipid scrambling, a feature of apoptotic cells, that caveola stability is acutely affected by the scrambling. We conclude that the predominant plasmalemmal anionic lipid PtdSer is essential for proper Cav clustering, caveola formation, and caveola dynamics and that membrane scrambling can perturb caveolar stability.


Assuntos
Cavéolas/metabolismo , Caveolina 1/metabolismo , Membrana Celular/metabolismo , Modelos Biológicos , Fosfatidilserinas/metabolismo , Proteínas de Ligação a RNA/metabolismo , Animais , Cavéolas/química , Cavéolas/ultraestrutura , Caveolina 1/antagonistas & inibidores , Caveolina 1/química , Caveolina 1/genética , Linhagem Celular , Membrana Celular/química , Membrana Celular/ultraestrutura , Rastreamento de Células , Cricetulus , Humanos , Cinética , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Mesocricetus , Microscopia Eletrônica de Transmissão , Microscopia de Vídeo , Fosfatidilinositol 4,5-Difosfato/química , Fosfatidilinositol 4,5-Difosfato/metabolismo , Fosfatos de Fosfatidilinositol/química , Fosfatos de Fosfatidilinositol/metabolismo , Fosfatidilserinas/química , Transporte Proteico , Interferência de RNA , Proteínas de Ligação a RNA/antagonistas & inibidores , Proteínas de Ligação a RNA/química , Proteínas de Ligação a RNA/genética , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/metabolismo , Imagem com Lapso de Tempo
12.
Cell Microbiol ; 19(12)2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-28778116

RESUMO

Caveolae are composed of 2 major proteins, caveolin 1 (CAV1) and cavin 1 or polymerase transcript release factor I (CAVIN1). Here, we demonstrate that CAV1 levels modulate invasion of Group A Streptococcus (GAS) into nonphagocytic mammalian cells. GAS showed enhanced internalisation into CAV1-knockout mouse embryonic fibroblasts and CAV1 knockdown human epithelial HEp-2 cells, whereas overexpression of CAV1 in HEp-2 cells reduced GAS invasion. This effect was not dependent on the expression of the GAS fibronectin binding protein SfbI, which had previously been implicated in caveolae-mediated uptake. Nor was this effect dependent on CAVIN1, as knockout of CAVIN1 in mouse embryonic fibroblasts resulted in reduced GAS internalisation. Although CAV1 restricted GAS invasion into host cells, we observed only minimal association of invading GAS (strain M1T15448 ) with CAV1 by immunofluorescence and very low association of invading M1T15448 with caveolae by transmission electron microscopy. These observations suggest that physical interaction with caveolae is not needed for CAV1 restriction of invading GAS. An indirect mechanism of action is also consistent with the finding that changing membrane fluidity reverses the increased invasion observed in CAV1-null cells. Together, these results suggest that CAV1 protects host cells against GAS invasion by a caveola-independent mechanism.


Assuntos
Caveolina 1/metabolismo , Endocitose , Células Epiteliais/imunologia , Fibroblastos/imunologia , Fatores Imunológicos/metabolismo , Streptococcus pyogenes/imunologia , Animais , Linhagem Celular , Células Epiteliais/microbiologia , Fibroblastos/microbiologia , Humanos , Camundongos Knockout
13.
Basic Res Cardiol ; 112(3): 24, 2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-28343262

RESUMO

Caveolae and associated cavin and caveolins may govern myocardial function, together with responses to mechanical and ischaemic stresses. Abnormalities in these proteins are also implicated in different cardiovascular disorders. However, specific roles of the cavin-1 protein in cardiac and coronary responses to mechanical/metabolic perturbation remain unclear. We characterised cardiovascular impacts of cavin-1 deficiency, comparing myocardial and coronary phenotypes and responses to stretch and ischaemia-reperfusion in hearts from cavin-1 +/+ and cavin-1 -/- mice. Caveolae and caveolins 1 and 3 were depleted in cavin-1 -/- hearts. Cardiac ejection properties in situ were modestly reduced in cavin-1 -/- mice. While peak contractile performance in ex vivo myocardium from cavin-1 -/- and cavin-1 +/+ mice was comparable, intrinsic beating rate, diastolic stiffness and Frank-Starling behaviour (stretch-dependent diastolic and systolic forces) were exaggerated in cavin-1 -/- hearts. Increases in stretch-dependent forces were countered by NOS inhibition (100 µM L-NAME), which exposed negative inotropy in cavin-1 -/- hearts, and were mimicked by 100 µM nitroprusside. In contrast, chronotropic differences appeared largely NOS-independent. Cavin-1 deletion also induced NOS-dependent coronary dilatation, ≥3-fold prolongation of reactive hyperaemic responses, and exaggerated pressure-dependence of coronary flow. Stretch-dependent efflux of lactate dehydrogenase and cardiac troponin I was increased and induction of brain natriuretic peptide and c-Fos inhibited in cavin-1 -/- hearts, while ERK1/2 phospho-activation was preserved. Post-ischaemic dysfunction and damage was also exaggerated in cavin-1 -/- hearts. Diverse effects of cavin-1 deletion reveal important roles in both NOS-dependent and -independent control of cardiac and coronary functions, together with governing sarcolemmal fragility and myocardial responses to stretch and ischaemia.


Assuntos
Coração/fisiologia , Proteínas de Membrana/metabolismo , Traumatismo por Reperfusão Miocárdica/metabolismo , Miocárdio/metabolismo , Proteínas de Ligação a RNA/metabolismo , Animais , Western Blotting , Fenômenos Fisiológicos Cardiovasculares , Modelos Animais de Doenças , Preparação de Coração Isolado , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Contração Miocárdica/fisiologia , Traumatismo por Reperfusão Miocárdica/fisiopatologia , Óxido Nítrico Sintase/metabolismo , Reação em Cadeia da Polimerase , Estresse Mecânico
15.
J Biol Chem ; 290(41): 24875-90, 2015 Oct 09.
Artigo em Inglês | MEDLINE | ID: mdl-26304117

RESUMO

The generation of caveolae involves insertion of the cholesterol-binding integral membrane protein caveolin-1 (Cav1) into the membrane, however, the precise molecular mechanisms are as yet unknown. We have speculated that insertion of the caveolin scaffolding domain (CSD), a conserved amphipathic region implicated in interactions with signaling proteins, is crucial for caveola formation. We now define the core membrane-juxtaposed region of Cav1 and show that the oligomerization domain and CSD are protected by tight association with the membrane in both mature mammalian caveolae and a model prokaryotic system for caveola biogenesis. Cryoelectron tomography reveals the core membrane-juxtaposed domain to be sufficient to maintain oligomerization as defined by polyhedral distortion of the caveolar membrane. Through mutagenesis we demonstrate the importance of the membrane association of the oligomerization domain/CSD for defined caveola biogenesis and furthermore, highlight the functional significance of the intramembrane domain and the CSD for defined caveolin-induced membrane deformation. Finally, we define the core structural domain of Cav1, constituting only 66 amino acids and of great potential to nanoengineering applications, which is required for caveolin-induced vesicle formation in a bacterial system. These results have significant implications for understanding the role of Cav1 in caveola formation and in regulating cellular signaling events.


Assuntos
Caveolina 1/metabolismo , Membrana Celular/metabolismo , Sequência de Aminoácidos , Animais , Caveolina 1/química , Membrana Celular/química , Cães , Endopeptidase K/metabolismo , Células Madin Darby de Rim Canino , Dados de Sequência Molecular , Multimerização Proteica , Estrutura Terciária de Proteína , Proteólise
16.
Hum Mol Genet ; 22(24): 5026-35, 2013 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-23900075

RESUMO

Defects such as cleft lip with or without cleft palate (CL/P) are among the most common craniofacial birth defects in humans. In many cases, the underlying molecular and cellular mechanisms that result in these debilitating anomalies remain largely unknown. Perturbed hedgehog (HH) signalling plays a major role in craniofacial development, and mutations in a number of pathway constituents underlie craniofacial disease. In particular, mutations in the gene encoding the major HH receptor and negative regulator, patched1 (PTCH1), are associated with both sporadic and familial forms of clefting, yet relatively little is known about how PTCH1 functions during craniofacial morphogenesis. To address this, we analysed the consequences of conditional loss of Ptch1 in mouse neural crest cell-derived facial mesenchyme. Using scanning electron microscopy (SEM) and live imaging of explanted facial primordia, we captured defective nasal pit invagination and CL in mouse embryos conditionally lacking Ptch1. Our analysis demonstrates interactions between HH and FGF signalling in the development of the upper lip, and reveals cell-autonomous and non-autonomous roles mediated by Ptch1. In particular, we show that deletion of Ptch1 in the facial mesenchyme alters cell morphology, specifically in the invaginating nasal pit epithelium. These findings highlight a critical link between the neural crest cells and olfactory epithelium in directing the morphogenesis of the mammalian lip and nose primordia. Importantly, these interactions are critically dependent on Ptch1 function for the prevention of orofacial clefts.


Assuntos
Encéfalo/anormalidades , Fenda Labial/genética , Fissura Palatina/genética , Crista Neural/metabolismo , Receptores de Superfície Celular/genética , Animais , Encéfalo/metabolismo , Morte Celular/genética , Proliferação de Células , Forma Celular/genética , Fenda Labial/metabolismo , Fissura Palatina/metabolismo , Modelos Animais de Doenças , Células Epiteliais/metabolismo , Fatores de Crescimento de Fibroblastos/metabolismo , Estudos de Associação Genética , Proteínas Hedgehog/metabolismo , Mesoderma/embriologia , Mesoderma/metabolismo , Camundongos , Camundongos Knockout , Morfogênese/genética , Mucosa Nasal/metabolismo , Crista Neural/enzimologia , Nariz/embriologia , Receptores Patched , Receptor Patched-1 , Fenótipo , Receptores de Superfície Celular/metabolismo , Transdução de Sinais , Proteína Wnt1/genética , Proteína Wnt1/metabolismo
17.
PLoS Pathog ; 9(8): e1003513, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23935497

RESUMO

Hepatitis C virus (HCV) is a single-stranded RNA virus that replicates on endoplasmic reticulum-derived membranes. HCV particle assembly is dependent on the association of core protein with cellular lipid droplets (LDs). However, it remains uncertain whether HCV assembly occurs at the LD membrane itself or at closely associated ER membranes. Furthermore, it is not known how the HCV replication complex and progeny genomes physically associate with the presumed sites of virion assembly at or near LDs. Using an unbiased proteomic strategy, we have found that Rab18 interacts with the HCV nonstructural protein NS5A. Rab18 associates with LDs and is believed to promote physical interaction between LDs and ER membranes. Active (GTP-bound) forms of Rab18 bind more strongly to NS5A than a constitutively GDP-bound mutant. NS5A colocalizes with Rab18-positive LDs in HCV-infected cells, and Rab18 appears to promote the physical association of NS5A and other replicase components with LDs. Modulation of Rab18 affects genome replication and possibly also the production of infectious virions. Our results support a model in which specific interactions between viral and cellular proteins may promote the physical interaction between membranous HCV replication foci and lipid droplets.


Assuntos
Hepacivirus/fisiologia , Membranas Intracelulares/metabolismo , Lipídeos de Membrana/metabolismo , Proteínas não Estruturais Virais/metabolismo , Replicação Viral/fisiologia , Proteínas rab de Ligação ao GTP/metabolismo , Linhagem Celular Tumoral , Hepatite C/genética , Hepatite C/metabolismo , Humanos , Membranas Intracelulares/virologia , Lipídeos de Membrana/genética , Proteômica , Proteínas não Estruturais Virais/genética , Proteínas rab de Ligação ao GTP/genética
18.
Hum Mol Genet ; 21(8): 1808-23, 2012 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-22228095

RESUMO

Mutations in components of the intraflagellar transport (IFT) machinery required for assembly and function of the primary cilium cause a subset of human ciliopathies characterized primarily by skeletal dysplasia. Recently, mutations in the IFT-A gene IFT144 have been described in patients with Sensenbrenner and Jeune syndromes, which are associated with short ribs and limbs, polydactyly and craniofacial defects. Here, we describe an N-ethyl-N-nitrosourea-derived mouse mutant with a hypomorphic missense mutation in the Ift144 gene. The mutant twinkle-toes (Ift144(twt)) phenocopies a number of the skeletal and craniofacial anomalies seen in patients with human skeletal ciliopathies. Like other IFT-A mouse mutants, Ift144 mutant embryos display a generalized ligand-independent expansion of hedgehog (Hh) signalling, in spite of defective ciliogenesis and an attenuation of the ability of mutant cells to respond to upstream stimulation of the pathway. This enhanced Hh signalling is consistent with cleft palate and polydactyly phenotypes in the Ift144(twt) mutant, although extensive rib branching, fusion and truncation phenotypes correlate with defects in early somite patterning and may reflect contributions from multiple signalling pathways. Analysis of embryos harbouring a second allele of Ift144 which represents a functional null, revealed a dose-dependent effect on limb outgrowth consistent with the short-limb phenotypes characteristic of these ciliopathies. This allelic series of mouse mutants provides a unique opportunity to uncover the underlying mechanistic basis of this intriguing subset of ciliopathies.


Assuntos
Anormalidades Múltiplas/genética , Cílios , Anormalidades Craniofaciais/genética , Proteínas/genética , Anormalidades Múltiplas/embriologia , Anormalidades Múltiplas/metabolismo , Animais , Mapeamento Cromossômico , Cílios/fisiologia , Cílios/ultraestrutura , Anormalidades Craniofaciais/embriologia , Anormalidades Craniofaciais/metabolismo , Proteínas do Citoesqueleto , Embrião de Mamíferos , Fatores de Crescimento de Fibroblastos/metabolismo , Membro Anterior/anormalidades , Membro Anterior/metabolismo , Proteínas Hedgehog/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular , Camundongos , Mutagênese , Mutação de Sentido Incorreto , Fenótipo , Polidactilia/embriologia , Polidactilia/genética , Polidactilia/metabolismo , Proteínas/química , Costelas/anormalidades , Transdução de Sinais
19.
PLoS Genet ; 7(7): e1002201, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21829381

RESUMO

Lipid droplets (LDs) are important cellular organelles that govern the storage and turnover of lipids. Little is known about how the size of LDs is controlled, although LDs of diverse sizes have been observed in different tissues and under different (patho)physiological conditions. Recent studies have indicated that the size of LDs may influence adipogenesis, the rate of lipolysis and the oxidation of fatty acids. Here, a genome-wide screen identifies ten yeast mutants producing "supersized" LDs that are up to 50 times the volume of those in wild-type cells. The mutated genes include: FLD1, which encodes a homologue of mammalian seipin; five genes (CDS1, INO2, INO4, CHO2, and OPI3) that are known to regulate phospholipid metabolism; two genes (CKB1 and CKB2) encoding subunits of the casein kinase 2; and two genes (MRPS35 and RTC2) of unknown function. Biochemical and genetic analyses reveal that a common feature of these mutants is an increase in the level of cellular phosphatidic acid (PA). Results from in vivo and in vitro analyses indicate that PA may facilitate the coalescence of contacting LDs, resulting in the formation of "supersized" LDs. In summary, our results provide important insights into how the size of LDs is determined and identify novel gene products that regulate phospholipid metabolism.


Assuntos
Metabolismo dos Lipídeos/genética , Ácidos Fosfatídicos/metabolismo , Espaço Intracelular/metabolismo , Fusão de Membrana , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Mutação/genética , Organelas/genética , Organelas/metabolismo , Fosfatidiletanolaminas/metabolismo , Fosfolipídeos/metabolismo , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Triglicerídeos/metabolismo
20.
Hepatology ; 55(5): 1574-84, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22105343

RESUMO

UNLABELLED: Caveolin-1 (CAV1) is a structural protein of caveolae involved in lipid homeostasis and endocytosis. Using newly generated pure Balb/C CAV1 null ((Balb/C)CAV1-/-) mice, CAV1-/- mice from Jackson Laboratories ((JAX)CAV1-/-), and CAV1-/- mice developed in the Kurzchalia Laboratory ((K)CAV1-/-), we show that under physiological conditions CAV1 expression in mouse tissues is necessary to guarantee an efficient progression of liver regeneration and mouse survival after partial hepatectomy. Absence of CAV1 in mouse tissues is compensated by the development of a carbohydrate-dependent anabolic adaptation. These results were supported by extracellular flux analysis of cellular glycolytic metabolism in CAV1-knockdown AML12 hepatocytes, suggesting cell autonomous effects of CAV1 loss in hepatic glycolysis. Unlike in (K)CAV1-/- livers, in (JAX)CAV1-/- livers CAV1 deficiency is compensated by activation of anabolic metabolism (pentose phosphate pathway and lipogenesis) allowing liver regeneration. Administration of 2-deoxy-glucose in (JAX)CAV1-/- mice indicated that liver regeneration in (JAX)CAV1-/- mice is strictly dependent on hepatic carbohydrate metabolism. Moreover, with the exception of regenerating (JAX)CAV1-/- livers, expression of CAV1 in mice is required for efficient hepatic lipid storage during fasting, liver regeneration, and diet-induced steatosis in the three CAV1-/- mouse strains. Furthermore, under these conditions CAV1 accumulates in the lipid droplet fraction in wildtype mouse hepatocytes. CONCLUSION: Our data demonstrate that lack of CAV1 alters hepatocyte energy metabolism homeostasis under physiological and pathological conditions.


Assuntos
Caveolina 1/metabolismo , Metabolismo Energético/fisiologia , Glucose/metabolismo , Regeneração Hepática/fisiologia , Análise de Variância , Animais , Análise Química do Sangue , Proliferação de Células , Cromatografia em Camada Fina/métodos , Desoxiglucose/farmacologia , Modelos Animais de Doenças , Feminino , Hepatectomia , Hepatócitos/metabolismo , Hepatócitos/fisiologia , Homeostase , Metabolismo dos Lipídeos/fisiologia , Regeneração Hepática/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos BALB C , Microscopia Eletrônica , Distribuição Aleatória , Reação em Cadeia da Polimerase em Tempo Real/métodos , Sensibilidade e Especificidade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA