Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
1.
PLoS Biol ; 16(4): e2005473, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29621251

RESUMEN

We describe the development and application of a suite of modular tools for high-resolution detection of proteins and intracellular protein complexes by electron microscopy (EM). Conditionally stable GFP- and mCherry-binding nanobodies (termed csGBP and csChBP, respectively) are characterized using a cell-free expression and analysis system and subsequently fused to an ascorbate peroxidase (APEX) enzyme. Expression of these cassettes alongside fluorescently labelled proteins results in recruitment and stabilisation of APEX, whereas unbound APEX nanobodies are efficiently degraded by the proteasome. This greatly simplifies correlative analyses, enables detection of less-abundant proteins, and eliminates the need to balance expression levels between fluorescently labelled and APEX nanobody proteins. Furthermore, we demonstrate the application of this system to bimolecular complementation ('EM split-fluorescent protein'), for localisation of protein-protein interactions at the ultrastructural level.


Asunto(s)
Ascorbato Peroxidasas/genética , Células Epiteliales/ultraestructura , Microscopía Electrónica/métodos , Microscopía Fluorescente/métodos , Imagen Individual de Molécula/métodos , Anticuerpos de Dominio Único/química , Animales , Ascorbato Peroxidasas/metabolismo , Línea Celular , Sistema Libre de Células , Cricetulus , Células Epiteliales/metabolismo , Expresión Génica , Genes Reporteros , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Unión Proteica , Mapeo de Interacción de Proteínas , Estabilidad Proteica , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Anticuerpos de Dominio Único/biosíntesis , Anticuerpos de Dominio Único/genética , Proteína Fluorescente Roja
2.
Traffic ; 16(7): 691-711, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25783006

RESUMEN

Actin has an ill-defined role in the trafficking of GLUT4 glucose transporter vesicles to the plasma membrane (PM). We have identified novel actin filaments defined by the tropomyosin Tpm3.1 at glucose uptake sites in white adipose tissue (WAT) and skeletal muscle. In Tpm 3.1-overexpressing mice, insulin-stimulated glucose uptake was increased; while Tpm3.1-null mice they were more sensitive to the impact of high-fat diet on glucose uptake. Inhibition of Tpm3.1 function in 3T3-L1 adipocytes abrogates insulin-stimulated GLUT4 translocation and glucose uptake. In WAT, the amount of filamentous actin is determined by Tpm3.1 levels and is paralleled by changes in exocyst component (sec8) and Myo1c levels. In adipocytes, Tpm3.1 localizes with MyoIIA, but not Myo1c, and it inhibits Myo1c binding to actin. We propose that Tpm3.1 determines the amount of cortical actin that can engage MyoIIA and generate contractile force, and in parallel limits the interaction of Myo1c with actin filaments. The balance between these actin filament populations may determine the efficiency of movement and/or fusion of GLUT4 vesicles with the PM.


Asunto(s)
Citoesqueleto de Actina/metabolismo , Glucosa/metabolismo , Tropomiosina/metabolismo , Células 3T3 , Adipocitos/metabolismo , Animales , Transportador de Glucosa de Tipo 4/metabolismo , Humanos , Ratones , Ratones Endogámicos C57BL , Miosina Tipo I/metabolismo , Unión Proteica , Transporte de Proteínas , Tropomiosina/genética
3.
Basic Res Cardiol ; 112(3): 24, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28343262

RESUMEN

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.


Asunto(s)
Corazón/fisiología , Proteínas de la Membrana/metabolismo , Daño por Reperfusión Miocárdica/metabolismo , Miocardio/metabolismo , Proteínas de Unión al ARN/metabolismo , Animales , Western Blotting , Fenómenos Fisiológicos Cardiovasculares , Modelos Animales de Enfermedad , Preparación de Corazón Aislado , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Contracción Miocárdica/fisiología , Daño por Reperfusión Miocárdica/fisiopatología , Óxido Nítrico Sintasa/metabolismo , Reacción en Cadena de la Polimerasa , Estrés Mecánico
4.
J Pathol ; 229(5): 685-96, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23224993

RESUMEN

Crim1 is a transmembrane protein that regulates the bioavailability of growth factors such as VEGFA. Crim1(KST264)(/)(KST264) hypomorphic mice develop renal disease characterized by glomerular cysts and loss of endothelial integrity, progressing to peritubular and pericystic fibrosis. Peritubular capillary endothelial cells display morphological changes as well as detachment from the basement membrane. In this study, gene expression profiling of CD31(+) endothelial cells isolated from Crim1(KST264)(/)(KST264) kidneys showed up-regulation of transcripts associated with fibrosis (Col3a1, Loxl1), endothelial dysfunction (Abp1, Dcn, Lcn2), biomarkers of renal damage (Lcn2, Havcr1/Kim1) as well as evidence for a TGFß1/TNF-associated inflammatory process. To determine whether the aberrant endothelium may in part contribute to the fibrogenic process, Tie2Cre-DsRed lineage tracing was undertaken in Crim1(KST264/KST264) mice. Approximately 31% of de novo αSMA(+) myofibroblasts detected within the tubulointerstitium were Tie2(+) DsRed(+) . However, 5.3% were F4/80(+) DsRed(+) , indicating a small population of myofibroblasts of monocytic rather than endothelial origin. In contrast, only 12% of myofibroblasts located around glomerular cysts were Tie2(+) DsRed(+) , with 7.7% being monocyte-derived (F4/80(+) DsRed(+) ). Collectively, this model supports the involvement of endothelial cells/monocytes in fibrosis within the tubulointerstitium, but also the heterogeneity of the fibrotic process even within distinct regions of the same kidney.


Asunto(s)
Receptores de Proteínas Morfogenéticas Óseas/genética , Linaje de la Célula , Células Endoteliales/patología , Enfermedades Renales/patología , Riñón/patología , Monocitos/patología , Mutación , Miofibroblastos/patología , Animales , Biomarcadores/metabolismo , Receptores de Proteínas Morfogenéticas Óseas/metabolismo , Linaje de la Célula/genética , Células Endoteliales/metabolismo , Transición Epitelial-Mesenquimal , Fibrosis , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Redes Reguladoras de Genes , Marcadores Genéticos , Genotipo , Integrasas/genética , Riñón/irrigación sanguínea , Riñón/metabolismo , Enfermedades Renales/genética , Enfermedades Renales/metabolismo , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Ratones , Ratones Transgénicos , Monocitos/metabolismo , Miofibroblastos/metabolismo , Fenotipo , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , ARN Mensajero/metabolismo , Proteínas Tirosina Quinasas Receptoras/genética , Receptor TIE-2
6.
Hepatology ; 55(5): 1574-84, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22105343

RESUMEN

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.


Asunto(s)
Caveolina 1/metabolismo , Metabolismo Energético/fisiología , Glucosa/metabolismo , Regeneración Hepática/fisiología , Análisis de Varianza , Animales , Análisis Químico de la Sangre , Proliferación Celular , Cromatografía en Capa Delgada/métodos , Desoxiglucosa/farmacología , Modelos Animales de Enfermedad , Femenino , Hepatectomía , Hepatocitos/metabolismo , Hepatocitos/fisiología , Homeostasis , Metabolismo de los Lípidos/fisiología , Regeneración Hepática/efectos de los fármacos , Ratones , Ratones Endogámicos BALB C , Microscopía Electrónica , Distribución Aleatoria , Reacción en Cadena en Tiempo Real de la Polimerasa/métodos , Sensibilidad y Especificidad
7.
BMC Biochem ; 14: 10, 2013 Apr 08.
Artículo en Inglés | MEDLINE | ID: mdl-23566155

RESUMEN

BACKGROUND: The androgen receptor (AR) is a member of the nuclear receptor (NR) superfamily of ligand-inducible DNA transcription factors, and is the major mediator of male sexual development, prostate growth and the pathogenesis of prostate cancer. Cell and gene specific regulation by the AR is determined by availability of and interaction with sets of key accessory cofactors. Ski-interacting protein (SKIP; SNW1, NCOA62) is a cofactor shown to interact with several NRs and a diverse range of other transcription factors. Interestingly, SKIP as part of the spliceosome is thought to link mRNA splicing with transcription. SKIP has not been previously shown to interact with the AR. RESULTS: The aim of this study was to investigate whether SKIP interacts with the AR and modulates AR-dependent transcription. Here, we show by co-immunoprecipitation experiments that SKIP is in a complex with the AR. Moreover, SKIP increased 5α-dihydrotestosterone (DHT) induced N-terminal/C-terminal AR interaction from 12-fold to almost 300-fold in a two-hybrid assay, and enhanced AR ligand-independent AF-1 transactivation. SKIP augmented ligand- and AR-dependent transactivation in PC3 prostate cancer cells. Live-cell imaging revealed a fast (half-time=129 s) translocation of AR from the cytoplasm to the nucleus upon DHT-stimulation. Förster resonance energy transfer (FRET) experiments suggest a direct AR-SKIP interaction in the nucleus upon translocation. CONCLUSIONS: Our results suggest that SKIP interacts with AR in the nucleus and enhances AR-dependent transactivation and N/C-interaction supporting a role for SKIP as an AR co-factor.


Asunto(s)
Coactivadores de Receptor Nuclear/metabolismo , Receptores Androgénicos/metabolismo , Animales , Células COS , Línea Celular Tumoral , Núcleo Celular/metabolismo , Chlorocebus aethiops , Dihidrotestosterona/farmacología , Transferencia Resonante de Energía de Fluorescencia , Genes Reporteros , Células HEK293 , Humanos , Inmunoprecipitación , Masculino , Coactivadores de Receptor Nuclear/genética , Unión Proteica/efectos de los fármacos , Estructura Terciaria de Proteína , Receptores Androgénicos/química , Receptores Androgénicos/genética , Activación Transcripcional
8.
Curr Biol ; 33(19): 4276-4284.e4, 2023 10 09.
Artículo en Inglés | MEDLINE | ID: mdl-37729911

RESUMEN

Plasma membrane rupture can result in catastrophic cell death. The skeletal muscle fiber plasma membrane, the sarcolemma, provides an extreme example of a membrane subject to mechanical stress since these cells specifically evolved to generate contraction and movement. A quantitative model correlating ultrastructural remodeling of surface architecture with tissue changes in vivo is required to understand how membrane domains contribute to the shape changes associated with tissue deformation in whole animals. We and others have shown that loss of caveolae, small invaginations of the plasma membrane particularly prevalent in the muscle sarcolemma, renders the plasma membrane more susceptible to rupture during stretch.1,2,3 While it is thought that caveolae are able to flatten and be absorbed into the bulk membrane to buffer local membrane expansion, a direct demonstration of this model in vivo has been unachievable since it would require measurement of caveolae at the nanoscale combined with detailed whole-animal morphometrics under conditions of perturbation. Here, we describe the development and application of the "active trapping model" where embryonic zebrafish are immobilized in a curved state that mimics natural body axis curvature during an escape response. The model is amenable to multiscale, multimodal imaging including high-resolution whole-animal three-dimensional quantitative electron microscopy. Using the active trapping model, we demonstrate the essential role of caveolae in maintaining sarcolemmal integrity and quantify the specific contribution of caveolar-derived membrane to surface expansion. We show that caveolae directly contribute to an increase in plasma membrane surface area under physiologically relevant membrane deformation conditions.


Asunto(s)
Caveolas , Pez Cebra , Animales , Membrana Celular , Caveolas/metabolismo , Fibras Musculares Esqueléticas , Microscopía Electrónica
9.
Dev Cell ; 58(5): 376-397.e4, 2023 03 13.
Artículo en Inglés | MEDLINE | ID: mdl-36858041

RESUMEN

Caveolae have been linked to many biological functions, but their precise roles are unclear. Using quantitative whole-cell proteomics of genome-edited cells, we show that the oxidative stress response is the major pathway dysregulated in cells lacking the key caveola structural protein, CAVIN1. CAVIN1 deletion compromised sensitivity to oxidative stress in cultured cells and in animals. Wound-induced accumulation of reactive oxygen species and apoptosis were suppressed in Cavin1-null zebrafish, negatively affecting regeneration. Oxidative stress triggered lipid peroxidation and induced caveolar disassembly. The resulting release of CAVIN1 from caveolae allowed direct interaction between CAVIN1 and NRF2, a key regulator of the antioxidant response, facilitating NRF2 degradation. CAVIN1-null cells with impaired negative regulation of NRF2 showed resistance to lipid-peroxidation-induced ferroptosis. Thus, caveolae, via lipid peroxidation and CAVIN1 release, maintain cellular susceptibility to oxidative-stress-induced cell death, demonstrating a crucial role for this organelle in cellular homeostasis and wound response.


Asunto(s)
Caveolas , Factor 2 Relacionado con NF-E2 , Animales , Caveolas/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , Pez Cebra/metabolismo , Peroxidación de Lípido , Proteínas de Unión al ARN/metabolismo , Estrés Oxidativo
10.
Elife ; 102021 02 16.
Artículo en Inglés | MEDLINE | ID: mdl-33591275

RESUMEN

Protein interaction networks are crucial for complex cellular processes. However, the elucidation of protein interactions occurring within highly specialised cells and tissues is challenging. Here, we describe the development, and application, of a new method for proximity-dependent biotin labelling in whole zebrafish. Using a conditionally stabilised GFP-binding nanobody to target a biotin ligase to GFP-labelled proteins of interest, we show tissue-specific proteomic profiling using existing GFP-tagged transgenic zebrafish lines. We demonstrate the applicability of this approach, termed BLITZ (Biotin Labelling In Tagged Zebrafish), in diverse cell types such as neurons and vascular endothelial cells. We applied this methodology to identify interactors of caveolar coat protein, cavins, in skeletal muscle. Using this system, we defined specific interaction networks within in vivo muscle cells for the closely related but functionally distinct Cavin4 and Cavin1 proteins.


Asunto(s)
Biotina/farmacología , Proteómica/métodos , Coloración y Etiquetado/métodos , Animales , Animales Modificados Genéticamente , Biotinilación , Caveolinas/metabolismo , Células Endoteliales/metabolismo , Proteínas Fluorescentes Verdes , Proteínas de la Membrana/metabolismo , Músculo Esquelético/metabolismo , Nanopartículas , Neuronas/metabolismo , Mapeo de Interacción de Proteínas , Pez Cebra
11.
Elife ; 102021 06 18.
Artículo en Inglés | MEDLINE | ID: mdl-34142659

RESUMEN

Caveolae-associated protein 3 (cavin3) is inactivated in most cancers. We characterized how cavin3 affects the cellular proteome using genome-edited cells together with label-free quantitative proteomics. These studies revealed a prominent role for cavin3 in DNA repair, with BRCA1 and BRCA1 A-complex components being downregulated on cavin3 deletion. Cellular and cell-free expression assays revealed a direct interaction between BRCA1 and cavin3 that occurs when cavin3 is released from caveolae that are disassembled in response to UV and mechanical stress. Overexpression and RNAi-depletion revealed that cavin3 sensitized various cancer cells to UV-induced apoptosis. Supporting a role in DNA repair, cavin3-deficient cells were sensitive to PARP inhibition, where concomitant depletion of 53BP1 restored BRCA1-dependent sensitivity to PARP inhibition. We conclude that cavin3 functions together with BRCA1 in multiple cancer-related pathways. The loss of cavin3 function may provide tumor cell survival by attenuating apoptotic sensitivity and hindering DNA repair under chronic stress conditions.


When cells become cancerous they often stop making certain proteins. This includes a protein known as cavin3 which resides in bulb-shaped pits of the membrane that surrounds the cell called caveolae. These structures work like stress detectors, picking up changes in the membrane and releasing proteins, such as cavin3, into the cell's interior. Past studies suggest that cavin3 might interact with a protein called BRCA1 that suppresses the formation of tumors. Cells with mutations in the gene for BRCA1 struggle to fix damage in their DNA, and have to rely on other repair proteins, such as PARPs (short for poly (ADP-ribose) polymerases). Blocking PARP proteins with drugs can kill cancer cells with problems in their BRCA1 proteins. However, it was unclear what role cavin3 plays in this mechanism. To investigate this, McMahon et al. exposed cells grown in the laboratory to DNA-damaging UV light to stimulate the release of cavin3 from caveolae. This revealed that cavin3 interacts with BRCA1 when cells are under stress, and helps stabilize the protein so it can perform DNA repairs. Cells without cavin3 showed decreased levels of the BRCA1 protein, but compensated for the loss of BRCA1 by increasing the levels of their PARP proteins. These cells also had increased DNA damage following treatment with drugs that block PARPs, similar to cancer cells carrying mutations in the gene for BRCA1. These findings suggest that cavin3 helps BRCA1 to suppress the formation of tumors, and therefore should be considered when developing new anti-cancer treatments.


Asunto(s)
Proteína BRCA1/metabolismo , Caveolas/metabolismo , Péptidos y Proteínas de Señalización Intracelular , Estrés Fisiológico/genética , Apoptosis/genética , Células HeLa , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteoma/genética , Proteómica
12.
Elife ; 102021 04 27.
Artículo en Inglés | MEDLINE | ID: mdl-33904409

RESUMEN

Genetic tags allow rapid localization of tagged proteins in cells and tissues. APEX, an ascorbate peroxidase, has proven to be one of the most versatile and robust genetic tags for ultrastructural localization by electron microscopy (EM). Here, we describe a simple method, APEX-Gold, which converts the diffuse oxidized diaminobenzidine reaction product of APEX into a silver/gold particle akin to that used for immunogold labelling. The method increases the signal-to-noise ratio for EM detection, providing unambiguous detection of the tagged protein, and creates a readily quantifiable particulate signal. We demonstrate the wide applicability of this method for detection of membrane proteins, cytoplasmic proteins, and cytoskeletal proteins. The method can be combined with different EM techniques including fast freezing and freeze substitution, focussed ion beam scanning EM, and electron tomography. Quantitation of expressed APEX-fusion proteins is achievable using membrane vesicles generated by a cell-free expression system. These membrane vesicles possess a defined quantum of signal, which can act as an internal standard for determination of the absolute density of expressed APEX-fusion proteins. Detection of fusion proteins expressed at low levels in cells from CRISPR-edited mice demonstrates the high sensitivity of the APEX-Gold method.


Asunto(s)
Tomografía con Microscopio Electrónico/métodos , Técnicas Genéticas , Imagenología Tridimensional/métodos , Animales , Ascorbato Peroxidasas , Congelación , Oro , Ratones , Proteínas
13.
J Cell Biol ; 220(12)2021 12 06.
Artículo en Inglés | MEDLINE | ID: mdl-34633413

RESUMEN

The cavin proteins are essential for caveola biogenesis and function. Here, we identify a role for the muscle-specific component, Cavin4, in skeletal muscle T-tubule development by analyzing two vertebrate systems, mouse and zebrafish. In both models, Cavin4 localized to T-tubules, and loss of Cavin4 resulted in aberrant T-tubule maturation. In zebrafish, which possess duplicated cavin4 paralogs, Cavin4b was shown to directly interact with the T-tubule-associated BAR domain protein Bin1. Loss of both Cavin4a and Cavin4b caused aberrant accumulation of interconnected caveolae within the T-tubules, a fragmented T-tubule network enriched in Caveolin-3, and an impaired Ca2+ response upon mechanical stimulation. We propose a role for Cavin4 in remodeling the T-tubule membrane early in development by recycling caveolar components from the T-tubule to the sarcolemma. This generates a stable T-tubule domain lacking caveolae that is essential for T-tubule function.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas Musculares/metabolismo , Músculo Esquelético/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Sarcolema/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Proteínas de Pez Cebra/metabolismo , Pez Cebra/metabolismo , Animales , Caveolas/metabolismo , Línea Celular , Embrión no Mamífero/metabolismo , Imagenología Tridimensional , Ratones Endogámicos C57BL , Fibras Musculares Esqueléticas/metabolismo , Fibras Musculares Esqueléticas/ultraestructura , Músculo Esquelético/ultraestructura , Unión Proteica , Sarcolema/ultraestructura , Pez Cebra/embriología
14.
Nat Commun ; 11(1): 3711, 2020 07 24.
Artículo en Inglés | MEDLINE | ID: mdl-32709891

RESUMEN

The skeletal muscle T-tubule is a specialized membrane domain essential for coordinated muscle contraction. However, in the absence of genetically tractable systems the mechanisms involved in T-tubule formation are unknown. Here, we use the optically transparent and genetically tractable zebrafish system to probe T-tubule development in vivo. By combining live imaging of transgenic markers with three-dimensional electron microscopy, we derive a four-dimensional quantitative model for T-tubule formation. To elucidate the mechanisms involved in T-tubule formation in vivo, we develop a quantitative screen for proteins that associate with and modulate early T-tubule formation, including an overexpression screen of the entire zebrafish Rab protein family. We propose an endocytic capture model involving firstly, formation of dynamic endocytic tubules at transient nucleation sites on the sarcolemma, secondly, stabilization by myofibrils/sarcoplasmic reticulum and finally, delivery of membrane from the recycling endosome and Golgi complex.


Asunto(s)
Contracción Muscular/fisiología , Proteínas Musculares/metabolismo , Músculo Esquelético/metabolismo , Músculo Esquelético/ultraestructura , Sarcolema/fisiología , Sarcolema/ultraestructura , Animales , Canales de Calcio/metabolismo , Canales de Calcio/ultraestructura , Canales de Calcio Tipo L/metabolismo , Proteínas Portadoras/metabolismo , Biología Evolutiva , Aparato de Golgi/metabolismo , Masculino , Microscopía Electrónica , Proteínas Musculares/química , Músculo Esquelético/química , Miofibrillas/metabolismo , Sarcolema/química , Retículo Sarcoplasmático/metabolismo , Pez Cebra
15.
Science ; 370(6514)2020 10 16.
Artículo en Inglés | MEDLINE | ID: mdl-33060333

RESUMEN

Lipid droplets (LDs) are the major lipid storage organelles of eukaryotic cells and a source of nutrients for intracellular pathogens. We demonstrate that mammalian LDs are endowed with a protein-mediated antimicrobial capacity, which is up-regulated by danger signals. In response to lipopolysaccharide (LPS), multiple host defense proteins, including interferon-inducible guanosine triphosphatases and the antimicrobial cathelicidin, assemble into complex clusters on LDs. LPS additionally promotes the physical and functional uncoupling of LDs from mitochondria, reducing fatty acid metabolism while increasing LD-bacterial contacts. Thus, LDs actively participate in mammalian innate immunity at two levels: They are both cell-autonomous organelles that organize and use immune proteins to kill intracellular pathogens as well as central players in the local and systemic metabolic adaptation to infection.


Asunto(s)
Bacterias/inmunología , Interacciones Huésped-Patógeno/inmunología , Inmunidad Innata , Gotas Lipídicas/inmunología , Animales , Péptidos Catiónicos Antimicrobianos/metabolismo , Ácidos Grasos/metabolismo , GTP Fosfohidrolasas/metabolismo , Células HEK293 , Humanos , Lipopolisacáridos/inmunología , Macrófagos/inmunología , Macrófagos/microbiología , Masculino , Ratones , Ratones Endogámicos C57BL , Mitocondrias/inmunología , Catelicidinas
16.
PLoS Pathog ; 3(6): e92, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17604452

RESUMEN

Candida albicans is a prevalent fungal pathogen amongst the immunocompromised population, causing both superficial and life-threatening infections. Since C. albicans is diploid, classical transmission genetics can not be performed to study specific aspects of its biology and pathogenesis. Here, we exploit the diploid status of C. albicans by constructing a library of 2,868 heterozygous deletion mutants and screening this collection using 35 known or novel compounds to survey chemically induced haploinsufficiency in the pathogen. In this reverse genetic assay termed the fitness test, genes related to the mechanism of action of the probe compounds are clearly identified, supporting their functional roles and genetic interactions. In this report, chemical-genetic relationships are provided for multiple FDA-approved antifungal drugs (fluconazole, voriconazole, caspofungin, 5-fluorocytosine, and amphotericin B) as well as additional compounds targeting ergosterol, fatty acid and sphingolipid biosynthesis, microtubules, actin, secretion, rRNA processing, translation, glycosylation, and protein folding mechanisms. We also demonstrate how chemically induced haploinsufficiency profiles can be used to identify the mechanism of action of novel antifungal agents, thereby illustrating the potential utility of this approach to antifungal drug discovery.


Asunto(s)
Antifúngicos/farmacología , Candida albicans/efectos de los fármacos , Diseño de Fármacos , Regulación Fúngica de la Expresión Génica/efectos de los fármacos , Genoma Fúngico , Candida albicans/genética , Candida albicans/metabolismo , ADN de Hongos/análisis , Perfilación de la Expresión Génica , Pruebas de Sensibilidad Microbiana , Análisis de Secuencia por Matrices de Oligonucleótidos , ARN Mensajero/efectos de los fármacos , ARN Mensajero/metabolismo
17.
Chem Biol ; 15(4): 363-74, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18420143

RESUMEN

Natural products provide an unparalleled source of chemical scaffolds with diverse biological activities and have profoundly impacted antimicrobial drug discovery. To further explore the full potential of their chemical diversity, we survey natural products for antifungal, target-specific inhibitors by using a chemical-genetic approach adapted to the human fungal pathogen Candida albicans and demonstrate that natural-product fermentation extracts can be mechanistically annotated according to heterozygote strain responses. Applying this approach, we report the discovery and characterization of a natural product, parnafungin, which we demonstrate, by both biochemical and genetic means, to inhibit poly(A) polymerase. Parnafungin displays potent and broad spectrum activity against diverse, clinically relevant fungal pathogens and reduces fungal burden in a murine model of disseminated candidiasis. Thus, mechanism-of-action determination of crude fermentation extracts by chemical-genetic profiling brings a powerful strategy to natural-product-based drug discovery.


Asunto(s)
Antifúngicos/farmacología , Antifúngicos/uso terapéutico , Productos Biológicos/farmacología , Productos Biológicos/uso terapéutico , Candida albicans/efectos de los fármacos , Candida albicans/genética , Evaluación Preclínica de Medicamentos/métodos , Polinucleotido Adenililtransferasa/antagonistas & inhibidores , Alelos , Secuencia de Aminoácidos , Animales , Antifúngicos/química , Antifúngicos/aislamiento & purificación , Aspergillus fumigatus/efectos de los fármacos , Aspergillus fumigatus/crecimiento & desarrollo , Aspergillus fumigatus/metabolismo , Productos Biológicos/química , Productos Biológicos/aislamiento & purificación , Candida albicans/metabolismo , Candidiasis/tratamiento farmacológico , Candidiasis/metabolismo , Mezclas Complejas/farmacología , Desoxiadenosinas/metabolismo , Desoxiadenosinas/farmacología , Farmacorresistencia Fúngica , Fermentación , Heterocigoto , Ratones , Pruebas de Sensibilidad Microbiana , Datos de Secuencia Molecular , Mutación , Poliadenilación/efectos de los fármacos , Polinucleotido Adenililtransferasa/genética , Polinucleotido Adenililtransferasa/metabolismo , ARN Mensajero/metabolismo , Saccharomyces cerevisiae/efectos de los fármacos , Saccharomyces cerevisiae/metabolismo , Resultado del Tratamiento
18.
Nat Commun ; 10(1): 3279, 2019 07 22.
Artículo en Inglés | MEDLINE | ID: mdl-31332168

RESUMEN

Caveolae are specialized domains of the plasma membrane. Formation of these invaginations is dependent on the expression of Caveolin-1 or -3 and proteins of the cavin family. In response to stress, caveolae disassemble and cavins are released from caveolae, allowing cavins to potentially interact with intracellular targets. Here, we describe the intracellular (non-plasma membrane) cavin interactome using biotin affinity proteomics and mass spectrometry. We validate 47 potential cavin-interactor proteins using a cell-free expression system and protein-protein binding assays. These data, together with pathway analyses, reveal unknown roles for cavin proteins in metabolism and stress signaling. We validated the interaction between one candidate interactor protein, protein phosphatase 1 alpha (PP1α), and Cavin-1 and -3 and show that UV treatment causes release of Cavin3 from caveolae allowing interaction with, and inhibition of, PP1α. This interaction increases H2AX phosphorylation to stimulate apoptosis, identifying a pro-apoptotic signaling pathway from surface caveolae to the nucleus.


Asunto(s)
Apoptosis/fisiología , Caveolas/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteína Fosfatasa 1/metabolismo , Proteínas de Unión al ARN/metabolismo , Apoptosis/efectos de la radiación , Caveolas/efectos de la radiación , Núcleo Celular/metabolismo , Histonas/metabolismo , Humanos , Espectrometría de Masas/métodos , Fosforilación/efectos de la radiación , Unión Proteica/efectos de la radiación , Transporte de Proteínas/efectos de la radiación , Proteómica/métodos , Rayos Ultravioleta
19.
Chem Biol ; 14(10): 1163-75, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17961828

RESUMEN

Mechanism-of-action (MOA) studies of bioactive compounds are fundamental to drug discovery. However, in vitro studies alone may not recapitulate a compound's MOA in whole cells. Here, we apply a chemogenomics approach in Candida albicans to evaluate compounds affecting purine metabolism. They include the IMP dehydrogenase inhibitors mycophenolic acid and mizoribine and the previously reported GMP synthase inhibitors acivicin and 6-diazo-5-oxo-L-norleucine (DON). We report important aspects of their whole-cell activity, including their primary target, off-target activity, and drug metabolism. Further, we describe ECC1385, an inhibitor of GMP synthase, and provide biochemical and genetic evidence supporting its MOA to be distinct from acivicin or DON. Importantly, GMP synthase activity is conditionally essential in C. albicans and Aspergillus fumigatus and is required for virulence of both pathogens, thus constituting an unexpected antifungal target.


Asunto(s)
Antifúngicos/farmacología , Aspergillus fumigatus/efectos de los fármacos , Candida albicans/efectos de los fármacos , Ligasas de Carbono-Nitrógeno/antagonistas & inhibidores , Inhibidores Enzimáticos/farmacología , Aspergillus fumigatus/enzimología , Candida albicans/enzimología , Diazooxonorleucina/farmacología , Farmacorresistencia Fúngica , Electroforesis en Gel de Poliacrilamida , IMP Deshidrogenasa/antagonistas & inhibidores , Isoxazoles/farmacología , Pruebas de Sensibilidad Microbiana , Ácido Micofenólico/farmacología , Purinas/metabolismo , Ribonucleósidos/farmacología
20.
Cell Death Discov ; 4: 19, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-29531816

RESUMEN

The Tasmanian devil faces extinction due to devil facial tumour disease (DFTD), a highly transmittable clonal form of cancer without available treatment. In this study, we report the cell-autonomous antiproliferative and cytotoxic activities exhibited by the spider peptide gomesin (AgGom) and gomesin-like homologue (HiGom) in DFTD cells. Mechanistically, both peptides caused a significant reduction at G0/G1 phase, in correlation with an augmented expression of the cell cycle inhibitory proteins p53, p27, p21, necrosis, exacerbated generation of reactive oxygen species and diminished mitochondrial membrane potential, all hallmarks of cellular stress. The screening of a novel panel of AgGom-analogues revealed that, unlike changes in the hydrophobicity and electrostatic surface, the cytotoxic potential of the gomesin analogues in DFTD cells lies on specific arginine substitutions in the eight and nine positions and alanine replacement in three, five and 12 positions. In conclusion, the evidence supports gomesin as a potential antiproliferative compound against DFTD disease.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA