Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Más filtros










Intervalo de año de publicación
1.
J Clin Invest ; 133(17)2023 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-37655664

RESUMEN

The lymphatic system (LS) is composed of lymphoid organs and a network of vessels that transport interstitial fluid, antigens, lipids, cholesterol, immune cells, and other materials in the body. Abnormal development or malfunction of the LS has been shown to play a key role in the pathophysiology of many disease states. Thus, improved understanding of the anatomical and molecular characteristics of the LS may provide approaches for disease prevention or treatment. Recent advances harnessing single-cell technologies, clinical imaging, discovery of biomarkers, and computational tools have led to the development of strategies to study the LS. This Review summarizes the outcomes of the NIH workshop entitled "Yet to be Charted: Lymphatic System in Health and Disease," held in September 2022, with emphasis on major areas for advancement. International experts showcased the current state of knowledge regarding the LS and highlighted remaining challenges and opportunities to advance the field.


Asunto(s)
Sistema Linfático , Vasos Linfáticos
2.
Front Physiol ; 14: 1099403, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36814475

RESUMEN

Enhancing our understanding of lymphatic anatomy from the microscopic to the anatomical scale is essential to discern how the structure and function of the lymphatic system interacts with different tissues and organs within the body and contributes to health and disease. The knowledge of molecular aspects of the lymphatic network is fundamental to understand the mechanisms of disease progression and prevention. Recent advances in mapping components of the lymphatic system using state of the art single cell technologies, the identification of novel biomarkers, new clinical imaging efforts, and computational tools which attempt to identify connections between these diverse technologies hold the potential to catalyze new strategies to address lymphatic diseases such as lymphedema and lipedema. This manuscript summarizes current knowledge of the lymphatic system and identifies prevailing challenges and opportunities to advance the field of lymphatic research as discussed by the experts in the workshop.

5.
JCI Insight ; 52019 03 14.
Artículo en Inglés | MEDLINE | ID: mdl-30870143

RESUMEN

Because injured mitochondria can accelerate cell death through the elaboration of oxidative free radicals and other mediators, it is striking that proliferator gamma coactivator 1-alpha (PGC1α), a stimulator of increased mitochondrial abundance, protects stressed renal cells instead of potentiating injury. Here we report that PGC1α's induction of lysosomes via transcription factor EB (TFEB) may be pivotal for kidney protection. CRISPR and stable gene transfer showed that PGC1α knockout tubular cells were sensitized to the genotoxic stressor cisplatin whereas transgenic cells were protected. The biosensor mtKeima unexpectedly revealed that cisplatin blunts mitophagy both in cells and mice. PGC1α not only counteracted this effect but also raised basal mitophagy, as did the downstream mediator nicotinamide adenine dinucleotide (NAD+). PGC1α did not consistently affect known autophagy pathways modulated by cisplatin. Instead RNA sequencing identified coordinated regulation of lysosomal biogenesis via TFEB. This effector pathway was sufficiently important that inhibition of TFEB or lysosomes unveiled a striking harmful effect of excess PGC1α in cells and conditional mice. These results uncover an unexpected effect of cisplatin on mitophagy and PGC1α's exquisite reliance on lysosomes for kidney protection. Finally, the data illuminate TFEB as a novel target for renal tubular stress resistance.


Asunto(s)
Lesión Renal Aguda/genética , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/metabolismo , Cisplatino/toxicidad , Túbulos Renales/metabolismo , Lisosomas/metabolismo , Mitocondrias/metabolismo , Biogénesis de Organelos , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/genética , Lesión Renal Aguda/inducido químicamente , Lesión Renal Aguda/metabolismo , Animales , Autofagia/efectos de los fármacos , Autofagia/genética , Sistemas CRISPR-Cas , Técnicas de Transferencia de Gen , Túbulos Renales/citología , Ratones , Ratones Noqueados , Ratones Transgénicos , Mitofagia/genética , NAD/metabolismo , Estrés Oxidativo/genética , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/metabolismo , Análisis de Secuencia de ARN
6.
J Mol Cell Cardiol ; 127: 270-276, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30639412

RESUMEN

Atherosclerosis is a chronic disorder of the vessel wall. One key regulator of disease progression is lipid handling in macrophages. However, the role of macrophage mitochondrial-dependent fatty acid ß-oxidation (FAO) in atherosclerosis is not well defined. To address this, we focused on carnitine palmitoyltransferase (CPT) 1 and 2, which play an essential role in the transport of long chain fatty acids (FAs) into the mitochondria. Using conditional alleles of these mitochondrial enzymes, we have generated myeloid-specific Cpt1a and Cpt2 knockout mutants (CPT1a M-KO and CPT2 M-KO). In culture, macrophages derived from CPT1a and CPT2 M-KO mice have impaired FAO, enhanced expression of the CD36 scavenger receptor, increased uptake of oxidized low-density lipoprotein (oxLDL), and augmented transformation into cholesterol-rich foam cells. In line with these in vitro observations, in the atherosclerosis-susceptible apolipoprotein E (ApoE) KO background, CPT2 M-KO mice demonstrated augmented atherosclerosis, accompanied by increased accumulation of aortic macrophages with elevated CD36 expression. These data suggest that macrophage FAO is athero-protective and that augmenting FAO may potentially slow atherosclerotic progression.


Asunto(s)
Aterosclerosis/metabolismo , Aterosclerosis/patología , Progresión de la Enfermedad , Ácidos Grasos/metabolismo , Macrófagos/metabolismo , Animales , Ratones , Ratones Noqueados , Células Mieloides/metabolismo , Oxidación-Reducción
7.
J Mol Cell Cardiol ; 123: 180-184, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30236923

RESUMEN

Certain organisms, including zebrafish, are capable of complete cardiac regeneration in response to injury. This response has also been observed in newborn mice, although in this case, the regenerative capacity is lost at approximately one week of age. The mechanisms regulating this short temporal window of cardiac regeneration in mice are not well understood. Here, we show that sonic hedgehog (Shh) signaling modulates the neonatal mouse regenerative response. In particular, we demonstrate that following apical resection of the heart on postnatal day 1, mice activate Shh ligand expression and downstream signaling. This response is largely absent when surgery is performed on non-regenerative, postnatal day 7 pups. Furthermore, an enhanced cardiac regeneration response was detected in ptch heterozygous mice which have a genetically-based constitutive increase in Shh signaling. We further show that Shh ligand is produced in the myocardium by non-myocytes and appears to regulate cardiomyocyte proliferation, as well as the recruitment of monocytes/macrophages to the regenerating area. Finally, we demonstrate that a small molecule activator of Shh signaling promotes heart regeneration, whereas an inhibitor of Shh signaling impairs the regenerative response. Together, these results implicate Shh signaling as a regulator of mammalian heart regeneration and suggest that modulating this pathway may lead to new potential therapies for cardiovascular diseases.


Asunto(s)
Corazón/fisiología , Proteínas Hedgehog/metabolismo , Miocardio/metabolismo , Regeneración , Transducción de Señal , Animales , Biomarcadores , Mamíferos , Ratones , Ratones Transgénicos
8.
Mol Cell ; 69(4): 689-698.e7, 2018 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-29429925

RESUMEN

Endothelial-to-mesenchymal transition (EndoMT) is a cellular process often initiated by the transforming growth factor ß (TGF-ß) family of ligands. Although required for normal heart valve development, deregulated EndoMT is linked to a wide range of pathological conditions. Here, we demonstrate that endothelial fatty acid oxidation (FAO) is a critical in vitro and in vivo regulator of EndoMT. We further show that this FAO-dependent metabolic regulation of EndoMT occurs through alterations in intracellular acetyl-CoA levels. Disruption of FAO via conditional deletion of endothelial carnitine palmitoyltransferase II (Cpt2E-KO) augments the magnitude of embryonic EndoMT, resulting in thickening of cardiac valves. Consistent with the known pathological effects of EndoMT, adult Cpt2E-KO mice demonstrate increased permeability in multiple vascular beds. Taken together, these results demonstrate that endothelial FAO is required to maintain endothelial cell fate and that therapeutic manipulation of endothelial metabolism could provide the basis for treating a growing number of EndoMT-linked pathological conditions.


Asunto(s)
Carnitina O-Palmitoiltransferasa/fisiología , Endotelio Vascular/metabolismo , Transición Epitelial-Mesenquimal , Ácidos Grasos/química , 3-Hidroxiacil-CoA Deshidrogenasas , Acetilcoenzima A/metabolismo , Acetil-CoA C-Aciltransferasa , Animales , Isomerasas de Doble Vínculo Carbono-Carbono , Células Cultivadas , Endotelio Vascular/citología , Enoil-CoA Hidratasa , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Oxidación-Reducción , Racemasas y Epimerasas , Transducción de Señal , Factor de Crecimiento Transformador beta/metabolismo
9.
Nat Protoc ; 12(8): 1576-1587, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28703790

RESUMEN

Mitophagy is a cellular process that selectively removes damaged, old or dysfunctional mitochondria. Defective mitophagy is thought to contribute to normal aging and to various neurodegenerative and cardiovascular diseases. Previous methods used to detect mitophagy in vivo were cumbersome, insensitive and difficult to quantify. We created a transgenic mouse model that expresses the pH-dependent fluorescent protein mt-Keima in order to more readily assess mitophagy. Keima is a pH-sensitive, dual-excitation ratiometric fluorescent protein that also exhibits resistance to lysosomal proteases. At the physiological pH of the mitochondria (pH 8.0), the shorter-wavelength excitation predominates. Within the acidic lysosome (pH 4.5) after mitophagy, mt-Keima undergoes a gradual shift to longer-wavelength excitation. In this protocol, we describe how to monitor mitophagic flux in living cells over an 18-h time frame, as well as how to quantify mitophagy using the mt-Keima probe. This protocol also describes how to use confocal microscopy to visualize mitophagy in living tissues obtained from mt-Keima transgenic mice. With this protocol, the mt-Keima probe can reliably be imaged within the first 60 min after tissue collection. We also describe how to apply mt-Keima with stimulated emission depletion (STED) microscopy, which can potentially provide substantially higher-resolution images. Typically, the approximate time frame for time-lapse fluorescence imaging of mt-Keima is 20 h for living cells. For confocal analysis of tissue from an mt-Keima mouse, the whole procedure generally takes no longer than 60 min, and the STED imaging usually takes <2 h.


Asunto(s)
Genes Reporteros , Microscopía Fluorescente/métodos , Mitofagia , Coloración y Etiquetado/métodos , Animales , Procesamiento de Imagen Asistido por Computador/métodos , Proteínas Luminiscentes/análisis , Proteínas Luminiscentes/genética , Ratones , Ratones Transgénicos , Microscopía Confocal , Proteínas Recombinantes/análisis , Proteínas Recombinantes/genética
10.
Adv Exp Med Biol ; 982: 49-63, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28551781

RESUMEN

The identification of the molecular composition of the mitochondrial calcium uniporter has allowed for the genetic manipulation of its components and the creation of various in vivo genetic models. Here, we review the initial attempts to modulate the expression of components of the calcium uniporter in a range of organisms from plants to mammals. This analysis has confirmed the strict requirement for the uniporter for in vivo mitochondrial calcium uptake and for maintaining mitochondrial calcium homeostasis. We further discuss the physiological effects following genetic manipulation of the uniporter on tissue bioenergetics and the threshold for cell death. Finally, we analyze the limited information regarding the role of various uniporter components in human disease.


Asunto(s)
Canales de Calcio/metabolismo , Señalización del Calcio , Proteínas de Unión al Calcio/metabolismo , Calcio/metabolismo , Proteínas de Transporte de Catión/metabolismo , Mitocondrias/metabolismo , Proteínas de Transporte de Membrana Mitocondrial/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Canales de Calcio/química , Canales de Calcio/genética , Proteínas de Unión al Calcio/química , Proteínas de Unión al Calcio/genética , Proteínas de Transporte de Catión/química , Proteínas de Transporte de Catión/genética , Muerte Celular , Metabolismo Energético , Genotipo , Humanos , Ratones Noqueados , Mitocondrias/patología , Proteínas de Transporte de Membrana Mitocondrial/química , Proteínas de Transporte de Membrana Mitocondrial/genética , Fenotipo , Conformación Proteica , Relación Estructura-Actividad
11.
Arch Biochem Biophys ; 613: 12-22, 2017 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-27983949

RESUMEN

We sought to explore the fate of the fatty acid synthesis pathway in human fibroblasts exposed to DNA damaging agents capable of inducing senescence, a state of irreversible growth arrest. Induction of premature senescence by doxorubicin or hydrogen peroxide led to a decrease in protein and mRNA levels of acetyl-CoA carboxylase 1 (ACC1), the enzyme that catalyzes the rate-limiting step in fatty-acid biosynthesis. ACC1 decay accompanied the activation of the DNA damage response (DDR), and resulted in decreased lipid synthesis. A reduction in protein and mRNA levels of ACC1 and in lipid synthesis was also observed in human primary fibroblasts that underwent replicative senescence. We also explored the consequences of inhibiting fatty acid synthesis in proliferating non-transformed cells. Using shRNA technology, we knocked down ACC1 in human fibroblasts. Interestingly, this metabolic perturbation was sufficient to arrest proliferation and trigger the appearance of several markers of the DDR and increase senescence associated ß-galactosidase activity. Reactive oxygen species and p38 mitogen activated protein kinase phosphorylation participated in the induction of senescence. Similar results were obtained upon silencing of fatty acid synthase (FAS) expression. Together our results point towards a tight coordination of fatty acid synthesis and cell proliferation in human fibroblasts.


Asunto(s)
Acetil-CoA Carboxilasa/metabolismo , Regulación Enzimológica de la Expresión Génica , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Proliferación Celular , Senescencia Celular , Ácidos Grasos/química , Fibroblastos/enzimología , Eliminación de Gen , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Humanos , Lentivirus , Lípidos/química , Sistema de Señalización de MAP Quinasas , Oxidantes/química , ARN Mensajero/metabolismo , Especies Reactivas de Oxígeno/metabolismo
13.
Mol Cell ; 60(4): 685-96, 2015 Nov 19.
Artículo en Inglés | MEDLINE | ID: mdl-26549682

RESUMEN

Alterations in mitophagy have been increasingly linked to aging and age-related diseases. There are, however, no convenient methods to analyze mitophagy in vivo. Here, we describe a transgenic mouse model in which we expressed a mitochondrial-targeted form of the fluorescent reporter Keima (mt-Keima). Keima is a coral-derived protein that exhibits both pH-dependent excitation and resistance to lysosomal proteases. Comparison of a wide range of primary cells and tissues generated from the mt-Keima mouse revealed significant variations in basal mitophagy. In addition, we have employed the mt-Keima mice to analyze how mitophagy is altered by conditions including diet, oxygen availability, Huntingtin transgene expression, the absence of macroautophagy (ATG5 or ATG7 expression), an increase in mitochondrial mutational load, the presence of metastatic tumors, and normal aging. The ability to assess mitophagy under a host of varying environmental and genetic perturbations suggests that the mt-Keima mouse should be a valuable resource.


Asunto(s)
Proteínas Luminiscentes/metabolismo , Ratones Transgénicos , Mitofagia , Envejecimiento/fisiología , Animales , Proteínas Luminiscentes/genética , Ratones , Especificidad de Órganos , Oxígeno/metabolismo
14.
Nat Cell Biol ; 15(12): 1464-72, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24212091

RESUMEN

Mitochondrial calcium has been postulated to regulate a wide range of processes from bioenergetics to cell death. Here, we characterize a mouse model that lacks expression of the recently discovered mitochondrial calcium uniporter (MCU). Mitochondria derived from MCU(-/-) mice have no apparent capacity to rapidly uptake calcium. Whereas basal metabolism seems unaffected, the skeletal muscle of MCU(-/-) mice exhibited alterations in the phosphorylation and activity of pyruvate dehydrogenase. In addition, MCU(-/-) mice exhibited marked impairment in their ability to perform strenuous work. We further show that mitochondria from MCU(-/-) mice lacked evidence for calcium-induced permeability transition pore (PTP) opening. The lack of PTP opening does not seem to protect MCU(-/-) cells and tissues from cell death, although MCU(-/-) hearts fail to respond to the PTP inhibitor cyclosporin A. Taken together, these results clarify how acute alterations in mitochondrial matrix calcium can regulate mammalian physiology.


Asunto(s)
Canales de Calcio/genética , Calcio/fisiología , Mitocondrias Musculares/metabolismo , Animales , Apoptosis , Canales de Calcio/deficiencia , Cardiotónicos/farmacología , Células Cultivadas , Tolerancia al Ejercicio/genética , Femenino , Fibroblastos/metabolismo , Estudios de Asociación Genética , Isoproterenol/farmacología , Masculino , Ratones , Ratones Noqueados , Proteínas de Transporte de Membrana Mitocondrial/metabolismo , Poro de Transición de la Permeabilidad Mitocondrial , Músculo Esquelético/metabolismo , Músculo Esquelético/fisiología , Daño por Reperfusión Miocárdica/metabolismo , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Fenotipo , Complejo Piruvato Deshidrogenasa/metabolismo
15.
Nat Med ; 19(10): 1281-7, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24056772

RESUMEN

Endothelial secretion of von Willebrand factor (VWF) from intracellular organelles known as Weibel-Palade bodies (WPBs) is required for platelet adhesion to the injured vessel wall. Here we demonstrate that WPBs are often found near or within autophagosomes and that endothelial autophagosomes contain abundant VWF protein. Pharmacological inhibitors of autophagy or knockdown of the essential autophagy genes Atg5 or Atg7 inhibits the in vitro secretion of VWF. Furthermore, although mice with endothelial-specific deletion of Atg7 have normal vessel architecture and capillary density, they exhibit impaired epinephrine-stimulated VWF release, reduced levels of high-molecular weight VWF multimers and a corresponding prolongation of bleeding times. Endothelial-specific deletion of Atg5 or pharmacological inhibition of autophagic flux results in a similar in vivo alteration of hemostasis. Thus, autophagy regulates endothelial VWF secretion, and transient pharmacological inhibition of autophagic flux may be a useful strategy to prevent thrombotic events.


Asunto(s)
Autofagia , Células Endoteliales/metabolismo , Factor de von Willebrand/metabolismo , Proteína 5 Relacionada con la Autofagia , Proteína 7 Relacionada con la Autofagia , Exocitosis , Hemostasis , Humanos , Proteínas Asociadas a Microtúbulos/genética , Enzimas Activadoras de Ubiquitina/genética , Cuerpos de Weibel-Palade/metabolismo
16.
Cell Rep ; 4(5): 913-20, 2013 Sep 12.
Artículo en Inglés | MEDLINE | ID: mdl-23994476

RESUMEN

We analyzed aging parameters using a mechanistic target of rapamycin (mTOR) hypomorphic mouse model. Mice with two hypomorphic (mTOR(Δ/Δ)) alleles are viable but express mTOR at approximately 25% of wild-type levels. These animals demonstrate reduced mTORC1 and mTORC2 activity and exhibit an approximately 20% increase in median survival. While mTOR(Δ/Δ) mice are smaller than wild-type mice, these animals do not demonstrate any alterations in normalized food intake, glucose homeostasis, or metabolic rate. Consistent with their increased lifespan, mTOR(Δ/Δ) mice exhibited a reduction in a number of aging tissue biomarkers. Functional assessment suggested that, as mTOR(Δ/Δ) mice age, they exhibit a marked functional preservation in many, but not all, organ systems. Thus, in a mammalian model, while reducing mTOR expression markedly increases overall lifespan, it affects the age-dependent decline in tissue and organ function in a segmental fashion.


Asunto(s)
Longevidad/fisiología , Serina-Treonina Quinasas TOR/deficiencia , Serina-Treonina Quinasas TOR/genética , Envejecimiento/fisiología , Animales , Femenino , Glucosa/metabolismo , Homeostasis , Masculino , Mamíferos , Ratones , Transducción de Señal , Serina-Treonina Quinasas TOR/biosíntesis
17.
Nature ; 492(7428): 199-204, 2012 Dec 13.
Artículo en Inglés | MEDLINE | ID: mdl-23201684

RESUMEN

Although initially viewed as unregulated, increasing evidence suggests that cellular necrosis often proceeds through a specific molecular program. In particular, death ligands such as tumour necrosis factor (TNF)-α activate necrosis by stimulating the formation of a complex containing receptor-interacting protein 1 (RIP1) and receptor-interacting protein 3 (RIP3). Relatively little is known regarding how this complex formation is regulated. Here, we show that the NAD-dependent deacetylase SIRT2 binds constitutively to RIP3 and that deletion or knockdown of SIRT2 prevents formation of the RIP1-RIP3 complex in mice. Furthermore, genetic or pharmacological inhibition of SIRT2 blocks cellular necrosis induced by TNF-α. We further demonstrate that RIP1 is a critical target of SIRT2-dependent deacetylation. Using gain- and loss-of-function mutants, we demonstrate that acetylation of RIP1 lysine 530 modulates RIP1-RIP3 complex formation and TNF-α-stimulated necrosis. In the setting of ischaemia-reperfusion injury, RIP1 is deacetylated in a SIRT2-dependent fashion. Furthermore, the hearts of Sirt2(-/-) mice, or wild-type mice treated with a specific pharmacological inhibitor of SIRT2, show marked protection from ischaemic injury. Taken together, these results implicate SIRT2 as an important regulator of programmed necrosis and indicate that inhibitors of this deacetylase may constitute a novel approach to protect against necrotic injuries, including ischaemic stroke and myocardial infarction.


Asunto(s)
Necrosis/enzimología , Sirtuina 2/genética , Sirtuina 2/metabolismo , Acetilación , Animales , Línea Celular , Femenino , Células HEK293 , Células HeLa , Humanos , Células Jurkat , Masculino , Ratones , Proteínas de Complejo Poro Nuclear/metabolismo , Unión Proteica , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo
18.
Science ; 336(6078): 225-8, 2012 Apr 13.
Artículo en Inglés | MEDLINE | ID: mdl-22499945

RESUMEN

Withdrawal of nutrients triggers an exit from the cell division cycle, the induction of autophagy, and eventually the activation of cell death pathways. The relation, if any, among these events is not well characterized. We found that starved mouse embryonic fibroblasts lacking the essential autophagy gene product Atg7 failed to undergo cell cycle arrest. Independent of its E1-like enzymatic activity, Atg7 could bind to the tumor suppressor p53 to regulate the transcription of the gene encoding the cell cycle inhibitor p21(CDKN1A). With prolonged metabolic stress, the absence of Atg7 resulted in augmented DNA damage with increased p53-dependent apoptosis. Inhibition of the DNA damage response by deletion of the protein kinase Chk2 partially rescued postnatal lethality in Atg7(-/-) mice. Thus, when nutrients are limited, Atg7 regulates p53-dependent cell cycle and cell death pathways.


Asunto(s)
Proteínas Asociadas a Microtúbulos/metabolismo , Estrés Fisiológico , Proteína p53 Supresora de Tumor/metabolismo , Enzimas Activadoras de Ubiquitina/metabolismo , Animales , Apoptosis , Autofagia , Proteína 7 Relacionada con la Autofagia , Ciclo Celular , Puntos de Control del Ciclo Celular , Línea Celular Tumoral , Células Cultivadas , Quinasa de Punto de Control 2 , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Daño del ADN , Regulación de la Expresión Génica , Humanos , Ratones , Proteínas Asociadas a Microtúbulos/genética , Fosforilación , Regiones Promotoras Genéticas , Unión Proteica , Multimerización de Proteína , Proteínas Serina-Treonina Quinasas/genética , Transcripción Genética , Enzimas Activadoras de Ubiquitina/genética
19.
Cell Cycle ; 11(7): 1383-92, 2012 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-22421146

RESUMEN

Oncogene-induced senescence (OIS) is characterized by permanent growth arrest and the acquisition of a secretory, pro-inflammatory state. Increasingly, OIS is viewed as an important barrier to tumorgenesis. Surprisingly, relatively little is known about the metabolic changes that accompany and therefore may contribute to OIS. Here, we have performed a metabolomic and bioenergetic analysis of Ras-induced senescence. Profiling approximately 300 different intracellular metabolites reveals that cells that have undergone OIS develop a unique metabolic signature that differs markedly from cells undergoing replicative senescence. A number of lipid metabolites appear uniquely increased in OIS cells, including a marked increase in the level of certain intracellular long chain fatty acids. Functional studies reveal that this alteration in the metabolome reflects substantial changes in overall lipid metabolism. In particular, Ras-induced senescent cells manifest a decline in lipid synthesis and a significant increase in fatty acid oxidation. Increased fatty acid oxidation results in an unexpectedly high rate of basal oxygen consumption in cells that have undergone OIS. Pharmacological or genetic inhibition of carnitine palmitoyltransferase 1, the rate-limiting step in mitochondrial fatty acid oxidation, restores a pre-senescent metabolic rate and, surprisingly, selectively inhibits the secretory, pro-inflammatory state that accompanies OIS. Thus, Ras-induced senescent cells demonstrate profound alterations in their metabolic and bioenergetic profiles, particularly with regards to the levels, synthesis and oxidation of free fatty acids. Furthermore, the inflammatory phenotype that accompanies OIS appears to be related to these underlying changes in cellular metabolism.


Asunto(s)
Senescencia Celular/genética , Metabolismo Energético/genética , Metabolismo de los Lípidos/genética , Oncogenes , Carnitina O-Palmitoiltransferasa/antagonistas & inhibidores , Carnitina O-Palmitoiltransferasa/metabolismo , Línea Celular Tumoral , Proliferación Celular , Citocinas/biosíntesis , Perfilación de la Expresión Génica , Humanos , Inflamación/genética , Metabolómica/métodos , Proteína Oncogénica p21(ras)/genética , Proteína Oncogénica p21(ras)/metabolismo , Consumo de Oxígeno
20.
Nat Commun ; 2: 593, 2011 Dec 20.
Artículo en Inglés | MEDLINE | ID: mdl-22186889

RESUMEN

The tumour suppressor BRCA1 is mutated in familial breast and ovarian cancer but its role in protecting other tissues from DNA damage has not been explored. Here we show a new role for BRCA1 as a gatekeeper of cardiac function and survival. In mice, loss of BRCA1 in cardiomyocytes results in adverse cardiac remodelling, poor ventricular function and higher mortality in response to ischaemic or genotoxic stress. Mechanistically, loss of cardiomyocyte BRCA1 results in impaired DNA double-strand break repair and activated p53-mediated pro-apoptotic signalling culminating in increased cardiomyocyte apoptosis, whereas deletion of the p53 gene rescues BRCA1-deficient mice from cardiac failure. In human adult and fetal cardiac tissues, ischaemia induces double-strand breaks and upregulates BRCA1 expression. These data reveal BRCA1 as a novel and essential adaptive response molecule shielding cardiomyocytes from DNA damage, apoptosis and heart dysfunction. BRCA1 mutation carriers, in addition to risk of breast and ovarian cancer, may be at a previously unrecognized risk of cardiac failure.


Asunto(s)
Apoptosis/genética , Proteína BRCA1/genética , Miocardio/metabolismo , Miocitos Cardíacos/metabolismo , Daño por Reperfusión/metabolismo , Proteína p53 Supresora de Tumor/genética , Adaptación Fisiológica , Adulto , Animales , Proteína BRCA1/deficiencia , Neoplasias de la Mama/genética , Roturas del ADN de Doble Cadena , Femenino , Eliminación de Gen , Humanos , Masculino , Ratones , Ratones Noqueados , Mutación , Miocardio/patología , Miocitos Cardíacos/citología , Neoplasias Ováricas/genética , Daño por Reperfusión/genética , Transducción de Señal , Proteína p53 Supresora de Tumor/deficiencia , Remodelación Ventricular/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA