Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 157(3): 565-79, 2014 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-24766806

RESUMEN

The mammalian heart has a remarkable regenerative capacity for a short period of time after birth, after which the majority of cardiomyocytes permanently exit cell cycle. We sought to determine the primary postnatal event that results in cardiomyocyte cell-cycle arrest. We hypothesized that transition to the oxygen-rich postnatal environment is the upstream signal that results in cell-cycle arrest of cardiomyocytes. Here, we show that reactive oxygen species (ROS), oxidative DNA damage, and DNA damage response (DDR) markers significantly increase in the heart during the first postnatal week. Intriguingly, postnatal hypoxemia, ROS scavenging, or inhibition of DDR all prolong the postnatal proliferative window of cardiomyocytes, whereas hyperoxemia and ROS generators shorten it. These findings uncover a protective mechanism that mediates cardiomyocyte cell-cycle arrest in exchange for utilization of oxygen-dependent aerobic metabolism. Reduction of mitochondrial-dependent oxidative stress should be an important component of cardiomyocyte proliferation-based therapeutic approaches.


Asunto(s)
Puntos de Control del Ciclo Celular , Miocitos Cardíacos/citología , Especies Reactivas de Oxígeno/metabolismo , Acetilcisteína/farmacología , Animales , Proliferación Celular/efectos de los fármacos , Daño del ADN , Depuradores de Radicales Libres/farmacología , Ratones , Mitocondrias/metabolismo , Miocitos Cardíacos/metabolismo , Pez Cebra
2.
Am J Respir Crit Care Med ; 206(1): 56-69, 2022 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-35417304

RESUMEN

Rationale: Genetic studies of idiopathic pulmonary fibrosis (IPF) have improved our understanding of this disease, but not all causal loci have been identified. Objectives: To identify genes enriched with rare deleterious variants in IPF and familial pulmonary fibrosis. Methods: We performed gene burden analysis of whole-exome data, tested single variants for disease association, conducted KIF15 (kinesin family member 15) functional studies, and examined human lung single-cell RNA sequencing data. Measurements and Main Results: Gene burden analysis of 1,725 cases and 23,509 control subjects identified heterozygous rare deleterious variants in KIF15, a kinesin involved in spindle separation during mitosis, and three telomere-related genes (TERT [telomerase reverse transcriptase], RTEL1 [regulator of telomere elongation helicase 1], and PARN [poly(A)-specific ribonuclease]). KIF15 was implicated in autosomal-dominant models of rare deleterious variants (odds ratio [OR], 4.9; 95% confidence interval [CI], 2.7-8.8; P = 2.55 × 10-7) and rare protein-truncating variants (OR, 7.6; 95% CI, 3.3-17.1; P = 8.12 × 10-7). Meta-analyses of the discovery and replication cohorts, including 2,966 cases and 29,817 control subjects, confirm the involvement of KIF15 plus the three telomere-related genes. A common variant within a KIF15 intron (rs74341405; OR, 1.6; 95% CI, 1.4-1.9; P = 5.63 × 10-10) is associated with IPF risk, confirming a prior report. Lymphoblastoid cells from individuals heterozygous for the common variant have decreased KIF15 and reduced rates of cell growth. Cell proliferation is dependent on KIF15 in the presence of an inhibitor of Eg5/KIF11, which has partially redundant function. KIF15 is expressed specifically in replicating human lung cells and shows diminished expression in replicating epithelial cells of patients with IPF. Conclusions: Both rare deleterious variants and common variants in KIF15 link a nontelomerase pathway of cell proliferation with IPF susceptibility.


Asunto(s)
Fibrosis Pulmonar Idiopática , Cinesinas , Telomerasa , Exoma , Humanos , Fibrosis Pulmonar Idiopática/genética , Cinesinas/genética , Telomerasa/genética , Telómero
3.
J Biol Chem ; 297(3): 101037, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34343565

RESUMEN

Besides contributing to anabolism, cellular metabolites serve as substrates or cofactors for enzymes and may also have signaling functions. Given these roles, multiple control mechanisms likely ensure fidelity of metabolite-generating enzymes. Acetate-dependent acetyl CoA synthetases (ACS) are de novo sources of acetyl CoA, a building block for fatty acids and a substrate for acetyltransferases. Eukaryotic acetate-dependent acetyl CoA synthetase 2 (Acss2) is predominantly cytosolic, but is also found in the nucleus following oxygen or glucose deprivation, or upon acetate exposure. Acss2-generated acetyl CoA is used in acetylation of Hypoxia-Inducible Factor 2 (HIF-2), a stress-responsive transcription factor. Mutation of a putative nuclear localization signal in endogenous Acss2 abrogates HIF-2 acetylation and signaling, but surprisingly also results in reduced Acss2 protein levels due to unmasking of two protein destabilization elements (PDE) in the Acss2 hinge region. In the current study, we identify up to four additional PDE in the Acss2 hinge region and determine that a previously identified PDE, the ABC domain, consists of two functional PDE. We show that the ABC domain and other PDE are likely masked by intramolecular interactions with other domains in the Acss2 hinge region. We also characterize mice with a prematurely truncated Acss2 that exposes a putative ABC domain PDE, which exhibits reduced Acss2 protein stability and impaired HIF-2 signaling. Finally, using primary mouse embryonic fibroblasts, we demonstrate that the reduced stability of select Acss2 mutant proteins is due to a shortened half-life, which is a result of enhanced degradation via a nonproteasome, nonautophagy pathway.


Asunto(s)
Acetato CoA Ligasa/química , Acetato CoA Ligasa/metabolismo , Acetatos/metabolismo , Acetato CoA Ligasa/genética , Secuencia de Aminoácidos , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Fibroblastos/química , Fibroblastos/enzimología , Humanos , Ratones , Unión Proteica , Dominios Proteicos , Estabilidad Proteica , Alineación de Secuencia
4.
Nature ; 523(7559): 226-30, 2015 Jul 09.
Artículo en Inglés | MEDLINE | ID: mdl-26098368

RESUMEN

Although the adult mammalian heart is incapable of meaningful functional recovery following substantial cardiomyocyte loss, it is now clear that modest cardiomyocyte turnover occurs in adult mouse and human hearts, mediated primarily by proliferation of pre-existing cardiomyocytes. However, fate mapping of these cycling cardiomyocytes has not been possible thus far owing to the lack of identifiable genetic markers. In several organs, stem or progenitor cells reside in relatively hypoxic microenvironments where the stabilization of the hypoxia-inducible factor 1 alpha (Hif-1α) subunit is critical for their maintenance and function. Here we report fate mapping of hypoxic cells and their progenies by generating a transgenic mouse expressing a chimaeric protein in which the oxygen-dependent degradation (ODD) domain of Hif-1α is fused to the tamoxifen-inducible CreERT2 recombinase. In mice bearing the creERT2-ODD transgene driven by either the ubiquitous CAG promoter or the cardiomyocyte-specific α myosin heavy chain promoter, we identify a rare population of hypoxic cardiomyocytes that display characteristics of proliferative neonatal cardiomyocytes, such as smaller size, mononucleation and lower oxidative DNA damage. Notably, these hypoxic cardiomyocytes contributed widely to new cardiomyocyte formation in the adult heart. These results indicate that hypoxia signalling is an important hallmark of cycling cardiomyocytes, and suggest that hypoxia fate mapping can be a powerful tool for identifying cycling cells in adult mammals.


Asunto(s)
Miocardio/citología , Miocitos Cardíacos/citología , Proteínas Recombinantes de Fusión/metabolismo , Animales , Hipoxia de la Célula , Proliferación Celular/genética , Femenino , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Masculino , Ratones , Ratones Transgénicos , Miocitos Cardíacos/metabolismo , Estructura Terciaria de Proteína , Proteínas Recombinantes de Fusión/genética , Recombinasas/genética , Recombinasas/metabolismo , Transducción de Señal , Células Madre/citología , Células Madre/metabolismo
5.
Bioconjug Chem ; 31(5): 1449-1462, 2020 05 20.
Artículo en Inglés | MEDLINE | ID: mdl-32302483

RESUMEN

Advances in bioconjugation, the ability to link biomolecules to each other, small molecules, surfaces, and more, can spur the development of advanced materials and therapeutics. We have discovered that pyrocinchonimide, the dimethylated analogue of maleimide, undergoes a surprising transformation with biomolecules. The reaction targets amines and involves an imide transfer, which has not been previously reported for bioconjugation purposes. Despite their similarity to maleimides, pyrocinchonimides do not react with free thiols. Though both lysine residues and the N-termini of proteins can receive the transferred imide, the reaction also exhibits a marked preference for certain amines that cannot solely be ascribed to solvent accessibility. This property is peculiar among amine-targeting reactions and can reduce combinatorial diversity when many available reactive amines are available, such as in the formation of antibody-drug conjugates. Unlike amides, the modification undergoes very slow reversion under high pH conditions. The reaction offers a thermodynamically controlled route to single or multiple modifications of proteins for a wide range of applications.


Asunto(s)
Aminas/química , Imidas/química , Proteínas/química , Concentración de Iones de Hidrógeno , Cinética , Lisina/química , Solventes/química , Compuestos de Sulfhidrilo/química , Termodinámica
7.
Proc Natl Acad Sci U S A ; 110(19): E1788-96, 2013 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-23610397

RESUMEN

Breathing and blood pressure are under constant homeostatic regulation to maintain optimal oxygen delivery to the tissues. Chemosensory reflexes initiated by the carotid body and catecholamine secretion from the adrenal medulla are the principal mechanisms for maintaining respiratory and cardiovascular homeostasis; however, the underlying molecular mechanisms are not known. Here, we report that balanced activity of hypoxia-inducible factor-1 (HIF-1) and HIF-2 is critical for oxygen sensing by the carotid body and adrenal medulla, and for their control of cardio-respiratory function. In Hif2α(+/-) mice, partial HIF-2α deficiency increased levels of HIF-1α and NADPH oxidase 2, leading to an oxidized intracellular redox state, exaggerated hypoxic sensitivity, and cardio-respiratory abnormalities, which were reversed by treatment with a HIF-1α inhibitor or a superoxide anion scavenger. Conversely, in Hif1α(+/-) mice, partial HIF-1α deficiency increased levels of HIF-2α and superoxide dismutase 2, leading to a reduced intracellular redox state, blunted oxygen sensing, and impaired carotid body and ventilatory responses to chronic hypoxia, which were corrected by treatment with a HIF-2α inhibitor. None of the abnormalities observed in Hif1α(+/-) mice or Hif2α(+/-) mice were observed in Hif1α(+/-);Hif2α(+/-) mice. These observations demonstrate that redox balance, which is determined by mutual antagonism between HIF-α isoforms, establishes the set point for hypoxic sensing by the carotid body and adrenal medulla, and is required for maintenance of cardio-respiratory homeostasis.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Cuerpo Carotídeo/fisiología , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Oxígeno/metabolismo , Médula Suprarrenal/fisiología , Animales , Presión Sanguínea , Sistema Cardiovascular , Cuerpo Carotídeo/metabolismo , Catecolaminas/metabolismo , Heterocigoto , Homeostasis , Hipoxia , Masculino , Glicoproteínas de Membrana/metabolismo , Ratones , Ratones Transgénicos , NADPH Oxidasa 2 , NADPH Oxidasas/metabolismo , Oxidación-Reducción , Células PC12 , Ratas , Superóxido Dismutasa/metabolismo
8.
Nat Chem Biol ; 9(4): 271-6, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23434853

RESUMEN

Hypoxia inducible factors (HIFs) are heterodimeric transcription factors induced in many cancers where they frequently promote the expression of protumorigenic pathways. Though transcription factors are typically considered 'undruggable', the PAS-B domain of the HIF-2α subunit contains a large cavity within its hydrophobic core that offers a unique foothold for small-molecule regulation. Here we identify artificial ligands that bind within this pocket and characterize the resulting structural and functional changes caused by binding. Notably, these ligands antagonize HIF-2 heterodimerization and DNA-binding activity in vitro and in cultured cells, reducing HIF-2 target gene expression. Despite the high sequence identity between HIF-2α and HIF-1α, these ligands are highly selective and do not affect HIF-1 function. These chemical tools establish the molecular basis for selective regulation of HIF-2, providing potential therapeutic opportunities to intervene in HIF-2-driven tumors, such as renal cell carcinomas.


Asunto(s)
Antineoplásicos/farmacología , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/antagonistas & inhibidores , Proteínas de Neoplasias/antagonistas & inhibidores , Bibliotecas de Moléculas Pequeñas/farmacología , Regulación Alostérica , Antineoplásicos/química , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Sitios de Unión , Línea Celular Tumoral , Cristalografía por Rayos X , Ensayos Analíticos de Alto Rendimiento , Humanos , Cinética , Ligandos , Simulación del Acoplamiento Molecular , Proteínas de Neoplasias/metabolismo , Resonancia Magnética Nuclear Biomolecular , Unión Proteica , Estructura Terciaria de Proteína , Bibliotecas de Moléculas Pequeñas/química
9.
Proc Natl Acad Sci U S A ; 108(7): 3065-70, 2011 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-21288809

RESUMEN

Cardiorespiratory functions in mammals are exquisitely sensitive to changes in arterial O(2) levels. Hypoxia-inducible factors (e.g., HIF-1 and HIF-2) mediate transcriptional responses to reduced oxygen availability. We demonstrate that haploinsufficiency for the O(2)-regulated HIF-2α subunit results in augmented carotid body sensitivity to hypoxia, irregular breathing, apneas, hypertension, and elevated plasma norepinephrine levels in adult Hif-2α(+/-) mice. These dysregulated autonomic responses were associated with increased oxidative stress and decreased mitochondrial electron transport chain complex I activity in adrenal medullae as a result of decreased expression of major cytosolic and mitochondrial antioxidant enzymes. Systemic administration of a membrane-permeable antioxidant prevented oxidative stress, normalized hypoxic sensitivity of the carotid body, and restored autonomic functions in Hif-2α(+/-) mice. Thus, HIF-2α-dependent redox regulation is required for maintenance of carotid body function and cardiorespiratory homeostasis.


Asunto(s)
Médula Suprarrenal/metabolismo , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Cuerpo Carotídeo/fisiología , Hipertensión/fisiopatología , Hipoxia/fisiopatología , Estrés Oxidativo/fisiología , Mecánica Respiratoria/fisiología , Análisis de Varianza , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Presión Sanguínea , Western Blotting , Complejo I de Transporte de Electrón/metabolismo , Perfilación de la Expresión Génica , Inmunohistoquímica , Ratones , Ratones Noqueados , Norepinefrina/metabolismo , Oxidación-Reducción , Consumo de Oxígeno , Pletismografía Total , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
10.
J Biol Chem ; 287(36): 30800-11, 2012 Aug 31.
Artículo en Inglés | MEDLINE | ID: mdl-22807441

RESUMEN

Hypoxia-inducible factors (HIFs) are oxygen-sensitive transcription factors. HIF-1α plays a prominent role in hypoxic gene induction. HIF-2α target genes are more restricted but include erythropoietin (Epo), one of the most highly hypoxia-inducible genes in mammals. We previously reported that HIF-2α is acetylated during hypoxia but is rapidly deacetylated by the stress-responsive deacetylase Sirtuin 1. We now demonstrate that the lysine acetyltransferases cAMP-response element-binding protein-binding protein (CBP) and p300 are required for efficient Epo induction during hypoxia. However, despite close structural similarity, the roles of CBP and p300 differ in HIF signaling. CBP acetylates HIF-2α, is a major coactivator for HIF-2-mediated Epo induction, and is required for Sirt1 augmentation of HIF-2 signaling during hypoxia in Hep3B cells. In comparison, p300 is a major contributor for HIF-1 signaling as indicated by induction of Pgk1. Whereas CBP can bind with HIF-2α independent of the HIF-2α C-terminal activation domain via enzyme/substrate interactions, p300 only complexes with HIF-2α through the C-terminal activation domain. Maximal CBP/HIF-2 signaling requires intact CBP acetyltransferase activity in both Hep3B cells as well as in mice.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Proteína de Unión a CREB/metabolismo , Fragmentos de Péptidos/metabolismo , Sialoglicoproteínas/metabolismo , Transducción de Señal/fisiología , Sirtuina 1/metabolismo , Factores de Transcripción p300-CBP/metabolismo , Acetilación , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Proteína de Unión a CREB/genética , Línea Celular , Eritropoyetina/biosíntesis , Eritropoyetina/genética , Humanos , Ratones , Fragmentos de Péptidos/genética , Fosfoglicerato Quinasa/genética , Fosfoglicerato Quinasa/metabolismo , Estructura Terciaria de Proteína , Sialoglicoproteínas/genética , Sirtuina 1/genética , Factores de Transcripción p300-CBP/genética
11.
Nat Med ; 12(7): 793-800, 2006 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16799557

RESUMEN

Vascular endothelial growth factor (VEGF) exerts crucial functions during pathological angiogenesis and normal physiology. We observed increased hematocrit (60-75%) after high-grade inhibition of VEGF by diverse methods, including adenoviral expression of soluble VEGF receptor (VEGFR) ectodomains, recombinant VEGF Trap protein and the VEGFR2-selective antibody DC101. Increased production of red blood cells (erythrocytosis) occurred in both mouse and primate models, and was associated with near-complete neutralization of VEGF corneal micropocket angiogenesis. High-grade inhibition of VEGF induced hepatic synthesis of erythropoietin (Epo, encoded by Epo) >40-fold through a HIF-1alpha-independent mechanism, in parallel with suppression of renal Epo mRNA. Studies using hepatocyte-specific deletion of the Vegfa gene and hepatocyte-endothelial cell cocultures indicated that blockade of VEGF induced hepatic Epo by interfering with homeostatic VEGFR2-dependent paracrine signaling involving interactions between hepatocytes and endothelial cells. These data indicate that VEGF is a previously unsuspected negative regulator of hepatic Epo synthesis and erythropoiesis and suggest that levels of Epo and erythrocytosis could represent noninvasive surrogate markers for stringent blockade of VEGF in vivo.


Asunto(s)
Eritropoyetina/fisiología , Hígado/fisiología , Factor A de Crecimiento Endotelial Vascular/fisiología , Animales , Hematócrito , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones SCID , Ratones Transgénicos , Modelos Animales , Policitemia/fisiopatología , Receptores de Factores de Crecimiento Endotelial Vascular/fisiología , Vasos Retinianos/fisiología
12.
Nat Genet ; 35(4): 331-40, 2003 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-14608355

RESUMEN

Hypoxia-inducible factor (HIF) transcription factors respond to multiple environmental stressors, including hypoxia and hypoglycemia. We report that mice lacking the HIF family member HIF-2alpha (encoded by Epas1) have a syndrome of multiple-organ pathology, biochemical abnormalities and altered gene expression patterns. Histological and ultrastructural analyses showed retinopathy, hepatic steatosis, cardiac hypertrophy, skeletal myopathy, hypocellular bone marrow, azoospermia and mitochondrial abnormalities in these mice. Serum and urine metabolite studies showed hypoglycemia, lactic acidosis, altered Krebs cycle function and dysregulated fatty acid oxidation. Biochemical assays showed enhanced generation of reactive oxygen species (ROS), whereas molecular analyses indicated reduced expression of genes encoding the primary antioxidant enzymes (AOEs). Transfection analyses showed that HIF-2alpha could efficiently transactivate the promoters of the primary AOEs. Prenatal or postnatal treatment of Epas1-/- mice with a superoxide dismutase (SOD) mimetic reversed several aspects of the null phenotype. We propose a rheostat role for HIF-2alpha that allows for the maintenance of ROS as well as mitochondrial homeostasis.


Asunto(s)
Anomalías Múltiples , Homeostasis/fisiología , Proteínas de Neoplasias , Especies Reactivas de Oxígeno , Transactivadores/metabolismo , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico , Hipoxia de la Célula , Complejo IV de Transporte de Electrones , Regulación de la Expresión Génica , Corazón/fisiología , Homocigoto , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Imitación Molecular , Músculo Esquelético/ultraestructura , Estrés Oxidativo , Peroxidasas , Peroxiredoxina III , Peroxirredoxinas , Superóxido Dismutasa , Superóxidos , Tasa de Supervivencia , Transactivadores/deficiencia , Transactivadores/genética , Transfección
13.
PLoS One ; 18(3): e0282223, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36862715

RESUMEN

The microenvironment of solid tumors is characterized by oxygen and glucose deprivation. Acss2/HIF-2 signaling coordinates essential genetic regulators including acetate-dependent acetyl CoA synthetase 2 (Acss2), Creb binding protein (Cbp), Sirtuin 1 (Sirt1), and Hypoxia Inducible Factor 2α (HIF-2α). We previously shown in mice that exogenous acetate augments growth and metastasis of flank tumors derived from fibrosarcoma-derived HT1080 cells in an Acss2/HIF-2 dependent manner. Colonic epithelial cells are exposed to the highest acetate levels in the body. We reasoned that colon cancer cells, like fibrosarcoma cells, may respond to acetate in a pro-growth manner. In this study, we examine the role of Acss2/HIF-2 signaling in colon cancer. We find that Acss2/HIF-2 signaling is activated by oxygen or glucose deprivation in two human colon cancer-derived cell lines, HCT116 and HT29, and is crucial for colony formation, migration, and invasion in cell culture studies. Flank tumors derived from HCT116 and HT29 cells exhibit augmented growth in mice when supplemented with exogenous acetate in an Acss2/HIF-2 dependent manner. Finally, Acss2 in human colon cancer samples is most frequently localized in the nucleus, consistent with it having a signaling role. Targeted inhibition of Acss2/HIF-2 signaling may have synergistic effects for some colon cancer patients.


Asunto(s)
Neoplasias del Colon , Fibrosarcoma , Humanos , Animales , Ratones , Acetato CoA Ligasa , Transducción de Señal , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Microambiente Tumoral
14.
J Biol Chem ; 286(16): 13869-78, 2011 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-21345792

RESUMEN

Hypoxia-inducible factors (HIFs) are stress-responsive transcriptional regulators of cellular and physiological processes involved in oxygen metabolism. Although much is understood about the molecular machinery that confers HIF responsiveness to oxygen, far less is known about HIF isoform-specific mechanisms of regulation, despite the fact that HIF-1 and HIF-2 exhibit distinct biological roles. We recently determined that the stress-responsive genetic regulator sirtuin 1 (Sirt1) selectively augments HIF-2 signaling during hypoxia. However, the mechanism by which Sirt1 maintains activity during hypoxia is unknown. In this report, we demonstrate that Sirt1 gene expression increases in a HIF-dependent manner during hypoxia in Hep3B and in HT1080 cells. Impairment of HIF signaling affects Sirt1 deacetylase activity as decreased HIF-1 signaling results in the appearance of acetylated HIF-2α, which is detected without pharmacological inhibition of Sirt1. We also find that Sirt1 augments HIF-2 mediated, but not HIF-1 mediated, transcriptional activation of the isolated Sirt1 promoter. These data in summary reveal a bidirectional link of HIF and Sirt1 signaling during hypoxia.


Asunto(s)
Regulación de la Expresión Génica , Factor 1 Inducible por Hipoxia/metabolismo , Hipoxia , Sirtuina 1/biosíntesis , Acetilación , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Línea Celular Tumoral , Inmunoprecipitación de Cromatina , Humanos , Ratones , Ratones Endogámicos C57BL , Procesamiento Proteico-Postraduccional , Transducción de Señal , Activación Transcripcional
15.
Proc Natl Acad Sci U S A ; 106(4): 1199-204, 2009 Jan 27.
Artículo en Inglés | MEDLINE | ID: mdl-19147445

RESUMEN

Intermittent hypoxia (IH) occurs in many pathological conditions including recurrent apneas. Hypoxia-inducible factors (HIFs) 1 and 2 mediate transcriptional responses to low O(2). A previous study showed that HIF-1 mediates some of the IH-evoked physiological responses. Because HIF-2alpha is an orthologue of HIF-1alpha, we examined the effects of IH on HIF-2alpha, the O(2)-regulated subunit expression, in pheochromocytoma 12 cell cultures. In contrast to the up-regulation of HIF-1alpha, HIF-2alpha was down-regulated by IH. Similar down-regulation of HIF-2alpha was also seen in carotid bodies and adrenal medullae from IH-exposed rats. Inhibitors of calpain proteases (ALLM, ALLN) prevented IH-evoked degradation of HIF-2alpha whereas inhibitors of prolyl hydroxylases or proteosome were ineffective. IH activated calpain proteases and down-regulated the endogenous calpain inhibitor calpastatin. IH-evoked HIF-2alpha degradation led to inhibition of SOD2 transcription, resulting in oxidative stress. Over-expression of transcriptionally active HIF-2alpha prevented IH-evoked oxidative stress and restored SOD2 activity. Systemic treatment of IH-exposed rats with ALLM rescued HIF-2alpha degradation and restored SOD2 activity, thereby preventing oxidative stress and hypertension. These observations demonstrate that, unlike continuous hypoxia, IH leads to down-regulation of HIF-2alpha via a calpain-dependent signaling pathway and results in oxidative stress as well as autonomic morbidities.


Asunto(s)
Apnea/enzimología , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Calpaína/metabolismo , Estrés Oxidativo , Procesamiento Proteico-Postraduccional , Animales , Apnea/mortalidad , Sistema Nervioso Autónomo/efectos de los fármacos , Sistema Nervioso Autónomo/patología , Señalización del Calcio/efectos de los fármacos , Proteínas de Unión al Calcio/metabolismo , Calpaína/antagonistas & inhibidores , Hipoxia de la Célula/efectos de los fármacos , Regulación hacia Abajo/efectos de los fármacos , Activación Enzimática/efectos de los fármacos , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Masculino , Oligopéptidos/administración & dosificación , Oligopéptidos/farmacología , Estrés Oxidativo/efectos de los fármacos , Células PC12 , Unión Proteica/efectos de los fármacos , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Superóxido Dismutasa/antagonistas & inhibidores
16.
Dev Biol ; 337(2): 396-404, 2010 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-19914232

RESUMEN

The mechanisms that regulate skeletal muscle differentiation, fiber type diversity and muscle regeneration are incompletely defined. Forkhead transcription factors are critical regulators of cellular fate determination, proliferation, and differentiation. We identified a forkhead/winged helix transcription factor, Foxj3, which was expressed in embryonic and adult skeletal muscle. To define the functional role of Foxj3, we examined Foxj3 mutant mice. Foxj3 mutant mice are viable but have significantly fewer Type I slow-twitch myofibers and have impaired skeletal muscle contractile function compared to their wild type controls. In response to a severe injury, Foxj3 mutant mice have impaired muscle regeneration. Foxj3 mutant myogenic progenitor cells have perturbed cell cycle kinetics and decreased expression of Mef2c. Examination of the skeletal muscle 5' upstream enhancer of the Mef2c gene revealed an evolutionary conserved forkhead binding site (FBS). Transcriptional assays in C2C12 myoblasts revealed that Foxj3 transcriptionally activates the Mef2c gene in a dose dependent fashion and binds to the conserved FBS. Together, these studies support the hypothesis that Foxj3 is an important regulator of myofiber identity and muscle regeneration through the transcriptional activation of the Mef2c gene.


Asunto(s)
Envejecimiento/metabolismo , Proteínas de Unión al ADN/metabolismo , Fibras Musculares Esqueléticas/metabolismo , Factores Reguladores Miogénicos/genética , Factores de Transcripción/metabolismo , Activación Transcripcional/genética , Animales , Secuencia de Bases , Ciclo Celular , Proliferación Celular , Proteínas de Unión al ADN/genética , Factores de Transcripción Forkhead , Regulación del Desarrollo de la Expresión Génica , Técnicas de Silenciamiento del Gen , Factores de Transcripción MEF2 , Ratones , Ratones Mutantes , Datos de Secuencia Molecular , Contracción Muscular/fisiología , Desarrollo de Músculos/genética , Fibras Musculares Esqueléticas/patología , Fibras Musculares Esqueléticas/ultraestructura , Mioblastos/citología , Mioblastos/metabolismo , Mioblastos/ultraestructura , Factores Reguladores Miogénicos/metabolismo , ARN Interferente Pequeño/metabolismo , Regeneración , Células Madre/citología , Células Madre/metabolismo , Células Madre/ultraestructura , Análisis de Supervivencia , Factores de Transcripción/genética
17.
J Appl Physiol (1985) ; 130(4): 1122-1132, 2021 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-33539264

RESUMEN

Olfactory receptor (Olfr) 78 is expressed in the carotid bodies (CB) and participates in CB responses to acute hypoxia. Olfr78 is also expressed in the kidney, which is a major site of erythropoietin (Epo) production by hypoxia. The present study examined the role of Olfr78 in cardiorespiratory and renal Epo gene responses to hypobaric hypoxia (HH), simulating low O2 condition experienced at high altitude. Studies were performed on adult, male wild-type (WT) and Olfr78 null mice treated with 18 h of HH (0.4 atmospheres). HH-treated WT mice exhibited increased baseline breathing, augmented hypoxic ventilatory response, elevated blood pressure, and plasma norepinephrine (NE) levels. These effects were associated with increased baseline CB sensory nerve activity and augmented CB sensory nerve response to subsequent acute hypoxia. In contrast, HH-treated Olfr78 null mice showed an absence of cardiorespiratory and CB sensory nerve responses, suggesting impaired CB-dependent cardiorespiratory adaptations. WT mice responded to HH with activation of the renal Epo gene expression and elevated plasma Epo levels, and these effects were attenuated or absent in Olfr78 null mice. The attenuated Epo activation by HH was accompanied with markedly reduced hypoxia-inducible factor (HIF)-2α protein and reduced activation of HIF-2 target gene Sod-1 in Olfr78 null mice, suggesting impaired transcriptional activation of HIF-2 contributes to attenuated Epo responses to HH. These results demonstrate a hitherto uncharacterized role for Olfr78 in cardiorespiratory adaptations and renal Epo gene activation by HH such as that experienced at high altitude.NEW & NOTEWORTHY In this study, we delineated a previously uncharacterized role for olfactory receptor 78 (Olfr78), a G-protein-coupled receptor in regulation of erythropoietin and cardiorespiratory responses to hypobaric hypoxia. Our results demonstrate a striking loss of cardiorespiratory adaptations accompanied by an equally striking absence of carotid body sensory nerve responses to hypobaric hypoxia in Olfr78 null mice. We further demonstrate a hitherto uncharacterized role for Olfr78 in erythropoietin activation by hypobaric hypoxia.


Asunto(s)
Cuerpo Carotídeo , Eritropoyetina , Receptores Odorantes , Animales , Hipoxia , Masculino , Ratones , Respiración
18.
Biol Reprod ; 82(6): 1227-36, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20181618

RESUMEN

Spermatogenesis, a process involving the differentiation of spermatogonial stem cells into mature spermatozoa, takes place throughout masculine life. A complex system in the testis, including endocrine signaling, physical interactions between germ and somatic cells, spermatocyte meiosis, and timely release of spermatozoa, controls this cycle. We demonstrate herein that decreased O(2) levels and Epas1 activation are critical components of spermatogenesis. Postnatal Epas1 ablation leads to male infertility, with reduced testis size and weight. While immature spermatogonia and spermatocytes are present in Epas1(Delta/Delta) testes, spermatid and spermatozoan numbers are dramatically reduced. This is not due to germ cell-intrinsic defects. Rather, Epas(Delta/Delta) Sertoli cells exhibit decreased ability to form tight junctions, thereby disrupting the blood-testis barrier necessary for proper spermatogenesis. Reduced numbers of tight junction complexes are due to decreased expression of multiple genes encoding tight junction proteins, including TJP1 (ZO1), TJP2 (ZO2), and occludin. Furthermore, Epas1(Delta/Delta) testes exhibit disrupted basement membranes surrounding the seminiferous tubules, causing the premature release of incompletely differentiated germ cells. We conclude that low O(2) levels in the male gonad regulate germ cell homeostasis in this organ via EPAS1.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Barrera Hematotesticular/metabolismo , Espermatogénesis , Espermatogonias/metabolismo , Testículo/metabolismo , Animales , Membrana Basal/química , Membrana Basal/patología , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/deficiencia , Barrera Hematotesticular/patología , Masculino , Proteínas de la Membrana/análisis , Ratones , Ocludina , Tamaño de los Órganos , Fosfoproteínas/análisis , Túbulos Seminíferos/química , Túbulos Seminíferos/patología , Células de Sertoli/química , Células de Sertoli/patología , Espermátides/metabolismo , Espermátides/patología , Espermatogonias/patología , Testículo/patología , Uniones Estrechas/química , Uniones Estrechas/patología , Proteína de la Zonula Occludens-1 , Proteína de la Zonula Occludens-2
19.
Circ Res ; 103(4): 388-95, 2008 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-18635823

RESUMEN

The ductus arteriosus is a specialized blood vessel containing highly differentiated and contractile vascular smooth muscle, derived largely from neural crest cells, that is essential for fetal life but typically closes after birth. Impaired development of the ductus arteriosus or disruption of signaling pathways that initiate postnatal closure can result in persistent patency of the ductus arteriosus, the third most common congenital heart defect. We found that Tfap2beta, a transcription factor associated with patent ductus arteriosus in humans, was uniquely expressed in mouse ductal smooth muscle. Endothelin-1 and the hypoxia-induced transcription factor, Hif2alpha were also highly enriched in ductal smooth muscle at embryonic day 13.5 and were dependent on Tfap2beta for their expression in this domain. Hif2alpha functioned as a negative regulator of Tfap2beta-induced transcription by disrupting protein-DNA interactions, suggesting a negative feedback loop regulating Tfap2beta activity. Our data indicate that Tfap2beta, Et-1, and Hif2alpha act in a transcriptional network during ductal smooth muscle development and that disruption of this pathway may contribute to patent ductus arteriosus by affecting the development of smooth muscle within the ductus arteriosus.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Conducto Arterioso Permeable/metabolismo , Conducto Arterial/metabolismo , Endotelina-1/metabolismo , Factor de Transcripción AP-2/metabolismo , Transcripción Genética/fisiología , Animales , Diferenciación Celular , Modelos Animales de Enfermedad , Conducto Arterial/fisiología , Conducto Arterioso Permeable/etiología , Conducto Arterioso Permeable/fisiopatología , Feto/metabolismo , Humanos , Ratones , Ratones Noqueados , Músculo Liso Vascular/citología , Músculo Liso Vascular/embriología , Músculo Liso Vascular/metabolismo , Mutación , Transducción de Señal/fisiología , Factor de Transcripción AP-2/genética , Transfección
20.
Circ Res ; 102(9): 1075-81, 2008 May 09.
Artículo en Inglés | MEDLINE | ID: mdl-18356544

RESUMEN

Stem and progenitor cell populations occupy a specialized niche and are consequently exposed to hypoxic as well as oxidative stresses. We have previously established that the multidrug resistance protein Abcg2 is the molecular determinant of the side population (SP) progenitor cell population. We observed that the cardiac SP cells increase in number more than 3-fold within 3 days of injury. Transcriptome analysis of the SP cells isolated from the injured adult murine heart reveals increased expression of cytoprotective transcripts. Overexpression of Abcg2 results in an increased ability to consume hydrogen peroxide and is associated with increased levels of alpha-glutathione reductase protein expression. Importantly, overexpression of Abcg2 also conferred a cell survival benefit following exposure to hydrogen peroxide. To further examine the molecular regulation of the Abcg2 gene, we demonstrated that hypoxia-inducible factor (HIF)-2alpha binds an evolutionary conserved HIF-2alpha response element in the murine Abcg2 promoter. Transcriptional assays reveal a dose-dependent activation of Abcg2 expression by HIF-2alpha. These results support the hypothesis that Abcg2 is a direct downstream target of HIF-2alpha which functions with other factors to initiate a cytoprotective program for this progenitor SP cell population that resides in the adult heart.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/metabolismo , Células Madre Adultas/metabolismo , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Citoprotección , Células Madre Embrionarias/metabolismo , Lesiones Cardíacas/prevención & control , Activación Transcripcional , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2 , Transportadoras de Casetes de Unión a ATP/genética , Células Madre Adultas/enzimología , Células Madre Adultas/patología , Animales , Línea Celular , Proliferación Celular , Células Cultivadas , Frío/efectos adversos , Citoprotección/genética , Modelos Animales de Enfermedad , Células Madre Embrionarias/patología , Perfilación de la Expresión Génica/métodos , Glutatión Reductasa/metabolismo , Lesiones Cardíacas/etiología , Lesiones Cardíacas/genética , Lesiones Cardíacas/metabolismo , Lesiones Cardíacas/patología , Peróxido de Hidrógeno/metabolismo , Ratones , Análisis de Secuencia por Matrices de Oligonucleótidos , Estrés Oxidativo , Regiones Promotoras Genéticas , Unión Proteica , ARN Mensajero/metabolismo , Transducción de Señal , Factores de Tiempo , Transcripción Genética , Transfección , Regulación hacia Arriba
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA