Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Brief Bioinform ; 22(4)2021 07 20.
Artículo en Inglés | MEDLINE | ID: mdl-33367491

RESUMEN

The human cerebral cortex undergoes profound structural and functional dynamic variations across the lifespan, whereas the underlying molecular mechanisms remain unclear. Here, with a novel method transcriptome-connectome correlation analysis (TCA), which integrates the brain functional magnetic resonance images and region-specific transcriptomes, we identify age-specific cortex (ASC) gene signatures for adolescence, early adulthood and late adulthood. The ASC gene signatures are significantly correlated with the cortical thickness (P-value <2.00e-3) and myelination (P-value <1.00e-3), two key brain structural features that vary in accordance with brain development. In addition to the molecular underpinning of age-related brain functions, the ASC gene signatures allow delineation of the molecular mechanisms of neuropsychiatric disorders, such as the regulation between ARNT2 and its target gene ETF1 involved in Schizophrenia. We further validate the ASC gene signatures with published gene sets associated with the adult cortex, and confirm the robustness of TCA on other brain image datasets. Availability: All scripts are written in R. Scripts for the TCA method and related statistics result can be freely accessed at https://github.com/Soulnature/TCA. Additional data related to this paper may be requested from the authors.


Asunto(s)
Envejecimiento/metabolismo , Translocador Nuclear del Receptor de Aril Hidrocarburo/biosíntesis , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/biosíntesis , Corteza Cerebral/metabolismo , Factores de Terminación de Péptidos/biosíntesis , Esquizofrenia/metabolismo , Transcriptoma , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Niño , Femenino , Humanos , Masculino , Persona de Mediana Edad
2.
Biochim Biophys Acta Mol Basis Dis ; 1863(6): 1640-1653, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28341552

RESUMEN

Coronary artery disease (CAD) is the leading cause of death worldwide. GWAS have identified >50 genomic loci for CAD, including ADTRP and MIA3/TANGO1. However, it is important to determine whether the GWAS genes form a molecular network. In this study, we have uncovered a novel molecular network between ADTRP and MIA3/TANGO1 for the pathogenesis of CAD. We showed that knockdown of ADTRP expression markedly down-regulated expression of MIA3/TANGO1. Mechanistically, ADTRP positively regulates expression of PIK3R3 encoding the regulatory subunit 3 of PI3K, which leads to activation of AKT, resulting in up-regulation of MIA3/TANGO1. Both ADTRP and MIA3/TANGO1 are involved in endothelial cell (EC) functions relevant to atherosclerosis. Knockdown of ADTRP expression by siRNA promoted oxidized-LDL-mediated monocyte adhesion to ECs and transendothelial migration of monocytes, inhibited EC proliferation and migration, and increased apoptosis, which was reversed by expression of constitutively active AKT1 and MIA3/TANGO1 overexpression, while the over-expression of ADTRP in ECs blunted these processes. Knockdown of MIA3/TANGO1 expression also promoted monocyte adhesion to ECs and transendothelial migration of monocytes, and vice versa for overexpression of MIA3/TANGO1. We found that ADTRP negatively regulates the levels of collagen VII and ApoB in HepG2 and endothelial cells, which are downstream regulatory targets of MIA3/TANGOI. In conclusion, we have uncovered a novel molecular signaling pathway for the pathogenesis of CAD, which involves a novel gene-gene regulatory network. We show that ADTRP positively regulates PIK3R3 expression, which leads to activation of AKT and up-regulation of MIA3/TANGO1, thereby regulating endothelial cell functions directly relevant to atherosclerosis.


Asunto(s)
Translocador Nuclear del Receptor de Aril Hidrocarburo/biosíntesis , Aterosclerosis/metabolismo , Enfermedad de la Arteria Coronaria/metabolismo , Redes Reguladoras de Genes , Predisposición Genética a la Enfermedad , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Proteínas de la Membrana/metabolismo , Transducción de Señal , Translocador Nuclear del Receptor de Aril Hidrocarburo/genética , Aterosclerosis/genética , Aterosclerosis/patología , Enfermedad de la Arteria Coronaria/genética , Enfermedad de la Arteria Coronaria/patología , Regulación de la Expresión Génica/genética , Estudio de Asociación del Genoma Completo , Células Hep G2 , Células Endoteliales de la Vena Umbilical Humana/patología , Humanos , Proteínas de la Membrana/genética , Fosfatidilinositol 3-Quinasas/biosíntesis , Fosfatidilinositol 3-Quinasas/genética
3.
Drug Metab Dispos ; 45(2): 118-129, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27856527

RESUMEN

The aryl hydrocarbon receptor (AHR) nuclear translocator (ARNT), as the AHR's heterodimerization partner, and NADPH-cytochrome P450 oxidoreductase (POR), as the key electron donor for all microsomal P450s, are independent and indispensable components in the adaptive and toxic responses to polycyclic aromatic hydrocarbons. Expression of both ARNT and POR in rat liver is induced by dexamethasone (DEX), a synthetic glucocorticoid known to activate both the glucocorticoid receptor (GR) and the pregnane X receptor (PXR). To better understand the role of GR and PXR in the in vivo DEX induction of rat hepatic ARNT and POR at the mRNA and protein levels, we studied the following: 1) the effects of DEX doses that activate GR (≥0.1 mg/kg) or PXR (≥10 mg/kg); 2) responses produced by GR- and PXR-selective agonists; 3) the impact of GR antagonism on DEX's inducing effects; and 4) whether biologic responses to DEX are altered in PXR-knockout rats. Our findings are consistent with a role for GR as a key mediator of the induction of rat hepatic ARNT expression by glucocorticoids; a role for PXR in the modulation of ARNT protein levels could not be excluded. Although GR activation may contribute to POR mRNA induction, regulation of POR expression and function by DEX is primarily PXR-mediated. This work suggests that the hepatic expression and function of ARNT and POR may be modulated by exposure to exogenous PXR activators and/or conditions that alter glucocorticoid levels such as stress, steroidal therapies, and diseases of excess or deficiency.


Asunto(s)
Translocador Nuclear del Receptor de Aril Hidrocarburo/biosíntesis , Dexametasona/farmacología , Microsomas Hepáticos/efectos de los fármacos , NADPH-Ferrihemoproteína Reductasa/biosíntesis , Receptores de Glucocorticoides/fisiología , Receptores de Esteroides/fisiología , Animales , Relación Dosis-Respuesta a Droga , Inducción Enzimática , Técnicas de Inactivación de Genes , Masculino , Microsomas Hepáticos/metabolismo , Receptor X de Pregnano , Ratas Endogámicas F344 , Ratas Sprague-Dawley , Receptores de Glucocorticoides/genética , Receptores de Esteroides/genética
4.
IUBMB Life ; 68(2): 122-35, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26707719

RESUMEN

The aryl hydrocarbon receptor nuclear translocator (ARNT/HIF1-ß) is an obligatory transcriptional partner of the aryl hydrocarbon receptor (AHR) and hypoxia-inducible factor-1α (HIF-1α). It has a basic helix-loop-helix domain that belongs to period-ARNT-single-minded (PAS) protein family. PAS proteins act as heterodimeric transcription factors with ARNT being master dimerization partner. The ARNT-HIF-1α complex is an important transcriptional regulator of the hypoxic response of the tumor cells. Previous studies have reported two transcript variants of the gene produced by alternative splicing in mouse. One transcript variant contains all 22 exons while the other variant lacks exon-E5. In our study, using combinatorial approach comprising bioinformatics tools and molecular biology techniques involving RT-PCR, semi-nested PCR, sequencing and qPCR, we have identified three novel transcript variants of Arnt gene in mouse. All three new transcripts arise as a result of alternative splicing of newly identified exons with exon-E2, replacing reported exon-E1. These transcripts encode for three protein isofoms having different N-termini. The expression of these transcripts was found to be different in different tissues of adult mice. In silico analysis of the upstream region of the new exons revealed three distinct promoter regions designated as PA, PB and PC present upstream of newly identified exons. These promoters possess potential signature sequences for common as well as different transcription factors suggesting complex regulation of Arnt gene. In silico post translational studies of the conceptually translated amino acid sequences of these transcripts show similarity in some of the properties while differ in others. The diversity at N-termini of protein isoforms suggests the possibility of forming different complexes in different tissues and may also be important for unique interactions with partner molecules.


Asunto(s)
Empalme Alternativo/genética , Translocador Nuclear del Receptor de Aril Hidrocarburo/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Isoformas de Proteínas/genética , Animales , Translocador Nuclear del Receptor de Aril Hidrocarburo/biosíntesis , Translocador Nuclear del Receptor de Aril Hidrocarburo/metabolismo , Hipoxia de la Célula/genética , Exones , Regulación de la Expresión Génica , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Ratones , Regiones Promotoras Genéticas , Isoformas de Proteínas/biosíntesis
5.
J Immunol ; 192(4): 1661-70, 2014 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-24403534

RESUMEN

The cytokine IL-10 has an important role in limiting inflammation in many settings, including toxoplasmosis. In the present studies, an IL-10 reporter mouse was used to identify the sources of this cytokine following challenge with Toxoplasma gondii. During infection, multiple cell types expressed the IL-10 reporter but NK cells were a major early source of this cytokine. These IL-10 reporter(+) NK cells expressed high levels of the IL-12 target genes T-bet, KLRG1, and IFN-γ, and IL-12 depletion abrogated reporter expression. However, IL-12 signaling alone was not sufficient to promote NK cell IL-10, and activation of the aryl hydrocarbon receptor (AHR) was also required for maximal IL-10 production. NK cells basally expressed the AHR, relevant chaperone proteins, and the AHR nuclear translocator, which heterodimerizes with the AHR to form a competent transcription factor. In vitro studies revealed that IL-12 stimulation increased NK cell AHR levels, and the AHR and AHR nuclear translocator were required for optimal production of IL-10. Additionally, NK cells isolated from T. gondii-infected Ahr(-/-) mice had impaired expression of IL-10, which was associated with increased resistance to this infection. Taken together, these data identify the AHR as a critical cofactor involved in NK cell production of IL-10.


Asunto(s)
Interleucina-10/biosíntesis , Interleucina-12/metabolismo , Células Asesinas Activadas por Linfocinas/metabolismo , Receptores de Hidrocarburo de Aril/metabolismo , Toxoplasma/inmunología , Animales , Translocador Nuclear del Receptor de Aril Hidrocarburo/biosíntesis , Translocador Nuclear del Receptor de Aril Hidrocarburo/metabolismo , Dimerización , Genes Reporteros , Inflamación/inmunología , Interferón gamma/biosíntesis , Lectinas Tipo C , Activación de Linfocitos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos CBA , Ratones Noqueados , Receptores de Hidrocarburo de Aril/deficiencia , Receptores de Hidrocarburo de Aril/genética , Receptores Inmunológicos/biosíntesis , Transducción de Señal/inmunología , Proteínas de Dominio T Box/biosíntesis , Toxoplasmosis Animal/inmunología
6.
Cell Physiol Biochem ; 35(3): 841-57, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25632940

RESUMEN

BACKGROUND/AIMS: Multipotent stem/stromal cells (MSC) are considered promising for cartilage tissue engineering. However, chondrogenic differentiation of MSC can ultimately lead to the formation of hypertrophic chondrocytes responsible for the calcification of cartilage. To prevent the production of this calcified matrix at the articular site, the late hypertrophic differentiation of MSCs must be carefully controlled. Given that articular cartilage is avascular, we hypothesized that in addition to its stimulatory role in the early differentiation of chondrogenic cells, hypoxia may prevent their late hypertrophic conversion. METHODS: Early and late chondrogenic differentiation were evaluated using human adipose MSC and murine ATDC5 cells cultured under either normoxic (21%O2) or hypoxic (5%O2) conditions. To investigate the effect of hypoxia on late chondrogenic differentiation, the transcriptional activity of hypoxia-inducible factor-1alpha (HIF-1α) and HIF-2α were evaluated using the NoShift DNA-binding assay and through modulation of their activity (chemical inhibitor, RNA interference). Results : Our data demonstrate that low oxygen tension not only stimulates the early chondrogenic commitment of two complementary models of chondrogenic cells, but also inhibits their hypertrophic differentiation. Conclusion : These results suggest that hypoxia can be used as an instrumental tool to prevent the formation of a calcified matrix in MSC-based cartilage tissue engineering.


Asunto(s)
Cartílago Articular/crecimiento & desarrollo , Diferenciación Celular/genética , Células Madre Mesenquimatosas/citología , Ingeniería de Tejidos , Animales , Translocador Nuclear del Receptor de Aril Hidrocarburo/biosíntesis , Calcinosis/genética , Calcinosis/patología , Cartílago Articular/citología , Hipoxia de la Célula , Condrocitos/citología , Condrogénesis/genética , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/biosíntesis , Células Madre Mesenquimatosas/metabolismo , Ratones , Oxígeno/metabolismo
7.
Nucleic Acids Res ; 41(11): 5626-38, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23599003

RESUMEN

Basic helix-loop-helix/Per-Arnt-Sim (bHLH/PAS) transcription factors function broadly in development, homeostasis and stress response. Active bHLH/PAS heterodimers consist of a ubiquitous signal-regulated subunit (e.g., hypoxia-inducible factors, HIF-1α/2α/3α; the aryl hydrocarbon receptor, AhR) or tissue-restricted subunit (e.g., NPAS1/3/4, Single Minded 1/2), paired with a general partner protein, aryl hydrocarbon receptor nuclear translocator (Arnt or Arnt2). We have investigated regulation of the neuron-enriched Arnt paralogue, Arnt2. We find high Arnt/Arnt2 ratios in P19 embryonic carcinoma cells and ES cells are dramatically reversed to high Arnt2/Arnt on neuronal differentiation. mRNA half-lives of Arnt and Arnt2 remain similar in both parent and neuronal differentiated cells. The GC-rich Arnt2 promoter, while heavily methylated in Arnt only expressing hepatoma cells, is methylation free in P19 and ES cells, where it is bivalent with respect to active H3K4me3 and repressive H3K27me3 histone marks. Typical of a 'transcription poised' developmental gene, H3K27me3 repressive marks are removed from Arnt2 during neuronal differentiation. Our data are consistent with a switch to predominant Arnt2 expression in neurons to allow specific functions of neuronal bHLH/PAS factors and/or to avoid neuronal bHLH/PAS factors from interfering with AhR/Arnt signalling.


Asunto(s)
Translocador Nuclear del Receptor de Aril Hidrocarburo/genética , Translocador Nuclear del Receptor de Aril Hidrocarburo/metabolismo , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Epigénesis Genética , Neurogénesis/genética , Neuronas/metabolismo , Animales , Translocador Nuclear del Receptor de Aril Hidrocarburo/biosíntesis , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/biosíntesis , Línea Celular Tumoral , Metilación de ADN , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Ratones , Regiones Promotoras Genéticas , Transcripción Genética
8.
J Cell Sci ; 125(Pt 14): 3320-32, 2012 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-22505606

RESUMEN

Previously we showed that spatial and developmental modulation of ARNT (HIF1ß) expression in mouse epidermis is essential for maintenance of keratinocyte differentiation, proper formation of the barrier and normal desquamation. Here, using lentiviral suppression or induction of ARNT in TERT-immortalized (N-TERT) and HaCaT cells we assessed the nature and mechanisms of ARNT involvement in control of differentiation in human epidermal keratinocytes. ARNT depletion did not affect the levels of basal keratins K5 and K14, but significantly induced expression of several key differentiation markers (an effect abolished by EGF supplementation). Furthermore, ARNT deficiency resulted in the downregulation of amphiregulin (AREG) - the most highly expressed EGFR ligand in human keratinocytes - whereas upregulation of ARNT showed the opposite. In ARNT-deficient monolayer cultures and 3D epidermal equivalents, the downregulation of AREG was concurrent with a decline of EGFR and ERK1/2 phosphorylation. TSA, a potent suppressor of HDAC activity, abolished the effects of ARNT deficiency, implying a role for HDACs in ARNT-dependent modulation of the AREG-EGFR pathway and downstream epidermal genes. Total HDAC activity was significantly increased in ARNT-depleted cells and decreased with ARNT overexpression. ARNT-dependent shifts in HDAC activity were specifically attributed to significant changes in the levels of HDAC1, HDAC2 and HDAC3 proteins (but not mRNA) in both monolayer and 3D cultures. Collectively, our results suggest that ARNT controls AREG expression and the downstream EGFR-ERK pathway in keratinocytes, at least in part, by modulating HDAC activity. This novel regulatory pathway targeting advanced stages of epidermal differentiation might have important implications for skin pathology such as psoriasis, atopic dermatitis and cancer.


Asunto(s)
Translocador Nuclear del Receptor de Aril Hidrocarburo/metabolismo , Receptores ErbB/metabolismo , Regulación de la Expresión Génica , Histona Desacetilasas/metabolismo , Queratinocitos/citología , Queratinocitos/metabolismo , Animales , Translocador Nuclear del Receptor de Aril Hidrocarburo/biosíntesis , Translocador Nuclear del Receptor de Aril Hidrocarburo/deficiencia , Translocador Nuclear del Receptor de Aril Hidrocarburo/genética , Diferenciación Celular/genética , Línea Celular , Células Cultivadas , Células Epidérmicas , Receptores ErbB/genética , Expresión Génica , Humanos , Queratinas/metabolismo , Ratones , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Fosforilación , Transducción de Señal
9.
Am J Pathol ; 182(6): 2391-406, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23583651

RESUMEN

Pulmonary arterial hypertension (PAH) is a chronic and progressive disease characterized by pulmonary vasculopathy with elevation of pulmonary artery pressure, often culminating in right ventricular failure. GATA-6, a member of the GATA family of zinc-finger transcription factors, is highly expressed in quiescent vasculature and is frequently lost during vascular injury. We hypothesized that endothelial GATA-6 may play a critical role in the molecular mechanisms underlying endothelial cell (EC) dysfunction in PAH. Here we report that GATA-6 is markedly reduced in pulmonary ECs lining both occluded and nonoccluded vessels in patients with idiopathic and systemic sclerosis-associated PAH. GATA-6 transcripts are also rapidly decreased in rodent PAH models. Endothelial GATA-6 is a direct transcriptional regulator of genes controlling vascular tone [endothelin-1, endothelin-1 receptor type A, and endothelial nitric oxide synthase (eNOS)], pro-inflammatory genes, CX3CL1 (fractalkine), 5-lipoxygenease-activating protein, and markers of vascular remodeling, including PAI-1 and RhoB. Mice with the genetic deletion of GATA-6 in ECs (Gata6-KO) spontaneously develop elevated pulmonary artery pressure and increased vessel muscularization, and these features are further exacerbated in response to hypoxia. Furthermore, innate immune cells including macrophages (CD11b(+)/F4/80(+)), granulocytes (Ly6G(+)/CD45(+)), and dendritic cells (CD11b(+)/CD11c(+)) are significantly increased in normoxic Gata6-KO mice. Together, our findings suggest a critical role of endothelial GATA-6 deficiency in development and disease progression in PAH.


Asunto(s)
Endotelio Vascular/metabolismo , Factor de Transcripción GATA6/deficiencia , Hipertensión Pulmonar/metabolismo , Adaptación Fisiológica/fisiología , Animales , Translocador Nuclear del Receptor de Aril Hidrocarburo/biosíntesis , Translocador Nuclear del Receptor de Aril Hidrocarburo/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/biosíntesis , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Estudios de Casos y Controles , Enfermedad Crónica , Progresión de la Enfermedad , Regulación hacia Abajo/fisiología , Células Endoteliales/fisiología , Hipertensión Pulmonar Primaria Familiar , Factor de Transcripción GATA6/metabolismo , Factor de Transcripción GATA6/fisiología , Regulación de la Expresión Génica/fisiología , Humanos , Hipertensión Pulmonar/etiología , Hipertrofia Ventricular Derecha/metabolismo , Hipertrofia Ventricular Derecha/fisiopatología , Hipoxia/complicaciones , Pulmón/irrigación sanguínea , Masculino , Ratones , Ratones Noqueados , Neumonía/metabolismo , ARN Mensajero/genética , Ratas , Ratas Sprague-Dawley , Esclerodermia Sistémica/complicaciones
10.
Mol Pharmacol ; 84(4): 591-602, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23907215

RESUMEN

The identification of molecular pathways in cancer cells is important for understanding the cells' underlying biology and for designing effective cancer therapies. We demonstrate that the expression of aryl hydrocarbon receptor nuclear translocator (ARNT) is critical during the development of cisplatin resistance. The reduced expression of ARNT was correlated with cisplatin-induced cell death in drug-sensitive cells. In addition, suppression of ARNT reversed the characteristics of cisplatin-resistant cells, making these cells cisplatin-sensitive, and significantly enhanced caspase-3 activation, DNA fragmentation, and apoptosis. The inhibition of colony formation, regulated by cisplatin, was more significant in ARNT-knockdown cells than in parental cells. In a xenograft analysis of severe combined immunodeficiency mice, cisplatin also efficiently inhibited ARNT-deficient c4 tumors but not ARNT-containing vT2 tumor formation. Furthermore, the downregulation of multidrug resistance 1 (MDR1) expression and retention of drugs in cells caused by suppression of ARNT, resulting in the resensitization of drug-resistant cells to cisplatin, was observed. When overexpressed, ARNT interacted with Sp1 to enhance the expression of MDR1 through Sp1-binding sites on the MDR1 promoter, resulting in a reversal of the effect of cisplatin on cell death. In addition, ARNT-induced MDR1 expression was inhibited in Sp1-knockdown cells. These results reveal previously unrecognized, multifaceted functions of ARNT in establishing the drug-resistant properties of cancer cells by the upregulation of MDR1, highlighting ARNT's potential as a therapeutic target in an important subset of cancers.


Asunto(s)
Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/biosíntesis , Translocador Nuclear del Receptor de Aril Hidrocarburo/biosíntesis , Cisplatino/farmacología , Resistencia a Antineoplásicos/genética , Regulación Neoplásica de la Expresión Génica , Regulación hacia Arriba/genética , Subfamilia B de Transportador de Casetes de Unión a ATP , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/genética , Animales , Translocador Nuclear del Receptor de Aril Hidrocarburo/genética , Resistencia a Antineoplásicos/efectos de los fármacos , Femenino , Células HeLa , Humanos , Ratones , Ratones SCID , Regulación hacia Arriba/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto/métodos
11.
Biochem Biophys Res Commun ; 434(1): 166-72, 2013 Apr 26.
Artículo en Inglés | MEDLINE | ID: mdl-23541582

RESUMEN

Solid tumors include hypoxic areas due to excessive cell proliferation. Adaptation to low oxygen levels is mediated by the hypoxia-inducible factor (HIF) pathway promoting invasion, metastasis, metabolic alterations, chemo-resistance and angiogenesis. The transcription factor HIF-1, the major player within this pathway consists of HIF-1α and HIF-1ß. The alpha subunit is continuously degraded under normoxia and becomes stabilized under reduced oxygen supply. In contrast, HIF-1ß is generally regarded as constitutively expressed and being present in excess within the cell. However, there is evidence that the expression of this subunit is more complex. The aim of this study was to investigate the role of HIF-1ß in human melanoma cells. Among a panel of five different cell lines, in 518A2 cells exposed to the hypoxia-mimetic cobalt chloride HIF-1ß was rapidly elevated on protein level. Knockdown experiments performed under cobalt chloride-exposure and hypoxia revealed that this effect was mediated by HIF-1α. The non-canonical relationship between these subunits was further confirmed by pharmacologic inhibition of HIF-1α and by expression of a dominant-negative HIF mutant. Overexpression of HIF-1α showed a time delay in HIF-1ß induction, thus arguing for HIF-1ß de novo synthesis rather than protein stabilization by heterodimerization. A Hen's egg test-chorioallantoic membrane model of angiogenesis and invasion indicated a local expression of HIF-1ß and implies a biological relevance of these findings. In summary, this study demonstrates the HIF-1α-dependent regulation of HIF-1ß under hypoxic conditions for the first time. The results indicate a novel cell specific mechanism which might prevent HIF-1ß to become a limiting factor.


Asunto(s)
Translocador Nuclear del Receptor de Aril Hidrocarburo/biosíntesis , Subunidad alfa del Factor 1 Inducible por Hipoxia/fisiología , Melanoma/metabolismo , Melanoma/patología , Regulación hacia Arriba/fisiología , Animales , Translocador Nuclear del Receptor de Aril Hidrocarburo/antagonistas & inhibidores , Translocador Nuclear del Receptor de Aril Hidrocarburo/genética , Hipoxia de la Célula/efectos de los fármacos , Hipoxia de la Célula/genética , Hipoxia de la Célula/fisiología , Línea Celular Tumoral , Embrión de Pollo , Cobalto/farmacología , Técnicas de Silenciamiento del Gen , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/antagonistas & inhibidores , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Melanoma/fisiopatología , Regulación hacia Arriba/efectos de los fármacos
12.
Toxicol Appl Pharmacol ; 264(2): 212-21, 2012 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-22917661

RESUMEN

Fibroblast growth factor-21 (FGF21) is a potential metabolic regulator with multiple beneficial effects on metabolic diseases. FGF21 is mainly expressed in the liver, but is also found in other tissues including the intestine, which expresses ß-klotho abundantly. The intestine is a unique organ that operates in a physiologically hypoxic environment, and is responsible for the fat absorption processes including triglyceride breakdown, re-synthesis and absorption into the portal circulation. In the present study, we investigated the effects of hypoxia and the chemical hypoxia inducer, cobalt chloride (CoCl(2)), on FGF21 expression in Caco-2 cells and the consequence of fat accumulation. Physical hypoxia (1% oxygen) and CoCl(2) treatment decreased both FGF21 mRNA and secreted protein levels. Gene silence and inhibition of hypoxia-inducible factor-α (HIFα) did not affect the reduction of FGF21 mRNA and protein levels by hypoxia. However, CoCl(2) administration caused a significant increase in oxidative stress. The addition of n-acetylcysteine (NAC) suppressed CoCl(2)-induced reactive oxygen species (ROS) formation and completely negated CoCl(2)-induced FGF21 loss. mRNA stability analysis demonstrated that the CoCl(2) administration caused a remarkable reduction in FGF21 mRNA stability. Furthermore, CoCl(2) increased intracellular triglyceride (TG) accumulation, along with a reduction in mRNA levels of lipid lipase, hormone sensitive lipase (HSL) and adipose triglyceride lipase (ATGL), and an increase of sterol regulatory element-binding protein-1c (SREBP1c) and stearoyl-coenzyme A (SCD1). Addition of both NAC and recombinant FGF21 significantly attenuated the CoCl(2)-induced TG accumulation. In conclusion, the decrease of FGF21 in Caco-2 cells by chemical hypoxia is independent of HIFα, but dependent on an oxidative stress-mediated mechanism. The regulation of FGF21 by hypoxia may contribute to intestinal lipid metabolism and absorption.


Asunto(s)
Cobalto/farmacología , Factores de Crecimiento de Fibroblastos/biosíntesis , Factor 1 Inducible por Hipoxia/fisiología , Estrés Oxidativo/efectos de los fármacos , Translocador Nuclear del Receptor de Aril Hidrocarburo/biosíntesis , Compuestos Azo , Western Blotting , Células CACO-2 , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Colorantes , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/biosíntesis , PPAR alfa/fisiología , PPAR gamma/fisiología , Interferencia de ARN , ARN Mensajero/biosíntesis , Reacción en Cadena en Tiempo Real de la Polimerasa , Superóxidos/metabolismo , Triglicéridos/metabolismo
13.
J Cell Biochem ; 112(3): 804-17, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21328454

RESUMEN

Cell therapy with bone marrow multipotential stromal cells (MSCs) represents a promising approach to promote wound healing and tissue regeneration. MSCs expanded in vitro lose early progenitors with differentiation and therapeutic potentials under normoxic condition, whereas hypoxic condition promotes MSC self-renewal through preserving colony forming early progenitors and maintaining undifferentiated phenotypes. Hypoxia inducible factor (HIF) pathway is a crucial signaling pathway activated in hypoxic condition. We evaluated the roles of HIFs in MSC differentiation, colony formation, and paracrine activity under hypoxic condition. Hypoxic condition reversibly decreased osteogenic and adipogenic differentiation. Decrease of osteogenic differentiation depended on HIF pathway; whereas decrease of adipogenic differentiation depended on the activation of unfolded protein response (UPR), but not HIFs. Hypoxia-mediated increase of MSC colony formation was not HIF-dependent also. Hypoxic exposure increased secretion of VEGF, HGF, and basic FGF in a HIF-dependent manner. These findings suggest that HIF has a limited, but pivotal role in enhancing MSC self-renewal and growth factor secretions under hypoxic condition.


Asunto(s)
Translocador Nuclear del Receptor de Aril Hidrocarburo/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Células Madre Mesenquimatosas/citología , Adipocitos/citología , Translocador Nuclear del Receptor de Aril Hidrocarburo/biosíntesis , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/biosíntesis , Técnicas de Cultivo de Célula , Diferenciación Celular/efectos de los fármacos , Hipoxia de la Célula , Genes Reporteros , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/biosíntesis , Osteocitos/citología , PPAR gamma/genética , PPAR gamma/metabolismo , Fenilbutiratos/farmacología , Interferencia de ARN , ARN Mensajero/metabolismo , Elementos de Respuesta , Respuesta de Proteína Desplegada
14.
Biochem Biophys Res Commun ; 381(1): 44-9, 2009 Mar 27.
Artículo en Inglés | MEDLINE | ID: mdl-19351592

RESUMEN

We have investigated the effect of benzo[a]pyrene (B[a]P), a carcinogen of tobacco smoke and an agonist for the aryl hydrocarbon receptor (AHR), on hypoxia-induced angiogenesis. Ischemia was induced by femoral artery ligation in wild-type and AHR-null mice, and the animals were subjected to oral administration of B[a]P (125 mg/kg) once a week. Exposure to B[a]P up-regulated the expression of metallothionein in the ischemic hindlimb and markedly inhibited ischemia-induced angiogenesis in wild-type mice. The amounts of interleukin-6 and of vascular endothelial growth factor (VEGF) mRNA in the ischemic hindlimb of wild-type mice were reduced by exposure to B[a]P. These various effects of B[a]P were markedly attenuated in AHR-null mice. Our observations suggest that the loss of the inhibitory effect of B[a]P on ischemia-induced angiogenesis apparent in AHR-null mice may be attributable to maintenance of interleukin-6 expression and consequent promotion of angiogenesis through up-regulation of VEGF expression.


Asunto(s)
Benzo(a)pireno/toxicidad , Carcinógenos/toxicidad , Isquemia/fisiopatología , Neovascularización Fisiológica/efectos de los fármacos , Receptores de Hidrocarburo de Aril/fisiología , Angiotensina I/biosíntesis , Animales , Translocador Nuclear del Receptor de Aril Hidrocarburo/biosíntesis , Citocromo P-450 CYP1A1/biosíntesis , Arteria Femoral/fisiopatología , Subunidad alfa del Factor 1 Inducible por Hipoxia/biosíntesis , Interleucina-6/biosíntesis , Extremidad Inferior/irrigación sanguínea , Masculino , Metalotioneína/biosíntesis , Ratones , Ratones Endogámicos , Receptores de Angiotensina/biosíntesis , Receptores de Hidrocarburo de Aril/agonistas , Receptores de Hidrocarburo de Aril/genética , Receptores de Factores de Crecimiento Endotelial Vascular/biosíntesis , Regulación hacia Arriba , Factor A de Crecimiento Endotelial Vascular/biosíntesis
15.
J Am Heart Assoc ; 7(11)2018 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-29858371

RESUMEN

BACKGROUND: Limb ischemia resulting from peripheral vascular disease is a common cause of morbidity. Vessel occlusion limits blood flow, creating a hypoxic environment that damages distal tissue, requiring therapeutic revascularization. Hypoxia-inducible factors (HIFs) are key transcriptional regulators of hypoxic vascular responses, including angiogenesis and arteriogenesis. Despite vascular smooth muscle cells' (VSMCs') importance in vessel integrity, little is known about their functional responses to hypoxia in peripheral vascular disease. This study investigated the role of VSMC HIF in mediating peripheral ischemic responses. METHODS AND RESULTS: We used ArntSMKO mice with smooth muscle-specific deletion of aryl hydrocarbon receptor nuclear translocator (ARNT, HIF-1ß), required for HIF transcriptional activity, in a femoral artery ligation model of peripheral vascular disease. ArntSMKO mice exhibit impaired perfusion recovery despite normal collateral vessel dilation and angiogenic capillary responses. Decreased blood flow manifests in extensive tissue damage and hypoxia in ligated limbs of ArntSMKO mice. Furthermore, loss of aryl hydrocarbon receptor nuclear translocator changes the proliferation, migration, and transcriptional profile of cultured VSMCs. ArntSMKO mice display disrupted VSMC morphologic features and wrapping around arterioles and increased vascular permeability linked to decreased local blood flow. CONCLUSIONS: Our data demonstrate that traditional vascular remodeling responses are insufficient to provide robust peripheral tissue reperfusion in ArntSMKO mice. In all, this study highlights HIF responses to hypoxia in arteriole VSMCs critical for the phenotypic and functional stability of vessels that aid in the recovery of blood flow in ischemic peripheral tissues.


Asunto(s)
Translocador Nuclear del Receptor de Aril Hidrocarburo/genética , Regulación de la Expresión Génica , Isquemia/genética , Extremidad Inferior/irrigación sanguínea , Músculo Liso Vascular/metabolismo , Enfermedades Vasculares Periféricas/genética , Animales , Translocador Nuclear del Receptor de Aril Hidrocarburo/biosíntesis , Western Blotting , Células Cultivadas , Modelos Animales de Enfermedad , Inmunohistoquímica , Isquemia/metabolismo , Isquemia/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía Confocal , Músculo Liso Vascular/patología , Enfermedades Vasculares Periféricas/metabolismo , Enfermedades Vasculares Periféricas/patología , ARN/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
16.
J Clin Invest ; 128(7): 3053-3070, 2018 07 02.
Artículo en Inglés | MEDLINE | ID: mdl-29664738

RESUMEN

Progression of chronic kidney disease associated with progressive fibrosis and impaired tubular epithelial regeneration is still an unmet biomedical challenge because, once chronic lesions have manifested, no effective therapies are available as of yet for clinical use. Prompted by various studies across multiple organs demonstrating that preconditioning regimens to induce endogenous regenerative mechanisms protect various organs from later incurring acute injuries, we here aimed to gain insights into the molecular mechanisms underlying successful protection and to explore whether such pathways could be utilized to inhibit progression of chronic organ injury. We identified a protective mechanism controlled by the transcription factor ARNT that effectively inhibits progression of chronic kidney injury by transcriptional induction of ALK3, the principal mediator of antifibrotic and proregenerative bone morphogenetic protein-signaling (BMP-signaling) responses. We further report that ARNT expression itself is controlled by the FKBP12/YY1 transcriptional repressor complex and that disruption of such FKBP12/YY1 complexes by picomolar FK506 at subimmunosuppressive doses increases ARNT expression, subsequently leading to homodimeric ARNT-induced ALK3 transcription. Direct targeting of FKBP12/YY1 with in vivo morpholino approaches or small molecule inhibitors, including GPI-1046, was equally effective for inducing ARNT expression, with subsequent activation of ALK3-dependent canonical BMP-signaling responses and attenuated chronic organ failure in models of chronic kidney disease, and also cardiac and liver injuries. In summary, we report an organ-protective mechanism that can be pharmacologically modulated by immunophilin ligands FK506 and GPI-1046 or therapeutically targeted by in vivo morpholino approaches.


Asunto(s)
Translocador Nuclear del Receptor de Aril Hidrocarburo/biosíntesis , Fallo Renal Crónico/tratamiento farmacológico , Fallo Renal Crónico/metabolismo , Animales , Receptores de Proteínas Morfogenéticas Óseas de Tipo 1/genética , Receptores de Proteínas Morfogenéticas Óseas de Tipo 1/metabolismo , Línea Celular , Progresión de la Enfermedad , Técnicas de Silenciamiento del Gen , Humanos , Riñón/efectos de los fármacos , Riñón/metabolismo , Riñón/patología , Fallo Renal Crónico/prevención & control , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Pirrolidinas/farmacología , Transducción de Señal/efectos de los fármacos , Tacrolimus/farmacología , Proteína 1A de Unión a Tacrolimus/antagonistas & inhibidores , Proteína 1A de Unión a Tacrolimus/metabolismo , Factor de Transcripción YY1/antagonistas & inhibidores , Factor de Transcripción YY1/genética , Factor de Transcripción YY1/metabolismo
17.
PLoS One ; 12(11): e0186262, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29117182

RESUMEN

Tissues of symbiotic Cnidarians are exposed to wide, rapid and daily variations of oxygen concentration. Indeed, during daytime, intracellular O2 concentration increases due to symbiont photosynthesis, while during night, respiration of both host cells and symbionts leads to intra-tissue hypoxia. The Hypoxia Inducible Factor 1 (HIF-1) is a heterodimeric transcription factor used for maintenance of oxygen homeostasis and adaptation to hypoxia. Here, we carried out a mechanistic study of the response to variations of O2 concentrations of the coral model Stylophora pistillata. In silico analysis showed that homologs of HIF-1 α (SpiHIF-1α) and HIF-1ß (SpiHIF-1ß) exist in coral. A specific SpiHIF-1 DNA binding on mammalian Hypoxia Response Element (HRE) sequences was shown in extracts from coral exposed to dark conditions. Then, we cloned the coral HIF-1α and ß genes and determined their expression and transcriptional activity. Although HIF-1α has an incomplete Oxygen-dependent Degradation Domain (ODD) relative to its human homolog, its protein level is increased under hypoxia when tested in mammalian cells. Moreover, co-transfection of SpiHIF-1α and ß in mammalian cells stimulated an artificial promoter containing HRE only in hypoxic conditions. This study shows the strong conservation of molecular mechanisms involved in adaptation to O2 concentration between Cnidarians and Mammals whose ancestors diverged about 1,200-1,500 million years ago.


Asunto(s)
Antozoos/genética , Translocador Nuclear del Receptor de Aril Hidrocarburo/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Oxígeno/metabolismo , Animales , Antozoos/fisiología , Translocador Nuclear del Receptor de Aril Hidrocarburo/biosíntesis , Clonación Molecular , Regulación de la Expresión Génica , Hipoxia/genética , Hipoxia/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/biosíntesis , Fotosíntesis/genética , Regiones Promotoras Genéticas , Elementos de Respuesta/genética , Simbiosis/genética
18.
Chemosphere ; 153: 198-204, 2016 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-27016815

RESUMEN

Litopenaeus vannamei has a high commercial value and is the primary cultured shellfish species globally. In this study, we have compared the hemolymph physiological responses between two L. vannamei strains under acute hypoxia. The results showed that hemocyanin concentration (HC) of strain A6410 was significantly higher than strain Zhengda; Total hemocyte counts (THC) decreased significantly in both strains under hypoxic stress (p < 0.05). We also investigated the temporal and spatial variations of hypoxia inducible factors 1 (HIF-1) by qRT-PCR. The results showed that hypoxia for 12 h increased the expression levels of HIF-1α in tissues of muscle and gill from the two strains (p < 0.05). In the hepatopancreas, the expression levels of HIF-1 increased significantly in strain Zhengda and decreased significantly in strain A6410 (p < 0.05). No significant changes of HIF-1 expression were detected in the same tissues between the two strains under hypoxia for 6 h (p > 0.05), but in the gills and hepatopancreas under hypoxia for 12 h (p < 0.05). Additionally, the expression level of HIF-1 was higher in the strain Zhengda than A6410 in the same tissue under hypoxia for 12 h. It was indicated that the hypoxic tolerance of Litopenaeus vannamei was closely correlated with the expression level of HIF-1, and the higher expression level of HIF-1 to hypoxia, the lower tolerance to hypoxia in the early stage of hypoxia. These results can help to better understand the molecular mechanisms of hypoxic tolerance and speed up the selective breeding process of hypoxia tolerance in L. vannamei.


Asunto(s)
Translocador Nuclear del Receptor de Aril Hidrocarburo/biosíntesis , Hipoxia de la Célula , Branquias/metabolismo , Hepatopáncreas/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/biosíntesis , Músculos/metabolismo , Penaeidae/metabolismo , Anaerobiosis , Animales , Hemocianinas , Hemocitos/metabolismo , Hemolinfa/metabolismo , Hemolinfa/fisiología
19.
Sci Rep ; 6: 21520, 2016 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-26861754

RESUMEN

Changes in cellular oxygen tension play important roles in physiological processes including development and pathological processes such as tumor promotion. The cellular adaptations to sustained hypoxia are mediated by hypoxia-inducible factors (HIFs) to regulate downstream target gene expression. With hypoxia, the stabilized HIF-α and aryl hydrocarbon receptor nuclear translocator (ARNT, also known as HIF-ß) heterodimer bind to hypoxia response elements (HREs) and regulate expression of target genes. Here, we report that WNT11 is induced by hypoxia in many cell types, and that transcription of WNT11 is regulated primarily by HIF-1α. We observed induced WNT11 expression in the hypoxic area of allograft tumors. In addition, in mice bearing orthotopic malignant gliomas, inhibition with bevacizumab of vascular endothelial growth factor, which is an important stimulus for angiogenesis, increased nuclear HIF-1α and HIF-2α, and expression of WNT11. Gain- and loss-of-function approaches revealed that WNT11 stimulates proliferation, migration and invasion of cancer-derived cells, and increases activity of matrix metalloproteinase (MMP)-2 and 9. Since tumor hypoxia has been proposed to increase tumor aggressiveness, these data suggest WNT11 as a possible target for cancer therapies, especially for tumors treated with antiangiogenic therapy.


Asunto(s)
Hipoxia de la Célula/fisiología , Movimiento Celular/genética , Proliferación Celular/genética , Glioma/patología , Subunidad alfa del Factor 1 Inducible por Hipoxia/biosíntesis , Invasividad Neoplásica/genética , Proteínas Wnt/biosíntesis , Inhibidores de la Angiogénesis/farmacología , Animales , Translocador Nuclear del Receptor de Aril Hidrocarburo/biosíntesis , Bevacizumab/farmacología , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica , Células HeLa , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neovascularización Patológica/tratamiento farmacológico , Oxígeno/metabolismo , Biosíntesis de Proteínas/genética , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau/genética , Proteínas Wnt/genética
20.
Mol Med Rep ; 14(5): 4383-4390, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27667582

RESUMEN

Pulmonary arterial hypertension (PAH) is characterized by endothelial dysfunction and structural remodeling of the pulmonary vasculature, mediated initially by reduced oxygen availability in the lungs. Hypoxia inducible factor (HIF), consisting of the functional subunit, HIF­1α, and the constitutively expressed HIF­1ß, is involved in the pathological processes associated with hypoxia. In the current study, the sequences of cDNAs and amino acids of HIF were characterized and analyzed using online bioinformatics tools. To further evaluate whether HIF accounts for the occurrence of PAH, the present study determine the expression and phosphorylation levels of HIF and its associated pathways, including extracellular signal­regulated kinase (Erk)1/2 and phosphoinositide 3­kinase (PI3K)/Akt, in the lungs of patients with PAH by reverse transcription­quantitative polymerase chain reaction and western blotting. The mRNA expression levels of PI3K, Erk2, and HIF­1α in the patients with PAH were significantly higher, compared with those in the control group, by 3.6­fold (P<0.01), 4.06­fold and 2.64­fold (P<0.05), respectively. No significant differences were found in the mRNA and protein levels of Akt between the two groups (P>0.05). The protein levels of phosphorylated (p­)Akt, Erk1/2, p­Erk1/2, HIF­1α and HIF­1ß were significantly increased by 5.89­, 0.5­, 0.59­, 1.46­ and 0.92­fold, respectively, in the patients with PAH, compared with those in the controls group (P<0.01 for p­Akt, Erk1/2; P<0.05 for p­Erk1/2, HIF­1α and HIF­1ß). These findings suggested that the mitogen­activated protein kinase and PI3K/Akt signaling pathways, and HIF­1 may perform a specific function in the pathogenesis of PAH.


Asunto(s)
Translocador Nuclear del Receptor de Aril Hidrocarburo/biosíntesis , Hipertensión Pulmonar/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Proteína Quinasa 1 Activada por Mitógenos/biosíntesis , Adolescente , Adulto , Translocador Nuclear del Receptor de Aril Hidrocarburo/genética , Femenino , Regulación de la Expresión Génica , Humanos , Hipertensión Pulmonar/patología , Subunidad alfa del Factor 1 Inducible por Hipoxia/biosíntesis , Pulmón/metabolismo , Pulmón/patología , Sistema de Señalización de MAP Quinasas/genética , Masculino , Persona de Mediana Edad , Proteína Quinasa 1 Activada por Mitógenos/genética , Fosfatidilinositol 3-Quinasas/genética , Fosforilación , Proteínas Proto-Oncogénicas c-akt/biosíntesis , Proteínas Proto-Oncogénicas c-akt/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA