Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
J Infect Dis ; 222(10): 1713-1722, 2020 10 13.
Artículo en Inglés | MEDLINE | ID: mdl-32453818

RESUMEN

BACKGROUND: Infection-induced preterm birth is a major cause of neonatal mortality and morbidity and leads to preterm premature rupture of placental chorioamniotic membranes. The loss of amniotic epithelial cells and tensile strength preceding membrane rupture is poorly understood. We hypothesized that intrauterine bacterial infection induces changes in microRNA (miRNA) expression, leading to amniotic epithelial cell loss and membrane weakening. METHODS: Ten pregnant pigtail macaques received choriodecidual inoculation of either group B Streptococcus (GBS) or saline (n = 5/group). Placental chorioamniotic membranes were studied using RNA microarray and immunohistochemistry. Chorioamniotic membranes from women with preterm premature rupture of membranes (pPROM) and normal term pregnancies were studied using transmission electron microscopy. RESULTS: In our model, an experimental GBS infection was associated with changes in the miRNA profile in the chorioamniotic membranes consistent with epithelial to mesenchymal transition (EMT) with loss of epithelial (E-cadherin) and gain of mesenchymal (vimentin) markers. Similarly, loss of desmosomes (intercellular junctions) was seen in placental tissues from women with pPROM. CONCLUSIONS: We describe EMT as a novel mechanism for infection-associated chorioamniotic membrane weakening, which may be a common pathway for many etiologies of pPROM. Therapy based on anti-miRNA targeting of EMT may prevent pPROM due to perinatal infection.


Asunto(s)
Transición Epitelial-Mesenquimal/fisiología , Rotura Prematura de Membranas Fetales/metabolismo , MicroARNs/metabolismo , Infecciones Estreptocócicas/metabolismo , Amnios/patología , Animales , Corioamnionitis/microbiología , Modelos Animales de Enfermedad , Femenino , Rotura Prematura de Membranas Fetales/etiología , Rotura Prematura de Membranas Fetales/microbiología , Rotura Prematura de Membranas Fetales/patología , Humanos , Inmunohistoquímica , Macaca nemestrina , MicroARNs/genética , Embarazo , Nacimiento Prematuro , Infecciones Estreptocócicas/complicaciones , Streptococcus agalactiae
2.
Am J Obstet Gynecol ; 218(4): 438.e1-438.e16, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29475580

RESUMEN

BACKGROUND: Most early preterm births are associated with intraamniotic infection and inflammation, which can lead to systemic inflammation in the fetus. The fetal inflammatory response syndrome describes elevations in the fetal interleukin-6 level, which is a marker for inflammation and fetal organ injury. An understanding of the effects of inflammation on fetal cardiac development may lead to insight into the fetal origins of adult cardiovascular disease. OBJECTIVE: The purpose of this study was to determine whether the fetal inflammatory response syndrome is associated with disruptions in gene networks that program fetal cardiac development. STUDY DESIGN: We obtained fetal cardiac tissue after necropsy from a well-described pregnant nonhuman primate model (pigtail macaque, Macaca nemestrina) of intrauterine infection (n=5) and controls (n=5). Cases with the fetal inflammatory response syndrome (fetal plasma interleukin-6 >11 pg/mL) were induced by either choriodecidual inoculation of a hypervirulent group B streptococcus strain (n=4) or intraamniotic inoculation of Escherichia coli (n=1). RNA and protein were extracted from fetal hearts and profiled by microarray and Luminex (Millipore, Billerica, MA) for cytokine analysis, respectively. Results were validated by quantitative reverse transcriptase polymerase chain reaction. Statistical and bioinformatics analyses included single gene analysis, gene set analysis, Ingenuity Pathway Analysis (Qiagen, Valencia, CA), and Wilcoxon rank sum. RESULTS: Severe fetal inflammation developed in the context of intraamniotic infection and a disseminated bacterial infection in the fetus. Interleukin-6 and -8 in fetal cardiac tissues were elevated significantly in fetal inflammatory response syndrome cases vs controls (P<.05). A total of 609 probe sets were expressed differentially (>1.5-fold change, P<.05) in the fetal heart (analysis of variance). Altered expression of select genes was validated by quantitative reverse transcriptase polymerase chain reaction that included several with known functions in cardiac injury, morphogenesis, angiogenesis, and tissue remodeling (eg, angiotensin I converting enzyme 2, STEAP family member 4, natriuretic peptide A, and secreted frizzled-related protein 4; all P<.05). Multiple gene sets and pathways that are involved in cardiac morphogenesis and vasculogenesis were downregulated significantly by gene set and Ingenuity Pathway Analysis (hallmark transforming growth factor beta signaling, cellular morphogenesis during differentiation, morphology of cardiovascular system; all P<.05). CONCLUSION: Disruption of gene networks for cardiac morphogenesis and vasculogenesis occurred in the preterm fetal heart of nonhuman primates with preterm labor, intraamniotic infection, and severe fetal inflammation. Inflammatory injury to the fetal heart in utero may contribute to the development of heart disease later in life. Development of preterm labor therapeutics must also target fetal inflammation to lessen organ injury and potential long-term effects on cardiac function.


Asunto(s)
Enfermedades Fetales/metabolismo , Miocardio/metabolismo , Síndrome de Respuesta Inflamatoria Sistémica/metabolismo , Enzima Convertidora de Angiotensina 2 , Animales , Factor Natriurético Atrial/genética , Biomarcadores/metabolismo , Corioamnionitis/metabolismo , Regulación hacia Abajo , Femenino , Corazón/microbiología , Interleucina-6/metabolismo , Interleucina-8/metabolismo , Macaca nemestrina , Proteínas de la Membrana/genética , Análisis por Micromatrices , Modelos Animales , Trabajo de Parto Prematuro , Oxidorreductasas/genética , Peptidil-Dipeptidasa A/genética , Embarazo , Proteínas Proto-Oncogénicas/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Regulación hacia Arriba
3.
Hum Mol Genet ; 23(6): 1606-18, 2014 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-24191263

RESUMEN

Expansion of CAG/CTG trinucleotide repeats causes numerous inherited neurological disorders, including Huntington's disease (HD), several spinocerebellar ataxias and myotonic dystrophy type 1. Expanded repeats are genetically unstable with a propensity to further expand when transmitted from parents to offspring. For many alleles with expanded repeats, extensive somatic mosaicism has been documented. For CAG repeat diseases, dramatic instability has been documented in the striatum, with larger expansions noted with advancing age. In contrast, only modest instability occurs in the cerebellum. Using microarray expression analysis, we sought to identify the genetic basis of these regional instability differences by comparing gene expression in the striatum and cerebellum of aged wild-type C57BL/6J mice. We identified eight candidate genes enriched in cerebellum, and validated four--Pcna, Rpa1, Msh6 and Fen1--along with a highly associated interactor, Lig1. We also explored whether expression levels of mismatch repair (MMR) proteins are altered in a line of HD transgenic mice, R6/2, that is known to show pronounced regional repeat instability. Compared with wild-type littermates, MMR expression levels were not significantly altered in R6/2 mice regardless of age. Interestingly, expression levels of these candidates were significantly increased in the cerebellum of control and HD human samples in comparison to striatum. Together, our data suggest that elevated expression levels of DNA replication and repair proteins in cerebellum may act as a safeguard against repeat instability, and may account for the dramatically reduced somatic instability present in this brain region, compared with the marked instability observed in the striatum.


Asunto(s)
Cerebelo/metabolismo , Cuerpo Estriado/metabolismo , Reparación de la Incompatibilidad de ADN , Enfermedad de Huntington/genética , Factores de Edad , Animales , ADN Ligasa (ATP) , ADN Ligasas/genética , Proteínas de Unión al ADN/genética , Femenino , Endonucleasas de ADN Solapado/genética , Regulación de la Expresión Génica , Humanos , Enfermedad de Huntington/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Inestabilidad de Microsatélites , Antígeno Nuclear de Célula en Proliferación/genética , Proteína de Replicación A/genética , Repeticiones de Trinucleótidos
4.
Am J Obstet Gynecol ; 213(6): 830.e1-830.e19, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26284599

RESUMEN

OBJECTIVE: Uterine overdistention is thought to induce preterm labor in women with twin and multiple pregnancies, but the pathophysiology remains unclear. We investigated for the first time the pathogenesis of preterm birth associated with rapid uterine distention in a pregnant nonhuman primate model. STUDY DESIGN: A nonhuman primate model of uterine overdistention was created using preterm chronically catheterized pregnant pigtail macaques (Macaca nemestrina) by inflation of intraamniotic balloons (N = 6), which were compared to saline controls (N = 5). Cesarean delivery was performed due to preterm labor or at experimental end. Microarray, quantitative reverse transcriptase polymerase chain reaction, Luminex (Austin, TX), and enzyme-linked immunosorbent assay were used to measure messenger RNA (mRNA) and/or protein levels from monkey (amniotic fluid, myometrium, maternal plasma) and human (amniocytes, amnion, myometrium) tissues. Statistical analysis employed analysis of covariance and Wilcoxon rank sum. Biomechanical forces were calculated using the law of Laplace. RESULTS: Preterm labor occurred in 3 of 6 animals after balloon inflation and correlated with greater balloon volume and uterine wall stress. Significant elevations of inflammatory cytokines and prostaglandins occurred following uterine overdistention in an "inflammatory pulse" that correlated with preterm labor (interleukin [IL]-1ß, tumor necrosis factor [TNF]-α, IL-6, IL-8, CCL2, prostaglandin E2, prostaglandin F2α, all P < .05). A similar inflammatory response was observed in amniocytes in vitro following mechanical stretch (IL1ß, IL6, and IL8 mRNA multiple time points, P < .05), in amnion of women with polyhydramnios (IL6 and TNF mRNA, P < .05) and in amnion (TNF-α) and myometrium of women with twins in early labor (IL6, IL8, CCL2, all P < .05). Genes differentially expressed in the nonhuman primate after balloon inflation and in women with polyhydramnios and twins are involved in tissue remodeling and muscle growth. CONCLUSION: Uterine overdistention by inflation of an intraamniotic balloon is associated with an inflammatory pulse that precedes and correlates with preterm labor. Our results indicate that inflammation is an early event after a mechanical stress on the uterus and leads to preterm labor when the stress is sufficiently great. Further, we find evidence of uterine tissue remodeling and muscle growth as a common, perhaps compensatory, response to uterine distension.


Asunto(s)
Inflamación/metabolismo , Trabajo de Parto Prematuro/fisiopatología , Estrés Mecánico , Útero/fisiopatología , Amnios/metabolismo , Animales , Citocinas/genética , Citocinas/metabolismo , Dinoprost/genética , Dinoprost/metabolismo , Dinoprostona/genética , Dinoprostona/metabolismo , Femenino , Humanos , Macaca nemestrina , Modelos Animales , Miometrio/metabolismo , Polihidramnios/metabolismo , Embarazo , Embarazo Múltiple/fisiología , ARN Mensajero/metabolismo
5.
Int J Cancer ; 134(4): 778-88, 2014 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-23929039

RESUMEN

Cirrhosis is the primary risk factor for the development of hepatocellular carcinoma (HCC), yet the mechanisms by which cirrhosis predisposes to carcinogenesis are poorly understood. Using a mouse model that recapitulates many aspects of the pathophysiology of human liver disease, we explored the mechanisms by which changes in the liver microenvironment induce dysplasia and HCC. Hepatic expression of platelet-derived growth factor C (PDGF-C) induces progressive fibrosis, chronic inflammation, neoangiogenesis and sinusoidal congestion, as well as global changes in gene expression. Using reporter mice, immunofluorescence, immunohistochemistry and liver cell isolation, we demonstrate that receptors for PDGF-CC are localized on hepatic stellate cells (HSCs), which proliferate, and transform into myofibroblast-like cells that deposit extracellular matrix and lead to production of growth factors and cytokines. We demonstrate induction of cytokine genes at 2 months, and stromal cell-derived hepatocyte growth factors that coincide with the onset of dysplasia at 4 months. Our results support a paracrine signaling model wherein hepatocyte-derived PDGF-C stimulates widespread HSC activation throughout the liver leading to chronic inflammation, liver injury and architectural changes. These complex changes to the liver microenvironment precede the development of HCC. Further, increased PDGF-CC levels were observed in livers of patients with nonalcoholic fatty steatohepatitis and correlate with the stage of disease, suggesting a role for this growth factor in chronic liver disease in humans. PDGF-C transgenic mice provide a unique model for the in vivo study of tumor-stromal interactions in the liver.


Asunto(s)
Carcinoma Hepatocelular/patología , Hígado Graso/patología , Células Estrelladas Hepáticas/patología , Neoplasias Hepáticas/patología , Linfocinas/metabolismo , Comunicación Paracrina , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Células del Estroma/patología , Animales , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Western Blotting , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/metabolismo , Estudios de Cohortes , Citocinas/genética , Citocinas/metabolismo , Hígado Graso/genética , Hígado Graso/metabolismo , Técnica del Anticuerpo Fluorescente , Perfilación de la Expresión Génica , Células Estrelladas Hepáticas/metabolismo , Hepatocitos/metabolismo , Hepatocitos/patología , Humanos , Técnicas para Inmunoenzimas , Inflamación/genética , Inflamación/metabolismo , Inflamación/patología , Hígado/metabolismo , Hígado/patología , Cirrosis Hepática/genética , Cirrosis Hepática/metabolismo , Cirrosis Hepática/patología , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Linfocinas/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Enfermedad del Hígado Graso no Alcohólico , Análisis de Secuencia por Matrices de Oligonucleótidos , Factor de Crecimiento Derivado de Plaquetas/genética , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células del Estroma/metabolismo
6.
J Toxicol Environ Health A ; 77(6): 293-312, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24593143

RESUMEN

Mercury (Hg) is neurotoxic and children may be particularly susceptible to this effect. A current major challenge is identification of children who may be uniquely susceptible to Hg toxicity because of genetic disposition. This study examined the hypothesis that genetic variants of catechol-O-methyltransferase (COMT) that are reported to alter neurobehavioral functions that are also affected by Hg in adults might modify the adverse neurobehavioral effects of Hg exposure in children. Five hundred and seven children, 8-12 yr of age at baseline, participated in a clinical trial to evaluate the neurobehavioral effects of Hg from dental amalgam tooth fillings. Subjects were evaluated at baseline and at seven subsequent annual intervals for neurobehavioral performance and urinary Hg levels. Following the clinical trial, genotyping assays were performed for single-nucleotide polymorphisms (SNPs) of COMT rs4680, rs4633, rs4818, and rs6269 on biological samples provided by 330 of the trial participants. Regression-modeling strategies were employed to evaluate associations between allelic status, Hg exposure, and neurobehavioral test outcomes. Similar analysis was performed using haplotypes of COMT SNPs. Among girls, few interactions for Hg exposure and COMT variants were found. In contrast, among boys, numerous gene-Hg interactions were observed between individual COMT SNPs, as well as with a common COMT haplotype affecting multiple domains of neurobehavioral function. These findings suggest increased susceptibility to the adverse neurobehavioral effects of Hg among children with common genetic variants of COMT, and may have important implications for strategies aimed at protecting children from the potential health risks associated with Hg exposure.


Asunto(s)
Catecol O-Metiltransferasa/genética , Amalgama Dental/toxicidad , Mercurio/toxicidad , Pruebas Neuropsicológicas , Polimorfismo de Nucleótido Simple , Catecol O-Metiltransferasa/sangre , Niño , Femenino , Haplotipos , Humanos , Masculino , Análisis de Regresión
7.
Cell Metab ; 4(5): 349-62, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17055784

RESUMEN

Huntington's disease (HD) is a fatal, dominantly inherited disorder caused by polyglutamine repeat expansion in the huntingtin (htt) gene. Here, we observe that HD mice develop hypothermia associated with impaired activation of brown adipose tissue (BAT). Although sympathetic stimulation of PPARgamma coactivator 1alpha (PGC-1alpha) was intact in BAT of HD mice, uncoupling protein 1 (UCP-1) induction was blunted. In cultured cells, expression of mutant htt suppressed UCP-1 promoter activity; this was reversed by PGC-1alpha expression. HD mice showed reduced food intake and increased energy expenditure, with dysfunctional BAT mitochondria. PGC-1alpha is a known regulator of mitochondrial function; here, we document reduced expression of PGC-1alpha target genes in HD patient and mouse striatum. Mitochondria of HD mouse brain show reduced oxygen consumption rates. Finally, HD striatal neurons expressing exogenous PGC-1alpha were resistant to 3-nitropropionic acid treatment. Altered PGC-1alpha function may thus link transcription dysregulation and mitochondrial dysfunction in HD.


Asunto(s)
Tejido Adiposo Pardo/fisiopatología , Regulación de la Temperatura Corporal/genética , Proteínas de Choque Térmico/metabolismo , Enfermedad de Huntington/etiología , Factores de Transcripción/metabolismo , Animales , Temperatura Corporal/genética , Células Cultivadas , Modelos Animales de Enfermedad , Proteínas de Choque Térmico/genética , Enfermedad de Huntington/genética , Enfermedad de Huntington/metabolismo , Ratones , Ratones Transgénicos , Mitocondrias/fisiología , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma , Transducción de Señal/genética , Factores de Transcripción/genética , Transcripción Genética
8.
Dev Neurosci ; 30(4): 231-42, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-17960054

RESUMEN

To identify neuroprotective changes in gene expression, we developed a neonatal mouse model of moderate to severe oxidative brain injury and hypothesized that recombinant erythropoietin (rEpo) would decrease the expression of proapoptotic and proinflammatory genes 24 and 48 h, respectively, after injury and increase the expression of neurogenic and angiogenic genes 168 h after injury. Postnatal day 10 BALB-c mice underwent sham surgery or right common carotid artery occlusion followed by alternating hypoxia and hyperoxia and were then treated with rEpo (5,000 U/kg s.c.) or saline (vehicle) daily for up to three doses. At death, gross brain injury was assessed, then hippocampus, cortex, and thalamus were isolated for RNA or protein extraction. Microarray analysis, real-time polymerase chain reaction, and Bio-Plex suspension array system validation were performed. rEpo decreased both incidence and severity of brain injury (median injury score 3 vs. 0, p < 0.0001) and reduced the injury-induced increases in interleukin-1alpha and interleukin-6 gene expression (p < 0.001), with corresponding effects on protein translation. Similarly, the expression of caspase-1, caspase-4, and caspase-6 and of p53 was increased by brain injury at 24 h, but mitigated by rEpo (p < 0.01). The interleukin-10 expression was higher in the rEpo-treated animals. Apoptotic and proinflammatory gene expression persisted for 168 h. There was no increase in angiogenic gene expression at the time points studied.


Asunto(s)
Modelos Animales de Enfermedad , Eritropoyetina/farmacología , Hipoxia Encefálica/tratamiento farmacológico , Ratones Endogámicos BALB C , Fármacos Neuroprotectores/farmacología , Animales , Caspasa 1/genética , Caspasa 6/genética , Caspasas/genética , Caspasas Iniciadoras , Expresión Génica/efectos de los fármacos , Hipoxia Encefálica/metabolismo , Hipoxia Encefálica/fisiopatología , Interleucina-10/metabolismo , Interleucina-1alfa/genética , Interleucina-1alfa/metabolismo , Interleucina-6/genética , Interleucina-6/metabolismo , Ratones , Análisis de Secuencia por Matrices de Oligonucleótidos , Estrés Oxidativo/efectos de los fármacos , Proteínas Recombinantes , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo , Proteína p53 Supresora de Tumor/genética
9.
Redox Biol ; 9: 264-275, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27596734

RESUMEN

Inhalation of multiwalled carbon nanotubes (MWCNTs) during their manufacture or incorporation into various commercial products may cause lung inflammation, fibrosis, and oxidative stress in exposed workers. Some workers may be more susceptible to these effects because of differences in their ability to synthesize the major antioxidant and immune system modulator glutathione (GSH). Accordingly, in this study we examined the influence of GSH synthesis and gender on MWCNT-induced lung inflammation in C57BL/6 mice. GSH synthesis was impaired through genetic manipulation of Gclm, the modifier subunit of glutamate cysteine ligase, the rate-limiting enzyme in GSH synthesis. Twenty-four hours after aspirating 25µg of MWCNTs, all male mice developed neutrophilia in their lungs, regardless of Gclm genotype. However, female mice with moderate (Gclm heterozygous) and severe (Gclm null) GSH deficiencies developed significantly less neutrophilia. We found no indications of MWCNT-induced oxidative stress as reflected in the GSH content of lung tissue and epithelial lining fluid, 3-nitrotyrosine formation, or altered mRNA or protein expression of several redox-responsive enzymes. Our results indicate that GSH-deficient female mice are rendered uniquely susceptible to an attenuated neutrophil response. If the same effects occur in humans, GSH-deficient women manufacturing MWCNTs may be at greater risk for impaired neutrophil-dependent clearance of MWCNTs from the lung. In contrast, men may have effective neutrophil-dependent clearance, but may be at risk for lung neutrophilia regardless of their GSH levels.


Asunto(s)
Glutatión/biosíntesis , Nanotubos de Carbono/efectos adversos , Oxidación-Reducción , Estrés Oxidativo , Neumonía/etiología , Neumonía/metabolismo , Animales , Líquido del Lavado Bronquioalveolar , Citocinas/metabolismo , Femenino , Fibrosis/genética , Fibrosis/metabolismo , Regulación de la Expresión Génica , Mediadores de Inflamación/metabolismo , Pulmón/metabolismo , Pulmón/patología , Masculino , Ratones , Ratones Noqueados , Factores Sexuales
10.
Arch Oral Biol ; 60(9): 1450-60, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26188427

RESUMEN

OBJECTIVE: Previous investigations suggest that the embryonic origins of the calvarial tissues (neural crest or mesoderm) may account for the molecular mechanisms underlying sutural development. The aim of this study was to evaluate the differences in the gene expression of human cranial tissues and assess the presence of an expression signature reflecting their embryonic origins. METHODS: Using microarray technology, we investigated global gene expression of cells from the frontal and parietal bones and the metopic and sagittal intrasutural mesenchyme (ISM) of four human foetal calvaria. qRT-PCR of a selected group of genes was done to validate the microarray analysis. Paired comparison and correlation analyses were performed on microarray results. RESULTS: Of six paired comparisons, frontal and parietal compartments (distinct tissue types of calvaria, either bone or intrasutural mesenchyme) had the most different gene expression profiles despite being composed of the same tissue type (bone). Correlation analysis revealed two distinct gene expression profiles that separate frontal and metopic compartments from parietal and sagittal compartments. TFAP2A, TFAP2B, ICAM1, SULF1, TNC and FOXF2 were among differentially expressed genes. CONCLUSION: Transcriptional profiles of two groups of tissues, frontal and metopic compartments vs. parietal and sagittal compartments, suggest differences in proliferation, differentiation and extracellular matrix production. Our data suggest that in the second trimester of human foetal development, a gene expression signature of neural crest origin still exists in frontal and metopic compartments while gene expression of parietal and sagittal compartments is more similar to mesoderm.


Asunto(s)
Feto/embriología , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Cráneo/embriología , Transcripción Genética , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Femenino , Humanos , Masculino , Análisis por Micromatrices , Osteogénesis/genética , Osteogénesis/fisiología , ARN/análisis , Reacción en Cadena en Tiempo Real de la Polimerasa , Cráneo/citología
11.
Cell Metab ; 22(5): 895-906, 2015 Nov 03.
Artículo en Inglés | MEDLINE | ID: mdl-26456335

RESUMEN

Many genes that affect replicative lifespan (RLS) in the budding yeast Saccharomyces cerevisiae also affect aging in other organisms such as C. elegans and M. musculus. We performed a systematic analysis of yeast RLS in a set of 4,698 viable single-gene deletion strains. Multiple functional gene clusters were identified, and full genome-to-genome comparison demonstrated a significant conservation in longevity pathways between yeast and C. elegans. Among the mechanisms of aging identified, deletion of tRNA exporter LOS1 robustly extended lifespan. Dietary restriction (DR) and inhibition of mechanistic Target of Rapamycin (mTOR) exclude Los1 from the nucleus in a Rad53-dependent manner. Moreover, lifespan extension from deletion of LOS1 is nonadditive with DR or mTOR inhibition, and results in Gcn4 transcription factor activation. Thus, the DNA damage response and mTOR converge on Los1-mediated nuclear tRNA export to regulate Gcn4 activity and aging.


Asunto(s)
Envejecimiento/genética , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/genética , Longevidad/genética , Proteínas de Complejo Poro Nuclear/genética , Proteínas de Saccharomyces cerevisiae/genética , Envejecimiento/metabolismo , Envejecimiento/patología , Animales , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/metabolismo , Caenorhabditis elegans/genética , Restricción Calórica , Daño del ADN/genética , Eliminación de Gen , Regulación de la Expresión Génica/genética , Genoma , ARN de Transferencia/genética , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Serina-Treonina Quinasas TOR/genética
12.
PLoS One ; 6(6): e20911, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21731631

RESUMEN

Methylazoxymethanol (MAM), the genotoxic metabolite of the cycad azoxyglucoside cycasin, induces genetic alterations in bacteria, yeast, plants, insects and mammalian cells, but adult nerve cells are thought to be unaffected. We show that the brains of adult C57BL6 wild-type mice treated with a single systemic dose of MAM acetate display DNA damage (O6-methyldeoxyguanosine lesions, O6-mG) that remains constant up to 7 days post-treatment. By contrast, MAM-treated mice lacking a functional gene encoding the DNA repair enzyme O6-mG DNA methyltransferase (MGMT) showed elevated O6-mG DNA damage starting at 48 hours post-treatment. The DNA damage was linked to changes in the expression of genes in cell-signaling pathways associated with cancer, human neurodegenerative disease, and neurodevelopmental disorders. These data are consistent with the established developmental neurotoxic and carcinogenic properties of MAM in rodents. They also support the hypothesis that early-life exposure to MAM-glucoside (cycasin) has an etiological association with a declining, prototypical neurodegenerative disease seen in Guam, Japan, and New Guinea populations that formerly used the neurotoxic cycad plant for food or medicine, or both. These findings suggest environmental genotoxins, specifically MAM, target common pathways involved in neurodegeneration and cancer, the outcome depending on whether the cell can divide (cancer) or not (neurodegeneration). Exposure to MAM-related environmental genotoxins may have relevance to the etiology of related tauopathies, notably, Alzheimer's disease.


Asunto(s)
Neoplasias Encefálicas/patología , Encéfalo/metabolismo , Daño del ADN , Acetato de Metilazoximetanol/análogos & derivados , Mutágenos/toxicidad , Enfermedades Neurodegenerativas/patología , Transducción de Señal/efectos de los fármacos , Animales , Sitios de Unión , Encéfalo/efectos de los fármacos , Neoplasias Encefálicas/metabolismo , Cycadopsida/química , Metilasas de Modificación del ADN/deficiencia , Metilasas de Modificación del ADN/metabolismo , Enzimas Reparadoras del ADN/deficiencia , Enzimas Reparadoras del ADN/metabolismo , Perfilación de la Expresión Génica , Redes Reguladoras de Genes/efectos de los fármacos , Guanosina/análogos & derivados , Guanosina/metabolismo , Humanos , Hígado/efectos de los fármacos , Hígado/metabolismo , Masculino , Acetato de Metilazoximetanol/toxicidad , Ratones , Ratones Endogámicos C57BL , Modelos Biológicos , Enfermedades Neurodegenerativas/metabolismo , Especificidad de Órganos/efectos de los fármacos , Factores de Transcripción/metabolismo , Transcripción Genética/efectos de los fármacos , Proteínas Supresoras de Tumor/deficiencia , Proteínas Supresoras de Tumor/metabolismo
13.
OMICS ; 13(6): 501-11, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20001863

RESUMEN

We have previously found that CHF1/Hey2 prevents the development of phenylephrine-induced cardiac hypertrophy. To determine the role of CHF1/Hey2 in pressure overload hypertrophy, we performed ascending aortic banding on wild-type and transgenic mice overexpressing CHF1/Hey2 in the myocardium. We found that both wild-type and transgenic mice developed increased ventricular weight to body weight ratios 1 week after aortic banding. Wild-type mice also developed decreased fractional shortening after 1 week when compared to preoperative echocardiograms and sham-operated controls. Transgenic mice, in comparison, demonstrated preserved fractional shortening. Histological examination of explanted heart tissue demonstrated extensive fibrosis in wild-type hearts, but minimal fibrosis in transgenic hearts. TUNEL staining demonstrated increased apoptosis in the wild-type hearts but not in the transgenic hearts. Exposure of cultured neonatal myocytes from wild-type and transgenic animals to hydrogen peroxide, a potent inducer of apoptosis, demonstrated increased apoptosis in the wild-type cells. Gene Set Analysis of microarray data from wild-type and transgenic hearts 1 week after banding revealed suppression and activation of multiple pathways involving apoptosis, cell signaling, and biosynthesis. These findings demonstrate that CHF1/Hey2 promotes physiological over pathological hypertrophy through suppression of apoptosis and regulation of multiple transcriptional pathways. These findings also suggest that CHF1/Hey2 and its downstream pathways provide a variety of targets for novel heart failure drug discovery, and that genetic polymorphisms in CHF1/Hey2 may affect susceptibility to hypertrophy and heart failure.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Cardiomegalia/metabolismo , Regulación de la Expresión Génica , Miocardio , Proteínas Represoras/metabolismo , Transducción de Señal/fisiología , Transcripción Genética , Animales , Apoptosis/fisiología , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Presión Sanguínea/fisiología , Cardiomegalia/patología , Células Cultivadas , Fibrosis/patología , Etiquetado Corte-Fin in Situ , Ratones , Ratones Transgénicos , Análisis por Micromatrices , Datos de Secuencia Molecular , Miocardio/metabolismo , Miocardio/patología , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Proteínas Represoras/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA