Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 80
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
J Infect Dis ; 222(10): 1713-1722, 2020 10 13.
Artigo em Inglês | MEDLINE | ID: mdl-32453818

RESUMO

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.


Assuntos
Transição Epitelial-Mesenquimal/fisiologia , Ruptura Prematura de Membranas Fetais/metabolismo , MicroRNAs/metabolismo , Infecções Estreptocócicas/metabolismo , Âmnio/patologia , Animais , Corioamnionite/microbiologia , Modelos Animais de Doenças , Feminino , Ruptura Prematura de Membranas Fetais/etiologia , Ruptura Prematura de Membranas Fetais/microbiologia , Ruptura Prematura de Membranas Fetais/patologia , Humanos , Imuno-Histoquímica , Macaca nemestrina , MicroRNAs/genética , Gravidez , Nascimento Prematuro , Infecções Estreptocócicas/complicações , Streptococcus agalactiae
2.
Hum Mol Genet ; 25(10): 2060-2069, 2016 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-26984941

RESUMO

The Werner syndrome (WS) is a prototypic adult Mendelian progeroid syndrome in which signs of premature aging are associated with genomic instability and an elevated risk of cancer. The WRN RECQ helicase protein binds and unwinds G-quadruplex (G4) DNA substrates in vitro, and we identified significant enrichment in G4 sequence motifs at the transcription start site and 5' ends of first introns (false discovery rate < 0.001) of genes down-regulated in WS patient fibroblasts. This finding provides strong evidence that WRN binds G4 DNA structures at many chromosomal sites to modulate gene expression. WRN appears to bind a distinct subpopulation of G4 motifs in human cells, when compared with the related Bloom syndrome RECQ helicase protein. Functional annotation of the genes and miRNAs altered in WS provided new insight into WS disease pathogenesis. WS patient fibroblasts displayed altered expression of multiple, mechanistically distinct, senescence-associated gene expression programs, with altered expression of disease-associated miRNAs, and dysregulation of canonical pathways that regulate cell signaling, genome stability and tumorigenesis. WS fibroblasts also displayed a highly statistically significant and distinct gene expression signature, with coordinate overexpression of nearly all of the cytoplasmic tRNA synthetases and associated ARS-interacting multifunctional protein genes. The 'non-canonical' functions of many of these upregulated tRNA charging proteins may together promote WS disease pathogenesis. Our results identify the human WRN RECQ protein as a G4 helicase that modulates gene expression in G4-dependent fashion at many chromosomal sites and provide several new and unexpected mechanistic insights into WS disease pathogenesis.


Assuntos
Proteínas de Ligação a DNA/genética , Instabilidade Genômica/genética , Neoplasias/genética , RecQ Helicases/genética , Síndrome de Werner/genética , Carcinogênese/genética , Proteínas de Ligação a DNA/metabolismo , Fibroblastos , Quadruplex G , Regulação da Expressão Gênica , Genoma Humano , Humanos , MicroRNAs , Neoplasias/patologia , Motivos de Nucleotídeos , RecQ Helicases/metabolismo
3.
Proc Natl Acad Sci U S A ; 111(27): 9905-10, 2014 Jul 08.
Artigo em Inglês | MEDLINE | ID: mdl-24958861

RESUMO

Bloom syndrome is a rare autosomal recessive disorder characterized by genetic instability and cancer predisposition, and caused by mutations in the gene encoding the Bloom syndrome, RecQ helicase-like (BLM) protein. To determine whether altered gene expression might be responsible for pathological features of Bloom syndrome, we analyzed mRNA and microRNA (miRNA) expression in fibroblasts from individuals with Bloom syndrome and in BLM-depleted control fibroblasts. We identified mRNA and miRNA expression differences in Bloom syndrome patient and BLM-depleted cells. Differentially expressed mRNAs are connected with cell proliferation, survival, and molecular mechanisms of cancer, and differentially expressed miRNAs target genes involved in cancer and in immune function. These and additional altered functions or pathways may contribute to the proportional dwarfism, elevated cancer risk, immune dysfunction, and other features observed in Bloom syndrome individuals. BLM binds to G-quadruplex (G4) DNA, and G4 motifs were enriched at transcription start sites (TSS) and especially within first introns (false discovery rate ≤ 0.001) of differentially expressed mRNAs in Bloom syndrome compared with normal cells, suggesting that G-quadruplex structures formed at these motifs are physiologic targets for BLM. These results identify a network of mRNAs and miRNAs that may drive the pathogenesis of Bloom syndrome.


Assuntos
Síndrome de Bloom/genética , DNA/química , Quadruplex G , Regulação Enzimológica da Expressão Gênica , RecQ Helicases/genética , Células Cultivadas , Perfilação da Expressão Gênica , Humanos , RNA Mensageiro/genética
4.
Hum Mol Genet ; 23(6): 1606-18, 2014 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-24191263

RESUMO

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.


Assuntos
Cerebelo/metabolismo , Corpo Estriado/metabolismo , Reparo de Erro de Pareamento de DNA , Doença de Huntington/genética , Fatores Etários , Animais , DNA Ligase Dependente de ATP , DNA Ligases/genética , Proteínas de Ligação a DNA/genética , Feminino , Endonucleases Flap/genética , Regulação da Expressão Gênica , Humanos , Doença de Huntington/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Instabilidade de Microssatélites , Antígeno Nuclear de Célula em Proliferação/genética , Proteína de Replicação A/genética , Repetições de Trinucleotídeos
5.
PLoS Pathog ; 10(3): e1003920, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24603861

RESUMO

Early events leading to intrauterine infection remain poorly defined, but may hold the key to preventing preterm delivery. To determine molecular pathways within fetal membranes (chorioamnion) associated with early choriodecidual infection that may progress to preterm premature rupture of membranes (PPROM), we examined the effects of a Group B Streptococcus (GBS) choriodecidual infection on chorioamnion in a nonhuman primate model. Ten chronically catheterized pregnant monkeys (Macaca nemestrina) at 118-125 days gestation (term = 172 days) received choriodecidual inoculation of either GBS (n = 5) or saline (n = 5). Cesarean section was performed in the first week after GBS or saline inoculation. RNA extracted from chorioamnion (inoculation site) was profiled by microarray. Single gene, Gene Set, and Ingenuity Pathway Analysis results were validated using qRT-PCR (chorioamnion), Luminex (amniotic fluid, AF), immunohistochemistry, and transmission electron microscopy (TEM). Despite uterine quiescence in most cases, significant elevations of AF cytokines (TNF-α, IL-8, IL-1ß, IL-6) were detected in GBS versus controls (p<0.05). Choriodecidual infection resolved by the time of cesarean section in 3 of 5 cases and GBS was undetectable by culture and PCR in the AF. A total of 331 genes were differentially expressed (>2-fold change, p<0.05). Remarkably, GBS exposure was associated with significantly downregulated expression of multiple cytokeratin (CK) and other cytoskeletal genes critical for maintenance of tissue tensile strength. Immunofluorescence revealed highly significant changes in the CK network within amniocytes with dense CK aggregates and retraction from the cell periphery (all p = 0.006). In human pregnancies affected by PPROM, there was further evidence of CK network retraction with significantly shorter amniocyte foot processes (p = 0.002). These results suggest early choriodecidual infection results in decreased cellular membrane integrity and tensile strength via dysfunction of CK networks. Downregulation of CK expression and perturbations in the amniotic epithelial cell intermediate filament network occur after GBS choriodecidual infection, which may contribute to PPROM.


Assuntos
Âmnio/patologia , Ruptura Prematura de Membranas Fetais/patologia , Queratinas/metabolismo , Efeitos Tardios da Exposição Pré-Natal/patologia , Infecções Estreptocócicas/patologia , Âmnio/microbiologia , Animais , Córion/microbiologia , Córion/patologia , Modelos Animais de Doenças , Feminino , Ruptura Prematura de Membranas Fetais/genética , Ruptura Prematura de Membranas Fetais/microbiologia , Imunofluorescência , Humanos , Imuno-Histoquímica , Macaca nemestrina , Microscopia Confocal , Microscopia Eletrônica de Transmissão , Análise de Sequência com Séries de Oligonucleotídeos , Gravidez , Efeitos Tardios da Exposição Pré-Natal/genética , Efeitos Tardios da Exposição Pré-Natal/microbiologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Infecções Estreptocócicas/genética , Streptococcus agalactiae , Transcriptoma
6.
Environ Sci Technol ; 50(4): 1990-9, 2016 Feb 16.
Artigo em Inglês | MEDLINE | ID: mdl-26730740

RESUMO

The use of sentinel species for population and ecosystem health assessments has been advocated as part of a One Health perspective. The Arctic is experiencing rapid change, including climate and environmental shifts, as well as increased resource development, which will alter exposure of biota to environmental agents of disease. Arctic canid species have wide geographic ranges and feeding ecologies and are often exposed to high concentrations of both terrestrial and marine-based contaminants. The domestic dog (Canis lupus familiaris) has been used in biomedical research for a number of years and has been advocated as a sentinel for human health due to its proximity to humans and, in some instances, similar diet. Exploiting the potential of molecular tools for describing the toxicogenomics of Arctic canids is critical for their development as biomedical models as well as environmental sentinels. Here, we present three approaches analyzing toxicogenomics of Arctic contaminants in both domestic and free-ranging canids (Arctic fox, Vulpes lagopus). We describe a number of confounding variables that must be addressed when conducting toxicogenomics studies in canid and other mammalian models. The ability for canids to act as models for Arctic molecular toxicology research is unique and significant for advancing our understanding and expanding the tool box for assessing the changing landscape of environmental agents of disease in the Arctic.


Assuntos
Ecotoxicologia/métodos , Exposição Ambiental/análise , Raposas , Perfilação da Expressão Gênica/métodos , Animais , Regiões Árticas , Cães/genética , Ecossistema , Monitoramento Ambiental/métodos , Poluentes Ambientais/toxicidade , Peixes , Raposas/genética , Mercúrio/toxicidade , Bifenilos Policlorados/toxicidade
7.
Proc Natl Acad Sci U S A ; 110(16): 6400-5, 2013 Apr 16.
Artigo em Inglês | MEDLINE | ID: mdl-23509287

RESUMO

The accumulation of damaged mitochondria has been proposed as a key factor in aging and the pathogenesis of many common age-related diseases, including Parkinson disease (PD). Recently, in vitro studies of the PD-related proteins Parkin and PINK1 have found that these factors act in a common pathway to promote the selective autophagic degradation of damaged mitochondria (mitophagy). However, whether Parkin and PINK1 promote mitophagy under normal physiological conditions in vivo is unknown. To address this question, we used a proteomic approach in Drosophila to compare the rates of mitochondrial protein turnover in parkin mutants, PINK1 mutants, and control flies. We found that parkin null mutants showed a significant overall slowing of mitochondrial protein turnover, similar to but less severe than the slowing seen in autophagy-deficient Atg7 mutants, consistent with the model that Parkin acts upstream of Atg7 to promote mitophagy. By contrast, the turnover of many mitochondrial respiratory chain (RC) subunits showed greater impairment in parkin than Atg7 mutants, and RC turnover was also selectively impaired in PINK1 mutants. Our findings show that the PINK1-Parkin pathway promotes mitophagy in vivo and, unexpectedly, also promotes selective turnover of mitochondrial RC subunits. Failure to degrade damaged RC proteins could account for the RC deficits seen in many PD patients and may play an important role in PD pathogenesis.


Assuntos
Proteínas de Drosophila/metabolismo , Transporte de Elétrons/fisiologia , Proteínas Mitocondriais/metabolismo , Mitofagia/fisiologia , Doença de Parkinson/etiologia , Proteínas Serina-Treonina Quinases/metabolismo , Transdução de Sinais/fisiologia , Ubiquitina-Proteína Ligases/metabolismo , Animais , Proteína 7 Relacionada à Autofagia , Encéfalo/metabolismo , Drosophila , Meia-Vida , Marcação por Isótopo , Espectrometria de Massas , Camundongos , Doença de Parkinson/metabolismo
8.
Infect Immun ; 83(10): 3909-17, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26195546

RESUMO

The mechanisms underlying fetal lung injury remain poorly defined. MicroRNAs (miRNAs) are small noncoding, endogenous RNAs that regulate gene expression and have been implicated in the pathogenesis of lung disease. Using a nonhuman primate model of choriodecidual infection, we sought to determine if differentially expressed miRNAs were associated with acute fetal lung injury. After inoculating 10 chronically catheterized pregnant monkeys (Macaca nemestrina) with either group B streptococcus (GBS) at 1 × 10(6) CFU (n = 5) or saline (n = 5) in the choriodecidual space, we extracted fetal lung mRNA and miRNA and profiled the changes in expression by microarray analysis. We identified 9 differentially expressed miRNAs in GBS-exposed fetal lungs, but of these, only miR-155-5p was validated by quantitative reverse transcription-PCR (P = 0.02). Significantly elevated miR-155-5p expression was also observed when immortalized human fetal airway epithelial (FeAE) cells were exposed to proinflammatory cytokines (interleukin-6 [IL-6] and tumor necrosis factor alpha [TNF-α]). Overexpression of miR-155-5p in FeAE cells in turn increased the production of IL-6 and CXCL10/gamma interferon-induced protein 10, which are implicated in leukocyte recruitment but also in protection from lung injury. Interestingly, while miR-155-5p decreased fibroblast growth factor 9 (FGF9) expression in a luciferase reporter assay, FGF9 levels were actually increased in GBS-exposed fetal lungs in vivo. FGF9 overexpression is associated with abnormal lung development. Thus, upregulation of miR-155-5p may serve as a compensatory mechanism to lessen the increase in FGF9 and prevent aberrant lung development. Understanding the complicated networks regulating lung development in the setting of infection is a key step in identifying how to prevent fetal lung injury leading to bronchopulmonary dysplasia.


Assuntos
Doenças Fetais/genética , Doenças Fetais/microbiologia , Pulmão/metabolismo , Infecções Estreptocócicas/embriologia , Infecções Estreptocócicas/genética , Streptococcus/fisiologia , Animais , Modelos Animais de Doenças , Feminino , Doenças Fetais/metabolismo , Humanos , Interleucina-6/genética , Interleucina-6/metabolismo , Pulmão/crescimento & desenvolvimento , Pulmão/microbiologia , Macaca nemestrina , Masculino , Gravidez , Infecções Estreptocócicas/metabolismo , Infecções Estreptocócicas/microbiologia , Streptococcus/genética , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/metabolismo
9.
Toxicol Appl Pharmacol ; 289(2): 240-50, 2015 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-26476918

RESUMO

Quantum dots (QDs) are engineered semiconductor nanoparticles with unique physicochemical properties that make them potentially useful in clinical, research and industrial settings. However, a growing body of evidence indicates that like other engineered nanomaterials, QDs have the potential to be respiratory hazards, especially in the context of the manufacture of QDs and products containing them, as well as exposures to consumers using these products. The overall goal of this study was to investigate the role of mouse strain in determining susceptibility to QD-induced pulmonary inflammation and toxicity. Male mice from 8 genetically diverse inbred strains (the Collaborative Cross founder strains) were exposed to CdSe-ZnS core-shell QDs stabilized with an amphiphilic polymer. QD treatment resulted in significant increases in the percentage of neutrophils and levels of cytokines present in bronchoalveolar lavage fluid (BALF) obtained from NOD/ShiLtJ and NZO/HlLtJ mice relative to their saline (Sal) treated controls. Cadmium measurements in lung tissue indicated strain-dependent differences in disposition of QDs in the lung. Total glutathione levels in lung tissue were significantly correlated with percent neutrophils in BALF as well as with lung tissue Cd levels. Our findings indicate that QD-induced acute lung inflammation is mouse strain dependent, that it is heritable, and that the choice of mouse strain is an important consideration in planning QD toxicity studies. These data also suggest that formal genetic analyses using additional strains or recombinant inbred strains from these mice could be useful for discovering potential QD-induced inflammation susceptibility loci.


Assuntos
Compostos de Cádmio/toxicidade , Pulmão/efeitos dos fármacos , Pneumonia/induzido quimicamente , Pontos Quânticos/toxicidade , Compostos de Selênio/toxicidade , Sulfetos/toxicidade , Compostos de Zinco/toxicidade , Animais , Líquido da Lavagem Broncoalveolar/imunologia , Análise por Conglomerados , Citocinas/metabolismo , Predisposição Genética para Doença , Glutationa/metabolismo , Hereditariedade , Pulmão/imunologia , Pulmão/metabolismo , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Masculino , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Infiltração de Neutrófilos/efeitos dos fármacos , Neutrófilos/efeitos dos fármacos , Neutrófilos/imunologia , Fenótipo , Pneumonia/genética , Pneumonia/imunologia , Pneumonia/metabolismo , Fatores de Risco , Especificidade da Espécie , Fatores de Tempo
10.
J Nat Prod ; 78(8): 1990-2000, 2015 Aug 28.
Artigo em Inglês | MEDLINE | ID: mdl-26186142

RESUMO

Silymarin, a characterized extract of the seeds of milk thistle (Silybum marianum), suppresses cellular inflammation. To define how this occurs, transcriptional profiling, metabolomics, and signaling studies were performed in human liver and T cell lines. Cellular stress and metabolic pathways were modulated within 4 h of silymarin treatment: activation of Activating Transcription Factor 4 (ATF-4) and adenosine monophosphate protein kinase (AMPK) and inhibition of mammalian target of rapamycin (mTOR) signaling, the latter being associated with induction of DNA-damage-inducible transcript 4 (DDIT4). Metabolomics analyses revealed silymarin suppression of glycolytic, tricarboxylic acid (TCA) cycle, and amino acid metabolism. Anti-inflammatory effects arose with prolonged (i.e., 24 h) silymarin exposure, with suppression of multiple pro-inflammatory mRNAs and signaling pathways including nuclear factor kappa B (NF-κB) and forkhead box O (FOXO). Studies with murine knock out cells revealed that silymarin inhibition of both mTOR and NF-κB was partially AMPK dependent, whereas silymarin inhibition of mTOR required DDIT4. Other natural products induced similar stress responses, which correlated with their ability to suppress inflammation. Thus, natural products activate stress and repair responses that culminate in an anti-inflammatory cellular phenotype. Natural products like silymarin may be useful as tools to define how metabolic, stress, and repair pathways regulate cellular inflammation.


Assuntos
Anti-Inflamatórios/farmacologia , Inflamação/tratamento farmacológico , Silybum marianum/química , Silimarina/farmacologia , Proteínas Quinases Ativadas por AMP/efeitos dos fármacos , Animais , Anti-Inflamatórios/química , Antioxidantes/farmacologia , Ciclo do Ácido Cítrico/efeitos dos fármacos , Fatores de Transcrição Forkhead/efeitos dos fármacos , Humanos , Inflamação/metabolismo , Células Jurkat , Fígado/metabolismo , Camundongos , Estrutura Molecular , NF-kappa B/antagonistas & inibidores , NF-kappa B/efeitos dos fármacos , Óxido Nítrico Sintase Tipo II , Transdução de Sinais/efeitos dos fármacos , Silimarina/química , Linfócitos T/metabolismo
11.
Int J Cancer ; 134(4): 778-88, 2014 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-23929039

RESUMO

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.


Assuntos
Carcinoma Hepatocelular/patologia , Fígado Gorduroso/patologia , Células Estreladas do Fígado/patologia , Neoplasias Hepáticas/patologia , Linfocinas/metabolismo , Comunicação Parácrina , Fator de Crescimento Derivado de Plaquetas/metabolismo , Células Estromais/patologia , Animais , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Western Blotting , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/metabolismo , Estudos de Coortes , Citocinas/genética , Citocinas/metabolismo , Fígado Gorduroso/genética , Fígado Gorduroso/metabolismo , Imunofluorescência , Perfilação da Expressão Gênica , Células Estreladas do Fígado/metabolismo , Hepatócitos/metabolismo , Hepatócitos/patologia , Humanos , Técnicas Imunoenzimáticas , Inflamação/genética , Inflamação/metabolismo , Inflamação/patologia , Fígado/metabolismo , Fígado/patologia , Cirrose Hepática/genética , Cirrose Hepática/metabolismo , Cirrose Hepática/patologia , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Linfocinas/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Hepatopatia Gordurosa não Alcoólica , Análise de Sequência com Séries de Oligonucleotídeos , Fator de Crescimento Derivado de Plaquetas/genética , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Células Estromais/metabolismo
12.
Drug Metab Dispos ; 41(2): 332-42, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23175668

RESUMO

Pregnancy-induced changes in drug pharmacokinetics can be explained by changes in expression of drug-metabolizing enzymes and transporters and/or normal physiology. In this study, we determined gestational age-dependent expression profiles for all metabolic enzyme and transporter genes in the maternal liver, kidney, small intestine, and placenta of pregnant mice by microarray analysis. We specifically examined the expression of genes important for xenobiotic, bile acid, and steroid hormone metabolism and disposition, namely, cytochrome P450s (Cyp), UDP-glucuronosyltranserases (Ugt), sulfotransferases (Sult), and ATP-binding cassette (Abc), solute carrier (Slc), and solute carrier organic anion (Slco) transporters. Few Ugt and Sult genes were affected by pregnancy. Cyp17a1 expression in the maternal liver increased 3- to 10-fold during pregnancy, which was the largest observed change in the maternal tissues. Cyp1a2, most Cyp2 isoforms, Cyp3a11, and Cyp3a13 expression in the liver decreased on gestation days (gd) 15 and 19 compared with nonpregnant controls (gd 0). In contrast, Cyp2d40, Cyp3a16, Cyp3a41a, Cyp3a41b, and Cyp3a44 in the liver were induced throughout pregnancy. In the placenta, Cyp expression on gd 10 and 15 was upregulated compared with gd 19. Notable changes were also observed in Abc and Slc transporters. Abcc3 expression in the liver and Abcb1a, Abcc4, and Slco4c1 expression in the kidney were downregulated on gd 15 and 19. In the placenta, Slc22a3 (Oct3) expression on gd 10 was 90% lower than that on gd 15 and 19. This study demonstrates important gestational age-dependent expression of metabolic enzyme and transporter genes, which may have mechanistic relevance to drug disposition in human pregnancy.


Assuntos
Transportadores de Cassetes de Ligação de ATP/metabolismo , Sistema Enzimático do Citocromo P-450/metabolismo , Glucuronosiltransferase/metabolismo , Intestino Delgado/enzimologia , Rim/enzimologia , Fígado/enzimologia , Transportadores de Ânions Orgânicos/metabolismo , Placenta/enzimologia , Transportadores de Cassetes de Ligação de ATP/genética , Animais , Sistema Enzimático do Citocromo P-450/genética , Feminino , Perfilação da Expressão Gênica/métodos , Regulação Enzimológica da Expressão Gênica , Idade Gestacional , Glucuronosiltransferase/genética , Isoenzimas , Camundongos , Análise de Sequência com Séries de Oligonucleotídeos , Transportadores de Ânions Orgânicos/genética , Gravidez , Reação em Cadeia da Polimerase em Tempo Real , Reprodutibilidade dos Testes , Especificidade por Substrato , Sulfotransferases/genética , Sulfotransferases/metabolismo
13.
Arterioscler Thromb Vasc Biol ; 32(12): 2910-8, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23042816

RESUMO

OBJECTIVE: Cholesterol accumulation by macrophages plays a key role in atherogenesis. To begin to develop a global picture of this process, we used proteomics and transcriptomics to analyze foam cells generated with acetyl-low-density lipoprotein, a classic ligand for scavenger receptors. METHODS AND RESULTS: Tandem mass spectrometry and stringent statistical analysis revealed that foam cells differentially expressed 15 of 542 proteins (2.8%) detected in macrophage-conditioned medium. Apolipoprotein E was one of the most upregulated proteins, confirming that proteins involved in lipid metabolism are important targets for regulation by sterol accumulation. However, levels of proteins linked to complement activation and lysosomal proteolysis also changed markedly. Transcriptional analysis demonstrated that 698 of 19,700 genes (3.5%) were regulated in foam cells, including many genes important in sterol metabolism. We also found that cholesterol accumulation regulated genes implicated in complement activation but failed to affect genes linked to proteolysis and macrophage polarization. Changes in protein levels in macrophage-conditioned medium were largely independent of changes in mRNA levels. CONCLUSIONS: Loading sterol into macrophages regulates levels of complement proteins and lysosomal proteases-key players in the immune system and plaque rupture. Posttranscriptional mechanisms are likely important for controlling levels of most of the proteins detected in macrophage medium.


Assuntos
Colesterol/metabolismo , Ativação do Complemento/fisiologia , Proteínas do Sistema Complemento/metabolismo , Células Espumosas/metabolismo , Proteínas de Membrana Lisossomal/metabolismo , Macrófagos/metabolismo , Proteólise , Animais , Apolipoproteínas E/metabolismo , Células Cultivadas , Células Espumosas/citologia , Perfilação da Expressão Gênica , Macrófagos/citologia , Camundongos , Camundongos Endogâmicos C57BL , Modelos Animais , Peptídeo Hidrolases/metabolismo , Proteômica , RNA Mensageiro/metabolismo
14.
Environ Sci Technol ; 47(13): 7466-74, 2013 Jul 02.
Artigo em Inglês | MEDLINE | ID: mdl-23745839

RESUMO

Although environmental trace metals, such as copper (Cu), can disrupt normal olfactory function in fish, the underlying molecular mechanisms of metal-induced olfactory injury have not been elucidated. Current research has suggested the involvement of epigenetic modifications. To address this hypothesis, we analyzed microRNA (miRNA) profiles in the olfactory system of Cu-exposed zebrafish. Our data revealed 2, 10, and 28 differentially expressed miRNAs in a dose-response manner corresponding to three increasing Cu concentrations. Numerous deregulated miRNAs were involved in neurogenesis (e.g., let-7, miR-7a, miR-128, and miR-138), indicating a role for Cu-mediated toxicity via interference with neurogenesis processes. Putative gene targets of deregulated miRNAs were identified when interrogating our previously published microarray database, including those involved in cell growth and proliferation, cell death, and cell morphology. Moreover, several miRNAs (e.g., miR-203a, miR-199*, miR-16a, miR-16c, and miR-25) may contribute to decreased mRNA levels of their host genes involved in olfactory signal transduction pathways and other critical neurological processes via a post-transcriptional mechanism. Our findings provide novel insight into the epigenetic regulatory mechanisms of metal-induced neurotoxicity of the fish olfactory system and identify novel miRNA biomarkers of metal exposures.


Assuntos
Cobre/toxicidade , MicroRNAs/metabolismo , Condutos Olfatórios/efeitos dos fármacos , Poluentes Químicos da Água/toxicidade , Animais , MicroRNAs/genética , Neurogênese/efeitos dos fármacos , Condutos Olfatórios/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , Peixe-Zebra
15.
Part Fibre Toxicol ; 10: 7, 2013 Mar 26.
Artigo em Inglês | MEDLINE | ID: mdl-23531317

RESUMO

BACKGROUND: Exposure to traffic-related air pollution (TRAP) is considered a trigger for acute cardiovascular events. Diesel Exhaust (DE) is a major contributor to TRAP in the world. We evaluated the effect of DE inhalation on circulating blood cell populations, hematological indices, and systemic inflammatory cytokines in humans using a specialized facility. METHODS: In a randomized double-blind crossover study balanced to order, 17 metabolic syndrome (MetS) and 15 healthy subjects inhaled filtered air (FA) or DE exposure in two-hour sessions on different days with a minimum 2-week washout period. We collected blood pre-exposure, 7, and 22 hours after exposure initiation and measured the complete blood count and differential. We performed multiplex cytokine assay to measure the changes in the systemic inflammatory cytokines, and endothelial adhesion molecules (n=15). A paired analysis compared the effect of DE and FA exposures for the change from pre-exposure to the subsequent time points. RESULTS: A significant increase in the hematocrit was noted 7 hrs after DE [1.4% (95% CI: 0.9 to 1.9%)] compared to FA exposure [0.5% (95% CI: -0.09 to 1.0%); p=0.008. The hemoglobin levels increased non-significantly at 7 hrs post DE [0.3 gm/dL (95% CI: 0.2 to 0.5 gm/dL)] versus FA exposure [0.2 gm/dL (95% CI: 0 to 0.3 gm/dL)]; p=0.06. Furthermore, the platelet count increased 22 hrs after DE exposure in healthy, but not in MetS subjects [DE: 16.6 (95% CI: 10.2 to 23) thousand platelets/mL versus [FA: 3.4 (95% CI: -9.5 to 16.3) thousand platelets/mL)]; p=0.04. No DE effect was observed for WBC, neutrophils, lymphocytes or erythrocytes. Using the multiplex assay, small borderline significant increases in matrix metalloproteinase-9, interleukins (IL)-1 beta, 6 and 10 occurred 7 hrs post exposure initiation, whereas E-selectin, intercellular adhesion molecule-1, and vascular cell adhesion molecule -1, and myeloperoxidase 22 hrs post exposure. CONCLUSIONS: Our results suggest that short-term DE exposure results in hemoconcentration and thrombocytosis, which are important determinants of acute cardiovascular events. Multiplex assay showed a non-significant increase in IL-1ß and IL-6 immediately post exposure followed by myeloperoxidase and endothelial activation molecules. Further specific assays in a larger population will improve our understanding of the systemic inflammatory mechanisms following acute exposure to TRAP.


Assuntos
Moléculas de Adesão Celular/sangue , Citocinas/sangue , Endotélio Vascular/efeitos dos fármacos , Mediadores da Inflamação/sangue , Exposição por Inalação/efeitos adversos , Síndrome Metabólica/sangue , Emissões de Veículos/toxicidade , Adulto , Biomarcadores/sangue , Contagem de Células Sanguíneas , Estudos Cross-Over , Método Duplo-Cego , Endotélio Vascular/imunologia , Endotélio Vascular/metabolismo , Feminino , Hematócrito , Hemoglobinas/metabolismo , Humanos , Masculino , Síndrome Metabólica/imunologia , Trombocitose/sangue , Trombocitose/induzido quimicamente , Fatores de Tempo , Washington , Adulto Jovem
16.
Proc Natl Acad Sci U S A ; 107(29): 13129-34, 2010 Jul 20.
Artigo em Inglês | MEDLINE | ID: mdl-20615956

RESUMO

Areas and layers of the cerebral cortex are specified by genetic programs that are initiated in progenitor cells and then, implemented in postmitotic neurons. Here, we report that Tbr1, a transcription factor expressed in postmitotic projection neurons, exerts positive and negative control over both regional (areal) and laminar identity. Tbr1 null mice exhibited profound defects of frontal cortex and layer 6 differentiation, as indicated by down-regulation of gene-expression markers such as Bcl6 and Cdh9. Conversely, genes that implement caudal cortex and layer 5 identity, such as Bhlhb5 and Fezf2, were up-regulated in Tbr1 mutants. Tbr1 implements frontal identity in part by direct promoter binding and activation of Auts2, a frontal cortex gene implicated in autism. Tbr1 regulates laminar identity in part by downstream activation or maintenance of Sox5, an important transcription factor controlling neuronal migration and corticofugal axon projections. Similar to Sox5 mutants, Tbr1 mutants exhibit ectopic axon projections to the hypothalamus and cerebral peduncle. Together, our findings show that Tbr1 coordinately regulates regional and laminar identity of postmitotic cortical neurons.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Mitose , Neocórtex/citologia , Neocórtex/embriologia , Neurônios/citologia , Animais , Biomarcadores/metabolismo , Proteínas do Citoesqueleto , Proteínas de Ligação a DNA/genética , Regulação para Baixo/genética , Regulação da Expressão Gênica no Desenvolvimento , Camundongos , Mutação/genética , Neocórtex/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , Especificidade de Órgãos , Ligação Proteica , Proteínas com Domínio T , Fatores de Transcrição , Ativação Transcricional , Regulação para Cima/genética
17.
Cell Metab ; 4(5): 349-62, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17055784

RESUMO

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.


Assuntos
Tecido Adiposo Marrom/fisiopatologia , Regulação da Temperatura Corporal/genética , Proteínas de Choque Térmico/metabolismo , Doença de Huntington/etiologia , Fatores de Transcrição/metabolismo , Animais , Temperatura Corporal/genética , Células Cultivadas , Modelos Animais de Doenças , Proteínas de Choque Térmico/genética , Doença de Huntington/genética , Doença de Huntington/metabolismo , Camundongos , Camundongos Transgênicos , Mitocôndrias/fisiologia , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo , Transdução de Sinais/genética , Fatores de Transcrição/genética , Transcrição Gênica
18.
Circulation ; 122(19): 1919-27, 2010 Nov 09.
Artigo em Inglês | MEDLINE | ID: mdl-20974999

RESUMO

BACKGROUND: High-density lipoprotein (HDL) protects the artery wall by removing cholesterol from lipid-laden macrophages. However, recent evidence suggests that HDL might also inhibit atherogenesis by combating inflammation. METHODS AND RESULTS: To identify potential antiinflammatory mechanisms, we challenged macrophages with lipopolysaccharide, an inflammatory microbial ligand for Toll-like receptor 4. HDL inhibited the expression of 30 (277 of 911) of the genes normally induced by lipopolysaccharide, microarray analysis revealed. One of its major targets was the type I interferon response pathway, a family of potent viral immunoregulators controlled by Toll-like receptor 4 and the TRAM/TRIF signaling pathway. Unexpectedly, the ability of HDL to inhibit gene expression was independent of macrophage cholesterol stores. Immunofluorescent studies suggested that HDL promoted TRAM translocation to intracellular compartments, which impaired subsequent signaling by Toll-like receptor 4 and TRIF. To examine the potential in vivo relevance of the pathway, we used mice deficient in apolipoprotein A-I, the major protein of HDL. After infection with Salmonella typhimurium, a Gram-negative bacterium that expresses lipopolysaccharide, apolipoprotein A-I-deficient mice had 6-fold higher plasma levels of interferon-ß, a key regulator of the type I interferon response, than did wild-type mice. CONCLUSIONS: HDL inhibits a subset of lipopolysaccharide-stimulated macrophage genes that regulate the type I interferon response, and its action is independent of sterol metabolism. These findings raise the possibility that regulation of macrophage genes by HDL might link innate immunity and cardioprotection.


Assuntos
Interferon Tipo I/imunologia , Lipopolissacarídeos/farmacologia , Lipoproteínas HDL/farmacologia , Macrófagos/imunologia , Animais , Quimiocina CXCL10/metabolismo , Quimiocinas/genética , Citocinas/genética , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/fisiologia , Terapia de Imunossupressão , Interferon beta/metabolismo , Interleucina-12/metabolismo , Macrófagos/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , RNA Mensageiro/genética , Transdução de Sinais/fisiologia , Tioglicolatos/farmacologia , Receptor 4 Toll-Like/agonistas , Receptor 4 Toll-Like/genética , Receptores Toll-Like/genética
19.
Mass Spectrom Rev ; 29(1): 79-125, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-19358229

RESUMO

Protein glycosylation regulates protein function and cellular distribution. Additionally, aberrant protein glycosylations have been recognized to play major roles in human disorders, including neurodegenerative diseases. Glycoproteomics, a branch of proteomics that catalogs and quantifies glycoproteins, provides a powerful means to systematically profile the glycopeptides or glycoproteins of a complex mixture that are highly enriched in body fluids, and therefore, carry great potential to be diagnostic and/or prognostic markers. Application of this mass spectrometry-based technology to the study of neurodegenerative disorders (e.g., Alzheimer's disease and Parkinson's disease) is relatively new, and is expected to provide insight into the biochemical pathogenesis of neurodegeneration, as well as biomarker discovery. In this review, we have summarized the current understanding of glycoproteins in biology and neurodegenerative disease, and have discussed existing proteomic technologies that are utilized to characterize glycoproteins. Some of the ongoing studies, where glycoproteins isolated from cerebrospinal fluid and human brain are being characterized in Parkinson's disease at different stages versus controls, are presented, along with future applications of targeted validation of brain specific glycoproteins in body fluids.


Assuntos
Glicoproteínas/análise , Glicoproteínas/metabolismo , Espectrometria de Massas/métodos , Doenças Neurodegenerativas/metabolismo , Proteômica/métodos , Sequência de Aminoácidos , Glicoproteínas/líquido cefalorraquidiano , Glicosilação , Humanos , Espectrometria de Massas/tendências , Dados de Sequência Molecular , Doenças Neurodegenerativas/líquido cefalorraquidiano , Doenças Neurodegenerativas/diagnóstico , Proteômica/tendências
20.
Pediatr Res ; 69(4): 285-92, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21178816

RESUMO

Critically ill preterm infants experience multiple stressors while hospitalized. Morphine is commonly prescribed to ameliorate their pain and stress. We hypothesized that neonatal stress will have a dose-dependent effect on hippocampal gene expression, and these effects will be altered by morphine treatment. Male C57BL/6 mice were exposed to five treatment conditions between postnatal d 5 and 9: 1) control, 2) mild stress + saline, 3) mild stress + morphine, 4) severe stress + saline, and 5) severe stress + morphine. Hippocampal RNA was extracted and analyzed using Affymetrix Mouse Gene 1.0 ST Arrays. Single gene analysis and gene set analysis were used to compare groups with validation by qPCR. Stress resulted in enrichment of gene sets related to fear response, oxygen carrying capacity, and NMDA receptor synthesis. Morphine down-regulated gene sets related to immune function. Stress + morphine resulted in enrichment of mitochondrial electron transport gene sets and down-regulation of gene sets related to brain development and growth. We conclude that neonatal stress alone influences hippocampal gene expression, and morphine alters a subset of stress-related changes in gene expression and influences other gene sets. Stress + morphine show interaction effects not present with either stimulus alone. These changes may alter neurodevelopment.


Assuntos
Analgésicos Opioides/farmacologia , Animais Recém-Nascidos , Expressão Gênica/efeitos dos fármacos , Hipocampo/efeitos dos fármacos , Hipocampo/fisiologia , Morfina/farmacologia , Estresse Fisiológico , Animais , Relação Dose-Resposta a Droga , Hipocampo/citologia , Humanos , Recém-Nascido , Recém-Nascido Prematuro , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Análise em Microsséries , Neurônios/efeitos dos fármacos , Neurônios/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA