Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
eNeuro ; 7(1)2020.
Artigo em Inglês | MEDLINE | ID: mdl-31996391

RESUMO

Visceral sensory neurons encode distinct sensations from healthy organs and initiate pain states that are resistant to common analgesics. Transcriptome analysis is transforming our understanding of sensory neuron subtypes but has generally focused on somatic sensory neurons or the total population of neurons in which visceral neurons form the minority. Our aim was to define transcripts specifically expressed by sacral visceral sensory neurons, as a step towards understanding the unique biology of these neurons and potentially leading to identification of new analgesic targets for pelvic visceral pain. Our strategy was to identify genes differentially expressed between sacral dorsal root ganglia (DRG) that include somatic neurons and sacral visceral neurons, and adjacent lumbar DRG that comprise exclusively of somatic sensory neurons. This was performed in adult and E18.5 male and female mice. By developing a method to restrict analyses to nociceptive Trpv1 neurons, a larger group of genes were detected as differentially expressed between spinal levels. We identified many novel genes that had not previously been associated with pelvic visceral sensation or nociception. Limited sex differences were detected across the transcriptome of sensory ganglia, but more were revealed in sacral levels and especially in Trpv1 nociceptive neurons. These data will facilitate development of new tools to modify mature and developing sensory neurons and nociceptive pathways.


Assuntos
Gânglios Espinais , Transcriptoma , Animais , Feminino , Masculino , Camundongos , Nociceptividade , Nociceptores , Dor , Células Receptoras Sensoriais
2.
J Thromb Haemost ; 17(1): 169-182, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30472780

RESUMO

Essentials Tissue factor (TF) isoforms are expressed in pancreatic neuroendocrine tumors (pNET). TF knockdown inhibits proliferation of human pNET cells in vitro. mTOR kinase inhibitor sapanisertib/MLN0128 suppresses TF expression in human pNET cells. Sapanisertib suppresses TF expression and activity and reduces the growth of pNET tumors in vivo. SUMMARY: Background Full-length tissue factor (flTF) and alternatively spliced TF (asTF) contribute to growth and spread of pancreatic ductal adenocarcinoma. It is unknown, however, if flTF and/or asTF contribute to the pathobiology of pancreatic neuroendocrine tumors (pNETs). Objective To assess TF expression in pNETs and the effects of mTOR complex 1/2 (mTORC1/2) inhibition on pNET growth. Methods Human pNET specimens were immunostained for TF. Human pNET cell lines QGP1 and BON were evaluated for TF expression and responsiveness to mTOR inhibition. shRNA were used to knock down TF in BON. TF cofactor activity was assessed using a two-step FXa generation assay. TF promoter activity was assessed using transient transfection of human TF promoter-driven reporter constructs into cells. Mice bearing orthotopic BON tumors were treated with the mTORC1/2 ATP site competitive inhibitor sapanisertib/MLN0128 (3 mg kg-1 , oral gavage) for 34 days. Results Immunostaining of pNET tissue revealed flTF and asTF expression. BON and QGP1 expressed both TF isoforms, with BON exhibiting higher levels. shRNA directed against TF suppressed BON proliferation in vitro. Treatment of BON with sapanisertib inhibited mTOR signaling and suppressed TF levels. BON tumors grown in mice treated with sapanisertib had significantly less TF protein and cofactor activity, and were smaller compared with tumors grown in control mice. Conclusions TF isoforms are expressed in pNETs. Sapanisertib suppresses TF mRNA and protein expression as well as TF cofactor activity in vitro and in vivo. Thus, further studies are warranted to evaluate the clinical utility of TF-suppressing mTORC1/2 inhibitor sapanisertib in pNET management.


Assuntos
Antineoplásicos/farmacologia , Proliferação de Células/efeitos dos fármacos , Tumores Neuroendócrinos/tratamento farmacológico , Neoplasias Pancreáticas/tratamento farmacológico , Inibidores de Proteínas Quinases/farmacologia , Pirazóis/farmacologia , Pirimidinas/farmacologia , Serina-Treonina Quinases TOR/antagonistas & inibidores , Tromboplastina/metabolismo , Animais , Linhagem Celular Tumoral , Humanos , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Alvo Mecanístico do Complexo 2 de Rapamicina/metabolismo , Camundongos Nus , Tumores Neuroendócrinos/enzimologia , Tumores Neuroendócrinos/genética , Tumores Neuroendócrinos/patologia , Neoplasias Pancreáticas/enzimologia , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patologia , Regiões Promotoras Genéticas , Transdução de Sinais , Serina-Treonina Quinases TOR/metabolismo , Tromboplastina/genética , Carga Tumoral/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
3.
Nat Commun ; 9(1): 4559, 2018 11 19.
Artigo em Inglês | MEDLINE | ID: mdl-30455465

RESUMO

Epigenetic regulators are attractive anticancer targets, but the promise of therapeutic strategies inhibiting some of these factors has not been proven in vivo or taken into account tumor cell heterogeneity. Here we show that the histone methyltransferase G9a, reported to be a therapeutic target in many cancers, is a suppressor of aggressive lung tumor-propagating cells (TPCs). Inhibition of G9a drives lung adenocarcinoma cells towards the TPC phenotype by de-repressing genes which regulate the extracellular matrix. Depletion of G9a during tumorigenesis enriches tumors in TPCs and accelerates disease progression metastasis. Depleting histone demethylases represses G9a-regulated genes and TPC phenotypes. Demethylase inhibition impairs lung adenocarcinoma progression in vivo. Therefore, inhibition of G9a is dangerous in certain cancer contexts, and targeting the histone demethylases is a more suitable approach for lung cancer treatment. Understanding cellular context and specific tumor populations is critical when targeting epigenetic regulators in cancer for future therapeutic development.


Assuntos
Progressão da Doença , Histona Desmetilases/metabolismo , Histona Metiltransferases/metabolismo , Neoplasias Pulmonares/metabolismo , Adenocarcinoma de Pulmão/metabolismo , Animais , Carcinogênese , Linhagem Celular Tumoral/efeitos dos fármacos , Sobrevivência Celular , Modelos Animais de Doenças , Matriz Extracelular/genética , Histona Desmetilases/efeitos dos fármacos , Histona-Lisina N-Metiltransferase/efeitos dos fármacos , Histona-Lisina N-Metiltransferase/metabolismo , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , Organoides/anatomia & histologia , Fenótipo , Proteínas Proto-Oncogênicas p21(ras)/genética
4.
Blood ; 130(10): 1213-1222, 2017 09 07.
Artigo em Inglês | MEDLINE | ID: mdl-28710059

RESUMO

Understanding and blocking the self-renewal pathway of preleukemia stem cells could prevent acute myeloid leukemia (AML) relapse. In this study, we show that increased FOXO1 represents a critical mechanism driving aberrant self-renewal in preleukemic cells expressing the t(8;21)-associated oncogene AML1-ETO (AE). Although generally considered as a tumor suppressor, FOXO1 is consistently upregulated in t(8;21) AML. Expression of FOXO1 in human CD34+ cells promotes a preleukemic state with enhanced self-renewal and dysregulated differentiation. The DNA binding domain of FOXO1 is essential for these functions. FOXO1 activates a stem cell molecular signature that is also present in AE preleukemia cells and preserved in t(8;21) patient samples. Genome-wide binding studies show that AE and FOXO1 share the majority of their binding sites, whereby FOXO1 binds to multiple crucial self-renewal genes and is required for their activation. In agreement with this observation, genetic and pharmacological ablation of FOXO1 inhibited the long-term proliferation and clonogenicity of AE cells and t(8;21) AML cell lines. Targeting of FOXO1 therefore provides a potential therapeutic strategy for elimination of stem cells at both preleukemic and leukemic stages.


Assuntos
Subunidade alfa 2 de Fator de Ligação ao Core/metabolismo , Proteína Forkhead Box O1/metabolismo , Redes Reguladoras de Genes , Leucemia Mieloide Aguda/genética , Proteínas de Fusão Oncogênica/metabolismo , Lesões Pré-Cancerosas/genética , Animais , Antígenos CD34/metabolismo , Linhagem Celular Tumoral , Proliferação de Células , Subunidade alfa 2 de Fator de Ligação ao Core/genética , Perfilação da Expressão Gênica , Regulação Leucêmica da Expressão Gênica , Genoma Humano , Células-Tronco Hematopoéticas/metabolismo , Humanos , Leucemia Mieloide Aguda/patologia , Camundongos SCID , Proteínas de Fusão Oncogênica/genética , Lesões Pré-Cancerosas/patologia , Proteína 1 Parceira de Translocação de RUNX1 , Regulação para Cima/genética
5.
Genes Immun ; 15(6): 361-9, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24920534

RESUMO

Eosinophilic esophagitis (EoE) is an allergic inflammatory disorder of the esophagus that is compounded by genetic predisposition and hypersensitivity to environmental antigens. Using high-density oligonucleotide expression chips, a disease-specific esophageal transcript signature was identified and was shown to be largely reversible with therapy. In an effort to expand the molecular signature of EoE, we performed RNA sequencing on esophageal biopsies from healthy controls and patients with active EoE and identified a total of 1607 significantly dysregulated transcripts (1096 upregulated, 511 downregulated). When clustered by raw expression levels, an abundance of immune cell-specific transcripts are highly induced in EoE but expressed at low (or undetectable) levels in healthy controls. Moreover, 66% of the gene signature identified by RNA sequencing was previously unrecognized in the EoE transcript signature by microarray-based expression profiling and included several long non-coding RNAs (lncRNA), an emerging class of transcriptional regulators. The lncRNA BRAF-activated non-protein coding RNA (BANCR) was upregulated in EoE and induced in interleukin-13 (IL-13)-treated primary esophageal epithelial cells. Repression of BANCR significantly altered the expression of IL-13-induced proinflammatory genes. Together, these data comprise new potential biomarkers of EoE and demonstrate a novel role for lncRNAs in EoE and IL-13-associated responses.


Assuntos
Esofagite Eosinofílica/genética , Análise de Sequência com Séries de Oligonucleotídeos/métodos , Análise de Sequência de RNA/métodos , Transcriptoma , Linhagem Celular , Células Cultivadas , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Humanos , Interleucina-13/farmacologia , Interferência de RNA , RNA não Traduzido/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Regulação para Cima
6.
Mucosal Immunol ; 5(4): 388-96, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22453679

RESUMO

Interleukin 13 (IL-13)-induced epithelial gene and protein expression changes are central to the pathogenesis of multiple allergic diseases. Herein, using human esophageal squamous and bronchial columnar epithelial cells, we identified microRNAs (miRNAs) that were differentially regulated after IL-13 stimulation. Among the IL-13-regulated miRNAs, miR-375 showed a conserved pattern of downregulation. Furthermore, miR-375 was downregulated in the lung of IL-13 lung transgenic mice. We subsequently analyzed miR-375 levels in a human disease characterized by IL-13 overproduction--the allergic disorder eosinophilic esophagitis (EE)--and observed downregulation of miR-375 in EE patient samples compared with control patients. MiR-375 expression levels reflected disease activity, normalized with remission, and inversely correlated with the degree of allergic inflammation. Using a lentiviral strategy and whole-transcriptome analysis in epithelial cells, miR-375 overexpression was sufficient to markedly modify IL-13-associated immunoinflammatory pathways in epithelial cells in vitro, further substantiating interactions between miR-375 and IL-13. Taken together, our results support a key role of miRNAs, particularly miR-375, in regulating and fine-tuning IL-13-mediated responses.


Assuntos
Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Fatores Imunológicos/farmacologia , Interleucina-13/farmacologia , MicroRNAs/genética , Transcriptoma , Animais , Linhagem Celular , Análise por Conglomerados , Esofagite Eosinofílica/genética , Esôfago/metabolismo , Perfilação da Expressão Gênica , Humanos , Camundongos , Camundongos Transgênicos , Mucosa Respiratória/efeitos dos fármacos , Mucosa Respiratória/metabolismo
7.
J Thromb Haemost ; 9(10): 2087-96, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21812913

RESUMO

BACKGROUND: TF is highly expressed in cancerous and atherosclerotic lesions. Monocyte recruitment is a hallmark of disease progression in these pathological states. OBJECTIVE: To examine the role of integrin signaling in TF-dependent recruitment of monocytes by endothelial cells. METHODS: The expression of flTF and asTF in cervical cancer and atherosclerotic lesions was examined. Biologic effects of the exposure of primary microvascular endothelial cells (MVEC) to truncated flTF ectodomain (LZ-TF) and recombinant asTF were assessed. RESULTS: flTF and asTF exhibited nearly identical expression patterns in cancer lesions and lipid-rich plaques. Tumor lesions, as well as stromal CD68(+) monocytes/macrophages, expressed both TF forms. Primary MVEC rapidly adhered to asTF and LZ-TF, and this was completely blocked by anti-ß1 integrin antibody. asTF- and LZ-TF-treatment of MVEC promoted adhesion of peripheral blood mononuclear cells (PBMCs) under orbital shear conditions and under laminar flow; asTF-elicited adhesion was more pronounced than that elicited by LZ-TF. Expression profiling and western blotting revealed a broad activation of cell adhesion molecules (CAMs) in MVEC following asTF treatment including E-selectin, ICAM-1 and VCAM-1. In transwell assays, asTF potentiated PMBC migration through MVEC monolayers by ∼3-fold under MCP-1 gradient. CONCLUSIONS: TF splice variants ligate ß1 integrins on MVEC, which induces the expression of CAMs in MVEC and leads to monocyte adhesion and transendothelial migration. asTF appears more potent than flTF in eliciting these effects. Our findings underscore the pathophysiologic significance of non-proteolytic, integrin-mediated signaling by the two naturally occurring TF variants in cancer and atherosclerosis.


Assuntos
Processamento Alternativo , Moléculas de Adesão Celular/metabolismo , Endotélio Vascular/citologia , Integrinas/metabolismo , Monócitos/citologia , Transdução de Sinais , Tromboplastina/genética , Western Blotting , Células Cultivadas , Feminino , Humanos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Neoplasias do Colo do Útero/metabolismo , Neoplasias do Colo do Útero/patologia
8.
Cancer Gene Ther ; 15(11): 733-41, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18551144

RESUMO

Oncolytic herpes simplex viruses (oHSVs) are promising anticancer therapeutics. We sought to characterize the functional genomic response of human cancer cells to oHSV infection using G207, an oHSV previously evaluated in a phase I trial. Five human malignant peripheral nerve sheath tumor cell lines, with differing sensitivity to oHSV, were infected with G207 for 6 h. Functional genomic analysis of virus-infected cells demonstrated large clusters of downregulated cellular mRNAs and smaller clusters of those upregulated, including 21 genes commonly upregulated in all five lines. Of these, 7 are known to be HSV-1 induced and 14 represent novel virus-regulated genes. Gene ontology analysis revealed that a majority of G207-upregulated genes are involved in Janus kinase/signal transducer and activator of transcription signaling, transcriptional regulation, nucleic acid metabolism, protein synthesis and apoptosis. Ingenuity networks highlighted nodes for AP-1 subunits and interferon signaling via STAT1, suppressor of cytokine signaling-1 (SOCS1), SOCS3 and RANTES. As biological confirmation, we found that virus-mediated upregulation of SOCS1 correlated with sensitivity to G207 and that depletion of SOCS1 impaired virus replication by >10-fold. Further characterization of roles provided by oHSV-induced cellular genes during virus replication may be utilized to predict oncolytic efficacy and to provide rational strategies for designing next-generation oncolytic viruses.


Assuntos
Regulação Neoplásica da Expressão Gênica/genética , Herpesvirus Humano 1/fisiologia , Neoplasias de Bainha Neural/terapia , Neoplasias de Bainha Neural/virologia , Terapia Viral Oncolítica/métodos , Proteínas Supressoras da Sinalização de Citocina/metabolismo , Replicação Viral/fisiologia , Western Blotting , Herpesvirus Humano 1/genética , Humanos , Análise de Sequência com Séries de Oligonucleotídeos , Proteína 1 Supressora da Sinalização de Citocina , Proteínas Supressoras da Sinalização de Citocina/genética , Replicação Viral/genética
9.
Oncogene ; 26(43): 6307-18, 2007 Sep 20.
Artigo em Inglês | MEDLINE | ID: mdl-17452985

RESUMO

Functional inactivation of the retinoblastoma tumor suppressor gene product (RB) is a common event in human cancers. Classically, RB functions to constrain cellular proliferation, and loss of RB is proposed to facilitate the hyperplastic proliferation associated with tumorigenesis. To understand the repertoire of regulatory processes governed by RB, two models of RB loss were utilized to perform microarray analysis. In murine embryonic fibroblasts harboring germline loss of RB, there was a striking deregulation of gene expression, wherein distinct biological pathways were altered. Specifically, genes involved in cell cycle control and classically associated with E2F-dependent gene regulation were upregulated via RB loss. In contrast, a program of gene expression associated with immune function and response to pathogens was significantly downregulated with the loss of RB. To determine the specific influence of RB loss during a defined period and without the possibility of developmental compensation as occurs in embryonic fibroblasts, a second system was employed wherein Rb was acutely knocked out in adult fibroblasts. This model confirmed the distinct regulation of cell cycle and immune modulatory genes through RB loss. Analyses of cis-elements supported the hypothesis that the majority of those genes upregulated with RB loss are regulated via the E2F family of transcription factors. In contrast, those genes whose expression was reduced with the loss of RB harbored different promoter elements. Consistent with these analyses, we found that disruption of E2F-binding function of RB was associated with the upregulation of gene expression. In contrast, cells harboring an RB mutant protein (RB-750F) that retains E2F-binding activity, but is specifically deficient in the association with LXCXE-containing proteins, failed to upregulate these same target genes. However, downregulation of genes involved in immune function was readily observed with disruption of the LXCXE-binding function of RB. Thus, these studies demonstrate that RB plays a significant role in both the positive and negative regulations of transcriptional programs and indicate that loss of RB has distinct biological effects related to both cell cycle control and immune function.


Assuntos
Ciclo Celular , Proteína do Retinoblastoma/deficiência , Proteína do Retinoblastoma/metabolismo , Retinoblastoma/metabolismo , Retinoblastoma/patologia , Transcrição Gênica/genética , Animais , Células Cultivadas , Regulação para Baixo , Fatores de Transcrição E2F/genética , Fatores de Transcrição E2F/metabolismo , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Camundongos , Camundongos Knockout , Regiões Promotoras Genéticas/genética , Retinoblastoma/genética , Retinoblastoma/imunologia , Proteína do Retinoblastoma/genética , Regulação para Cima
10.
Neuroreport ; 15(11): 1735-8, 2004 Aug 06.
Artigo em Inglês | MEDLINE | ID: mdl-15257138

RESUMO

Clusterin accumulates in significant quantity in prion protein lesions associated with bovine spongiform encephalopathy (BSE) and we therefore sought to elucidate its ability to alter BSE pathogenesis and incubation time by comparison of wild type C57BL/6J mice and clusterin knock out (ko) mice. The ko mice had a 40 day increase in mean incubation time compared to wild type mice. PrP deposition in the medulla was less aggregated in clusterin knock out mice when compared to wild type BSE infected mice and a more marked astrocytosis, as determined by GFAP staining, was evident. The vacuolation profiles did not differ between the two strains of mice. Taken together these results suggest that clusterin alters the extracellular deposition of PrP(BSE) and accelerates BSE pathogenesis.


Assuntos
Encefalopatia Espongiforme Bovina/metabolismo , Encefalopatia Espongiforme Bovina/patologia , Glicoproteínas/fisiologia , Chaperonas Moleculares/fisiologia , Animais , Bovinos , Clusterina , Feminino , Glicoproteínas/deficiência , Glicoproteínas/genética , Masculino , Bulbo/metabolismo , Bulbo/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Chaperonas Moleculares/genética
11.
Rheumatology (Oxford) ; 43(8): 973-9, 2004 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15150433

RESUMO

OBJECTIVE: To evaluate the ability of microarray-based methods to identify genes with disease-specific expression patterns in peripheral blood mononuclear cells (PBMC) and synovial fluid mononuclear cells (SFMC) of juvenile arthritis patients and healthy controls. METHODS: Microarray data (Affymetrix U95Av2) from 26 PBMC and 20 SFMC samples collected from patients with active disease (classified by course according to ACR criteria) were analysed for expression patterns that correlated with disease characteristics. For comparison, PBMC gene expression profiles were obtained from 15 healthy controls. Real-time PCR was used for confirmation of gene expression differences. RESULTS: Statistical analysis of gene expression patterns in PBMC identified 378 probe sets corresponding to 342 unique genes with differing expression levels between polyarticular course patients and controls (t test, P<0.0001). The genes represented by these probe sets were enriched for functions related to regulation of immune cell functions, receptor signalling as well as protein metabolism and degradation. Included in these probe sets were a group of CXCL chemokines with functions related to angiogenesis. Further analysis showed that, whereas angiogenic CXCL (ELR+) gene expression was elevated in polyarticular PBMC, expression of angiostatic CXCL (ELR-) chemokines was lower in polyarticular SFMC compared with corresponding pauciarticular samples (t test, P<0.05). CONCLUSIONS: This pilot study demonstrates that juvenile arthritis patients exhibit complex patterns of gene expression in PBMC and SFMC. The presence of disease-correlated biologically relevant gene expression patterns suggests that the power of this approach will allow better understanding of disease mechanisms, identify distinct clinical phenotypes in disease subtypes, and suggest new therapeutic approaches.


Assuntos
Artrite Juvenil/genética , Quimiocinas CXC/genética , Expressão Gênica/genética , Leucócitos Mononucleares/fisiologia , Espondiloartropatias/genética , Líquido Sinovial/fisiologia , Adolescente , Adulto , Células Cultivadas , Criança , Perfilação da Expressão Gênica/métodos , Humanos , Neovascularização Patológica/genética , Análise de Sequência com Séries de Oligonucleotídeos/métodos , Projetos Piloto , Proteínas Tirosina Quinases/genética , Estudos Retrospectivos , Transdução de Sinais/genética , Transativadores/genética
12.
Physiol Genomics ; 6(2): 105-16, 2001 Jul 17.
Artigo em Inglês | MEDLINE | ID: mdl-11459926

RESUMO

Placental development results from a highly dynamic differentiation program. We used DNA microarray analysis to characterize the process by which human cytotrophoblast cells differentiate into syncytiotrophoblast cells in a purified cell culture system. Of 6,918 genes analyzed, 141 genes were induced and 256 were downregulated by more than 2-fold. Dynamically regulated genes were divided by the K-means algorithm into 9 kinetic pattern groups, then by biologic classification into 6 overall functional categories: cell and tissue structural dynamics, cell cycle and apoptosis, intercellular communication, metabolism, regulation of gene expression, and expressed sequence tag (EST) and function unknown. Gene expression changes within key functional categories were tightly coupled to morphological changes. In several key gene function categories, such as cell and tissue structure, many gene members of the category were strongly activated while others were strongly repressed. These findings suggest that differentiation is augmented by "categorical reprogramming" in which the function of induced genes is enhanced by preventing the further synthesis of categorically related gene products.


Assuntos
Transcrição Gênica , Trofoblastos/fisiologia , Diferenciação Celular , Células Cultivadas , Gonadotropina Coriônica/farmacologia , Feminino , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Humanos , Cinética , Modelos Biológicos , Análise de Sequência com Séries de Oligonucleotídeos , RNA Mensageiro/biossíntese , Trofoblastos/efeitos dos fármacos
13.
Physiol Genomics ; 6(1): 19-28, 2001 Jun 06.
Artigo em Inglês | MEDLINE | ID: mdl-11395543

RESUMO

To define molecular mechanisms of cardiac hypertrophy, genes whose expression was perturbed by any of four different transgenic mouse hypertrophy models [protein kinase C-epsilon activation peptide (PsiepsilonRACK), calsequestrin (CSQ), calcineurin (CN), and Galpha(q)] were compared by DNA microarray analyses using the approximately 8,800 genes present on the Incyte mouse GEM1. The total numbers of regulated genes (tens to hundreds) correlated with phenotypic severity of the model (Galpha(q) > CN > CSQ > PsiepsilonRACK), but demonstrated that no single gene was consistently upregulated. Of the three models exhibiting pathological hypertrophy, only atrial natriuretic peptide was consistently upregulated, suggesting that transcriptional alterations are highly specific to individual genetic causes of hypertrophy. However, hierarchical-tree and K-means clustering analyses revealed that subsets of the upregulated genes did exhibit coordinate regulatory patterns that were unique or overlapping across the different hypertrophy models. One striking set consisted of apoptotic genes uniquely regulated in the apoptosis-prone Galpha(q) model. Thus, rather than identifying a single common hypertrophic cardiomyopathy gene program, these data suggest that extensive groups of genes may be useful for the prediction of specific underlying genetic determinants and condition-specific therapeutic approaches.


Assuntos
Cardiomegalia/genética , Animais , Apoptose/genética , Calcineurina/genética , Calsequestrina/genética , Cardiomegalia/metabolismo , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP , Perfilação da Expressão Gênica , Proteínas Heterotriméricas de Ligação ao GTP/genética , Isoenzimas/metabolismo , Camundongos , Camundongos Transgênicos , Análise de Sequência com Séries de Oligonucleotídeos , Proteína Quinase C/metabolismo , Proteína Quinase C-épsilon , RNA Mensageiro/biossíntese , Transcrição Gênica , Regulação para Cima
14.
Nat Med ; 7(3): 338-43, 2001 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-11231633

RESUMO

Clusterin, also known as apolipoprotein J, is a ubiquitously expressed molecule thought to influence a variety of processes including cell death. In the brain, it accumulates in dying neurons following seizures and hypoxic-ischemic (H-I) injury. Despite this, in vivo evidence that clusterin directly influences cell death is lacking. Following neonatal H-I brain injury in mice (a model of cerebral palsy), there was evidence of apoptotic changes (neuronal caspase-3 activation), as well as accumulation of clusterin in dying neurons. Clusterin-deficient mice had 50% less brain injury following neonatal H-I. Surprisingly, the absence of clusterin had no effect on caspase-3 activation, and clusterin accumulation and caspase-3 activation did not colocalize to the same cells. Studies with cultured cortical neurons demonstrated that exogenous purified astrocyte-secreted clusterin exacerbated oxygen/glucose-deprivation-induced necrotic death. These results indicate that clusterin may be a new therapeutic target to modulate non-caspase-dependent neuronal death following acute brain injury.


Assuntos
Encéfalo/patologia , Caspases/metabolismo , Glicoproteínas/fisiologia , Hipóxia-Isquemia Encefálica/patologia , Chaperonas Moleculares/fisiologia , Animais , Animais Recém-Nascidos , Western Blotting , Caspase 3 , Morte Celular/fisiologia , Clusterina , Imunofluorescência , Glicoproteínas/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microscopia Imunoeletrônica , Chaperonas Moleculares/genética
15.
Physiol Genomics ; 7(2): 135-48, 2001 Dec 21.
Artigo em Inglês | MEDLINE | ID: mdl-11773600

RESUMO

Gene induction and categorical reprogramming during in vitro human endometrial fibroblast decidualization. Physiol Genomics 7: 135-148, 2001. First published September 21, 2001; 10.1152/physiolgenomics.00061.2001.-Human decidual fibroblasts undergo a differentiative commitment to the acquisition of endocrine, metabolic, and structural cell functions in a process known as decidualization. Decidualization is critical for embryo implantation and placental function. We characterized gene expression pattern kinetics during decidual fibroblast differentiation by microarray analysis. Of 6,918 genes analyzed, 121 genes were induced by more than twofold, 110 were downregulated, and 50 showed biphasic behavior. Dynamically regulated genes were could be fit into nine K-means algorithm-based kinetic pattern groups, and by biologic classification, into five categories: cell and tissue function, cell and tissue structure, regulation of gene expression, expressed sequence tag (EST), and "function unknown." Reprogramming of genes within specific functional groups and gene families was a prominent feature that consisted of simultaneous induction and downregulation of a set of genes with related function. We previously observed a conceptually similar process during fetal trophoblast differentiation, in which the same phenomena applied to different genes. Of the 569 dynamically regulated genes regulated by either model, only 81 of these were in common. These results suggest that reprogramming of gene expression within focused functional categories represents a fundamental aspect of cellular differentiation.


Assuntos
Diferenciação Celular/fisiologia , Decídua , Endométrio , Fibroblastos/metabolismo , Regulação da Expressão Gênica/fisiologia , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Análise por Conglomerados , AMP Cíclico/farmacologia , Decídua/citologia , Regulação para Baixo , Endométrio/citologia , Estradiol/farmacologia , Feminino , Fibroblastos/citologia , Fibroblastos/efeitos dos fármacos , Perfilação da Expressão Gênica , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Proteína 1 de Ligação a Fator de Crescimento Semelhante à Insulina/biossíntese , Proteína 1 de Ligação a Fator de Crescimento Semelhante à Insulina/genética , Análise de Sequência com Séries de Oligonucleotídeos , Gravidez , Progesterona/farmacologia , Prolactina/biossíntese , Prolactina/genética , RNA Mensageiro/metabolismo , Ativação Transcricional , Regulação para Cima , Vimentina/biossíntese
16.
Adv Exp Med Biol ; 500: 479-87, 2001.
Artigo em Inglês | MEDLINE | ID: mdl-11764985

RESUMO

In summary, acute lung injury is a severe (>40% mortality) respiratory disease associated with numerous precipitating factors. Despite extensive research since its initial description over 30 years ago, questions remain about the basic pathophysiological mechanisms and their relationship to therapeutic strategies. Histopathology reveals surfactant disruption, epithelial perturbation and sepsis, either as initiating factors or as secondary complications, which in turn increase the expression of cytokines that sequester and activate inflammatory cells, most notably, neutrophils. Concomitant release of reactive oxygen and nitrogen species subsequently modulates endothelial function. Together these events orchestrate the principal clinical manifestations of the syndrome, pulmonary edema and atelectasis. To better understand the gene-environmental interactions controlling this complex process, we examined the relative sensitivity of inbred mouse strains to acute lung injury induced by ozone, ultrafine PTFE, or fine particulate NiSO4 (0.2 microm MMAD, 15-150 microg/m3). Measuring survival time, protein and neutrophils in bronchoalveolar lavage, lung wet: dry weight, and histology, we found that these responses varied between inbred mouse strains, and susceptibility is heritable. To assess the molecular progression of NiSO4-induced acute lung injury, temporal relationships of 8734 genes and expressed sequence tags were assessed by cDNA microarray analysis. Clustering of co-regulated genes (displaying similar temporal expression patterns) revealed the altered expression of relatively few genes. Enhanced expression occurred mainly in genes associated with oxidative stress, anti-proteolytic function, and repair of the extracellular matrix. Concomitantly, surfactant proteins and Clara cell secretory protein mRNA expression decreased. Genome wide analysis of 307 mice generated from the backcross of resistant B6xA F1 with susceptible A strain identified significant linkage to a region on chromosome 6 (proposed as Aliq4) and suggestive linkages on chromosomes 1, 8, and 12. Combining of these QTLs with two additional possible modifying loci (chromosome 9 and 16) accounted for the difference in survival time noted in the A and B6 parental strains. Combining these findings with those of the microarray analysis has enabled prioritization of candidate genes. These candidates, in turn, can be directed to the lung epithelium in transgenic mice or abated in inducible and constitutive gene-targeted mice. Initial results are encouraging and suggest that several of these mice vary in their susceptibility to oxidant-induced lung injury. Thus, these combined approaches have led to new insights into functional genomics of lung injury and diseases.


Assuntos
Exposição Ambiental/efeitos adversos , Predisposição Genética para Doença/genética , Lesão Pulmonar , Oxidantes/efeitos adversos , Animais , Fator de Crescimento Epidérmico/metabolismo , Genômica , Humanos , Níquel/efeitos adversos , Ozônio/efeitos adversos , Politetrafluoretileno/efeitos adversos , Característica Quantitativa Herdável , Fator de Crescimento Transformador alfa/metabolismo
17.
Ann Neurol ; 50(6): 699-707, 2001 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11761467

RESUMO

Using microarray technology, we investigated whether the gene expression profile in white blood cells could be used as a fingerprint of different disease states. Adult rats were subjected to ischemic strokes, hemorrhagic strokes, sham surgeries, kainate-induced seizures, hypoxia, or insulin-induced hypoglycemia, and compared with controls. The white blood cell RNA expression patterns were assessed 24 hours later using oligonucleotide microarrays. Results showed that many genes were upregulated or downregulated at least twofold in white blood cells after each experimental condition. Blood genomic response patterns were different for each condition. These results demonstrate the potential of blood gene expression profiling for diagnostic, mechanistic, and therapeutic assessment of a wide variety of disease states.


Assuntos
Perfilação da Expressão Gênica , Hipoglicemia/genética , Hipóxia/genética , Leucócitos/fisiologia , Análise de Sequência com Séries de Oligonucleotídeos , Convulsões/genética , Acidente Vascular Cerebral/genética , Animais , Hemorragia Cerebral/sangue , Hemorragia Cerebral/induzido quimicamente , Hemorragia Cerebral/metabolismo , Agonistas de Aminoácidos Excitatórios , Marcadores Genéticos , Glucose/farmacologia , Humanos , Hipoglicemia/sangue , Hipoglicemia/metabolismo , Hipóxia/sangue , Hipóxia/metabolismo , Insulina/farmacologia , Ácido Caínico , Leucócitos/efeitos dos fármacos , Masculino , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Convulsões/sangue , Convulsões/induzido quimicamente , Convulsões/metabolismo , Acidente Vascular Cerebral/sangue , Acidente Vascular Cerebral/metabolismo
18.
Res Rep Health Eff Inst ; (105): 5-58; discussion 59-71, 2001 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11954676

RESUMO

To begin identifying genes controlling individual susceptibility to particulate matter, responses of inbred mouse strains exposed to nickel sulfate (NiSO4*) were compared with those of mice exposed to ozone (O3) or polytetrafluoroethylene (PTFE). The A strain was sensitive to NiSO4-induced lung injury (quantified by survival time), the C3H/He (C3) strain and several other strains were intermediate in their responses, and the C57BL/6 (B6) strain was resistant. The strains showed a pattern of response similar to the patterns of response to O3 and PTFE. The phenotype of A x B6 offspring (B6AF1) resembled that of the resistant B6 parental strain, with strains exhibiting sensitivity in the order A > C3 > B6 = B6AF1. Pathology was comparable for the A and B6 mice, and exposure to NiSO4 at 15 microg/m3 produced 20% mortality in A mice. Strain sensitivity for the presence of protein or neutrophils in lavage fluid differed from strain sensitivity for survival time, suggesting that they are not causally linked but are controlled by an independent gene or genes. In the B6 strain, exposure to nickel oxide (NiO) by instillation (40 to 1000 nm) or inhalation (50 nm) produced no changes, whereas inhalation of NiSO4 (60 or 250 nm) increased lavage proteins and neutrophils. Complementary DNA (cDNA) microarray analysis with 8,734 sequence-verified clones revealed a temporal pattern of increased oxidative stress, extracellular matrix repair, cell proliferation, and hypoxia, followed by a decrease in surfactant-associated proteins (SPs). Certain expressed sequence tags (ESTs), clustered with known genes, suggest possible coregulation and novel roles in pulmonary injury. Finally, locus number estimation (Wright equation) and a genomewide analysis suggested 5 genes could explain the survival time and identified significant linkage for a quantitative trait locus (QTL) on chromosome 6, Aliq4 (acute lung injury QTL4). Haplotype analysis identified an allelic combination of 5 QTLs that could explain the difference in sensitivity to acute lung injury between parental strains. Positional candidate genes for Aliq4 include aquaporin-1 (Aqp1), SP-B, and transforming growth factor-alpha (TGF-alpha). Transgenic mice expressing TGF-alpha were rescued from NiSO4 injury (that is, they had diminished SP-B loss and increased survival time). These findings suggest that NiSO4-induced acute lung injury is a complex trait controlled by at least 5 genes (all possibly involved in cell proliferation and surfactant function). Future assessment of these susceptibility genes (including evaluations of human synteny and function) could provide valuable insights into individual susceptibility to the adverse effects of particulate matter.


Assuntos
Poluentes Atmosféricos/efeitos adversos , Regulação da Expressão Gênica/efeitos dos fármacos , Inflamação/fisiopatologia , Exposição por Inalação , Irritantes/efeitos adversos , Pneumopatias/etiologia , Níquel/efeitos adversos , Oxidantes Fotoquímicos/efeitos adversos , Ozônio/efeitos adversos , Politetrafluoretileno/efeitos adversos , Animais , Northern Blotting , Lavagem Broncoalveolar , Divisão Celular , Mapeamento Cromossômico , Modelos Animais de Doenças , Pneumopatias/genética , Pneumopatias/veterinária , Camundongos , Camundongos Endogâmicos , Análise de Sequência com Séries de Oligonucleotídeos , Tamanho da Partícula , Fenótipo , Tensoativos , Análise de Sobrevida
19.
J Clin Invest ; 106(9): 1105-13, 2000 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11067863

RESUMO

Apolipoprotein J/clusterin (apoJ/clusterin), an intriguing protein with unknown function, is induced in myocarditis and numerous other inflammatory injuries. To test its ability to modify myosin-induced autoimmune myocarditis, we generated apoJ-deficient mice. ApoJ-deficient and wild-type mice exhibited similar initial onset of myocarditis, as evidenced by the induction of two early markers of the T cell-mediated immune response, MHC-II and TNF receptor p55. Furthermore, autoantibodies against the primary antigen cardiac myosin were induced to the same extent. Although the same proportion of challenged animals exhibited some degree of inflammatory infiltrate, inflammation was more severe in apoJ-deficient animals. Inflammatory lesions were more diffuse and extensive in apoJ-deficient mice, particularly in females. In marked contrast to wild-type animals, the development of a strong generalized secondary response against cardiac antigens in apoJ-deficient mice was predictive of severe myocarditis. Wild-type mice with a strong Ab response to secondary antigens appeared to be protected from severe inflammation. After resolution of inflammation, apoJ-deficient, but not wild-type, mice exhibited cardiac function impairment and severe myocardial scarring. These results suggest that apoJ limits progression of autoimmune myocarditis and protects the heart from postinflammatory tissue destruction.


Assuntos
Doenças Autoimunes/etiologia , Glicoproteínas/fisiologia , Chaperonas Moleculares , Miocardite/etiologia , Animais , Antígenos CD/biossíntese , Autoanticorpos/biossíntese , Doenças Autoimunes/imunologia , Doenças Autoimunes/patologia , Sequência de Bases , Clusterina , Primers do DNA/genética , Feminino , Glicoproteínas/deficiência , Glicoproteínas/genética , Antígenos de Histocompatibilidade Classe II/biossíntese , Masculino , Camundongos , Camundongos Knockout , Miocardite/imunologia , Miocardite/patologia , Miosinas/imunologia , Receptores do Fator de Necrose Tumoral/biossíntese , Receptores Tipo I de Fatores de Necrose Tumoral , Linfócitos T/imunologia
20.
Oncogene ; 18(4): 1103-11, 1999 Jan 28.
Artigo em Inglês | MEDLINE | ID: mdl-10023687

RESUMO

The c-myb gene encodes a sequence specific transactivator that is required for fetal hematopoiesis, but its potential role in other tissues is less clear because of the early fetal demise of mice with targeted deletions of the c-myb gene and incomplete of knowledge about c-myb's expression pattern. In the hematopoietic system, c-Myb protein acts on target genes whose expression is restricted to individual lineages, despite Myb's presence and role in multiple immature lineages. This suggests that c-Myb actions within different cell type-specific contexts are strongly affected by combinatorial interactions. To consider the possibility of similar c-Myb actions could extend into non-hematopoietic systems in other cell and tissue compartments, we characterized c-myb expression in developing and adult mice using in situ hybridization and correlated this with stage-specific differentiation and mitotic activity. Diverse tissues exhibited strong c-myb expression during development, notably tooth buds, the thyroid primordium, developing trachea and proximal branching airway epithelium, hair follicles, hematopoietic cells, and gastrointestinal crypt epithelial cells. The latter three of these all maintained high expression into adulthood, but with characteristic restriction to immature cell lineages prior to their terminal differentiation. In all sites, during fetal and adult stages, loss of c-Myb expression correlated strikingly with the initiation of terminal differentiation, but not the loss of mitotic activity. Based on these data, we hypothesize that c-Myb's function during cellular differentiation is both an activator of immature gene expression and a suppressor of terminal differentiation in diverse lineages.


Assuntos
Diferenciação Celular/fisiologia , Desenvolvimento Embrionário e Fetal/genética , Oncogenes , Proteínas Proto-Oncogênicas/fisiologia , Transativadores/fisiologia , Fatores Etários , Animais , Sistema Digestório/embriologia , Folículo Piloso/embriologia , Hibridização In Situ , Fígado/embriologia , Camundongos , Mitose , Odontogênese , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-myb , Timo/embriologia , Glândula Tireoide/embriologia , Traqueia/embriologia , Transativadores/genética , Transativadores/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...