Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
J Neurogenet ; 26(3-4): 397-412, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22783900

RESUMO

Disturbances in sleep are encountered in the majority of patients with depressive disorder. To elucidate the molecular mechanisms behind this relationship, we examined gene expression changes in a rodent model for disturbed sleep and depression. The animals were treated with daily injections of clomipramine to affect their sleep during early infancy. This early interference with sleep is known to induce depression-like behavior in adult animals. After 2 weeks of treatment, the change in gene expression was examined using the Affymetrix Rat 230.2 chip. We studied the gene expression in the basal forebrain, hippocampus, and frontal cortex and combined the results to reveal the otherwise indissectible networks between and around the tissues. The major disrupted pathways between the three brain areas were related to synaptic transmission, regulation of translation, and ubiquitinylation. The involved pathways were within the cellular components of the axons, growth cones, melanosomes, and pigment granules. A network analysis allowing for additional interactors, in the form of chemicals or gene products, revealed a disturbed communicational network between the different brain areas. This disturbed network is centered around serotonin, Mn(II), and Rhoa. The findings elucidate inter-tissue pathways and networks in the brain that are involved in sleep and mood regulation. The findings are of uttermost interest, some are quite predictable and obvious, but some are novel or have only been proposed by rare theoretical speculations (such as the melanosome and Mn(II) involvement). Equally important as the findings are the methods described in this article. In this study, we present two novel simple ways to perform system biological analysis based on gene expression array data. We used two already existing tools in a new way, and by careful planning of the input data, managed to extrapolate intricate hidden inter-tissue networks to build a molecular picture of disease.


Assuntos
Depressão/etiologia , Depressão/patologia , Hipocampo/metabolismo , Córtex Pré-Frontal/metabolismo , Prosencéfalo/metabolismo , Transtornos do Sono-Vigília/complicações , Animais , Animais Recém-Nascidos , Antidepressivos Tricíclicos/toxicidade , Clomipramina/toxicidade , Modelos Animais de Doenças , Feminino , Preferências Alimentares/efeitos dos fármacos , Preferências Alimentares/fisiologia , Perfilação da Expressão Gênica , Regulação da Expressão Gênica/efeitos dos fármacos , Redes Reguladoras de Genes/efeitos dos fármacos , Redes Reguladoras de Genes/fisiologia , Masculino , Vias Neurais/fisiologia , Análise de Sequência com Séries de Oligonucleotídeos , Ratos , Ratos Wistar , Transtornos do Sono-Vigília/induzido quimicamente , Sacarose/administração & dosagem , Edulcorantes/administração & dosagem
2.
Int J Cancer ; 130(7): 1558-66, 2012 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-21544814

RESUMO

Defects in the mismatch repair system lead to microsatellite instability (MSI), a feature observed in ∼ 15% of all colorectal cancers (CRCs). Microsatellite mutations that drive tumourigenesis, typically inactivation of tumour suppressors, are selected for and are frequently detected in MSI cancers. Here, we evaluated somatic mutations in microsatellite repeats of 790 genes chosen based on reduced expression in MSI CRC and existence of a coding mononucleotide repeat of 6-10 bp in length. All the repeats were initially sequenced in 30 primary MSI CRC samples and whenever frameshift mutations were identified in >20%, additional 70 samples were sequenced. To distinguish driver mutations from passengers, we similarly analyzed the occurrence of frameshift mutations in 121 intronic control repeats and utilized a statistical regression model to determine cut-off mutation frequencies for repeats of all types (A/T and C/G, 6-10 bp). Along with several know target genes, including TGFBR2, ACVR2, and MSH3, six novel candidate driver genes emerged that harbored significantly more mutations than identical control repeats. The mutation frequencies in 100 MSI CRC samples were 51% in G8 of GLYR1, 47% in T9 of ABCC5, 43% in G8 of WDTC1, 33% in A8 of ROCK1, 30% in T8 of OR51E2, and 28% in A8 of TCEB3. Immunohistochemical staining of GLYR1 revealed defective protein expression in tumors carrying biallelic mutations, supporting a loss of function hypothesis. This is a large scale, unbiased effort to identify genes that when mutated are likely to contribute to MSI CRC development.


Assuntos
Neoplasias Colorretais/genética , DNA de Neoplasias/genética , Instabilidade de Microssatélites , Linhagem Celular Tumoral , Mutação da Fase de Leitura , Células HCT116 , Humanos , Imuno-Histoquímica/métodos , Repetições de Microssatélites , Taxa de Mutação , Análise de Regressão
3.
Mol Cancer Res ; 8(7): 952-60, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20551151

RESUMO

The circadian clock regulates daily variations in physiologic processes. CLOCK acts as a regulator in the circadian apparatus controlling the expression of other clock genes, including PER1. Clock genes have been implicated in cancer-related functions; in this work, we investigated CLOCK as a possible target of somatic mutations in microsatellite unstable colorectal cancers. Combining microarray gene expression data and public gene sequence information, we identified CLOCK as 1 of 790 putative novel microsatellite instability (MSI) target genes. A total of 101 MSI colorectal carcinomas (CRC) were sequenced for a coding microsatellite in CLOCK. The effect of restoring CLOCK expression was studied in LS180 cells lacking wild-type CLOCK by stably expressing GST-CLOCK or glutathione S-transferase empty vector and testing the effects of UV-induced apoptosis and radiation by DNA content analysis using flow cytometry. Putative novel CLOCK target genes were searched by using ChIP-seq. CLOCK mutations occurred in 53% of MSI CRCs. Restoring CLOCK expression in cells with biallelic CLOCK inactivation resulted in protection against UV-induced apoptosis and decreased G(2)-M arrest in response to ionizing radiation. Using ChIP-Seq, novel CLOCK-binding elements were identified near DNA damage genes p21, NBR1, BRCA1, and RAD50. CLOCK is shown to be mutated in cancer, and altered response to DNA damage provides one plausible mechanism of tumorigenesis.


Assuntos
Adenocarcinoma/genética , Proteínas CLOCK/genética , Neoplasias Colorretais/genética , Sequência de Aminoácidos , Sequência de Bases , Linhagem Celular Tumoral , Mutação da Fase de Leitura , Perfilação da Expressão Gênica , Células HCT116 , Humanos , Repetições de Microssatélites , Dados de Sequência Molecular , Mutação
4.
Eur J Neurosci ; 31(8): 1465-73, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20384783

RESUMO

Disturbances in sleep are encountered in the majority of patients with depression. To elucidate the possible molecular mechanisms behind this relationship we examined gene expression changes in a rodent model for depression and disturbed sleep. Animals were treated with daily injections of clomipramine in their early infancy, after which gene expression in basal forebrain was examined using Affymetrix Rat 230.2 chips. We tested the levels of both single transcripts and involved pathways, and searched for common nominators (i.e. transcription factors) that could explain these changes. We identified 72 differentially expressed gene transcripts, many of which are involved in epigenetic regulation, such as DNMT2. Analysis of functional pathways revealed statistically significant changes of the biological process of synaptic transmission, the cellular compartment of the synapse and the molecular function of GABA signalling, showing that transcripts with altered expression are functionally related. Finally, promoter analysis of the differentially expressed genes showed a clear enrichment of binding sites for the transcription factor CREB1, a molecule also involved in epigenetic regulation (cAMP response element-binding protein induces histone modifications). These results indicate that CREB1 may constitute one of the major links between disturbed sleep and mood. The results also highlight the molecular mechanisms in the murine clomipramine model, previously shown to be a valid model for depression.


Assuntos
Transtorno Depressivo/genética , Expressão Gênica , Transtornos do Sono-Vigília/genética , Animais , Clomipramina , Transtorno Depressivo/induzido quimicamente , Transtorno Depressivo/metabolismo , Modelos Animais de Doenças , Feminino , Masculino , Análise de Sequência com Séries de Oligonucleotídeos , Regiões Promotoras Genéticas , Prosencéfalo/metabolismo , Ratos , Ratos Wistar , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/genética , Transtornos do Sono-Vigília/induzido quimicamente , Transtornos do Sono-Vigília/metabolismo
5.
Int J Cancer ; 125(3): 530-40, 2009 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-19431147

RESUMO

High biological activity of the transforming growth factor (TGF)-beta-Smad pathway characterizes the malignant phenotype of malignant gliomas and confers poor prognosis to glioma patients. Accordingly, TGF-beta has become a novel target for the experimental treatment of these tumors. TGF-beta is processed by furin-like proteases (FLP) and secreted from cells in a latent complex with its processed propeptide, the latency-associated peptide (LAP). Latent TGF-beta-binding protein 1 (LTBP-1) covalently binds to this small latent TGF-beta complex (SLC) and regulates its function, presumably via interaction with the extracellular matrix (ECM). We report here that the levels of LTBP-1 protein in vivo increase with the grade of malignancy in gliomas. LTBP-1 is associated with the ECM as well as secreted into the medium in cultured malignant glioma cells. The release of LTBP-1 into the medium is decreased by the inhibition of FLP activity. Gene-transfer mediated overexpression of LTBP-1 in glioma cell lines results in an increase inTGF-beta activity. Accordingly, Smad2 phosphorylation as an intracellular marker of TGF-beta activity is enhanced. Conversely, LTBP-1 gene silencing reduces TGF-beta activity and Smad2 phosphorylation without affecting TGF-beta protein levels. Collectively, we identify LTBP-1 as an important modulator of TGF-beta activation in glioma cells, which may contribute to the malignant phenotype of these tumors.


Assuntos
Neoplasias Encefálicas/metabolismo , Glioma/metabolismo , Proteínas de Ligação a TGF-beta Latente/metabolismo , Transdução de Sinais , Proteína Smad2/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Astrocitoma/metabolismo , Linhagem Celular Tumoral , Proliferação de Células , Matriz Extracelular/metabolismo , Regulação Neoplásica da Expressão Gênica , Glioblastoma/metabolismo , Humanos , Immunoblotting , Imuno-Histoquímica , Análise de Sequência com Séries de Oligonucleotídeos , Fenótipo , Fosforilação , Reação em Cadeia da Polimerase , Regulação para Cima
6.
Nat Genet ; 41(1): 47-55, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19060911

RESUMO

Recent genome-wide association (GWA) studies of lipids have been conducted in samples ascertained for other phenotypes, particularly diabetes. Here we report the first GWA analysis of loci affecting total cholesterol (TC), low-density lipoprotein (LDL) cholesterol, high-density lipoprotein (HDL) cholesterol and triglycerides sampled randomly from 16 population-based cohorts and genotyped using mainly the Illumina HumanHap300-Duo platform. Our study included a total of 17,797-22,562 persons, aged 18-104 years and from geographic regions spanning from the Nordic countries to Southern Europe. We established 22 loci associated with serum lipid levels at a genome-wide significance level (P < 5 x 10(-8)), including 16 loci that were identified by previous GWA studies. The six newly identified loci in our cohort samples are ABCG5 (TC, P = 1.5 x 10(-11); LDL, P = 2.6 x 10(-10)), TMEM57 (TC, P = 5.4 x 10(-10)), CTCF-PRMT8 region (HDL, P = 8.3 x 10(-16)), DNAH11 (LDL, P = 6.1 x 10(-9)), FADS3-FADS2 (TC, P = 1.5 x 10(-10); LDL, P = 4.4 x 10(-13)) and MADD-FOLH1 region (HDL, P = 6 x 10(-11)). For three loci, effect sizes differed significantly by sex. Genetic risk scores based on lipid loci explain up to 4.8% of variation in lipids and were also associated with increased intima media thickness (P = 0.001) and coronary heart disease incidence (P = 0.04). The genetic risk score improves the screening of high-risk groups of dyslipidemia over classical risk factors.


Assuntos
Doença das Coronárias/sangue , Doença das Coronárias/genética , Predisposição Genética para Doença , Lipídeos/sangue , Locos de Características Quantitativas/genética , População Branca/genética , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Índice de Massa Corporal , HDL-Colesterol/sangue , Estudos de Coortes , Feminino , Testes Genéticos , Genoma Humano/genética , Humanos , Masculino , Redes e Vias Metabólicas/genética , Pessoa de Meia-Idade , Fenótipo , Polimorfismo de Nucleotídeo Único/genética , Caracteres Sexuais
7.
Am J Hum Genet ; 83(6): 787-94, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19061986

RESUMO

Although high-density SNP genotyping platforms generate a momentum for detailed genome-wide association (GWA) studies, an offshoot is a new insight into population genetics. Here, we present an example in one of the best-known founder populations by scrutinizing ten distinct Finnish early- and late-settlement subpopulations. By determining genetic distances, homozygosity, and patterns of linkage disequilibrium, we demonstrate that population substructure, and even individual ancestry, is detectable at a very high resolution and supports the concept of multiple historical bottlenecks resulting from consecutive founder effects. Given that genetic studies are currently aiming at identifying smaller and smaller genetic effects, recognizing and controlling for population substructure even at this fine level becomes imperative to avoid confounding and spurious associations. This study provides an example of the power of GWA data sets to demonstrate stratification caused by population history even within a seemingly homogeneous population, like the Finns. Further, the results provide interesting lessons concerning the impact of population history on the genome landscape of humans, as well as approaches to identify rare variants enriched in these subpopulations.


Assuntos
Efeito Fundador , Variação Genética , Genoma Humano , Desequilíbrio de Ligação , População , Alelos , Cromossomos Humanos Par 22 , Frequência do Gene , Estudo de Associação Genômica Ampla/métodos , Homozigoto , Humanos , Polimorfismo de Nucleotídeo Único
8.
BMC Med Genomics ; 1: 55, 2008 Nov 11.
Artigo em Inglês | MEDLINE | ID: mdl-19014429

RESUMO

BACKGROUND: Studies on asbestos-induced tumourigenesis have indicated the role of, e.g., reactive oxygen/nitrogen species, mitochondria, as well as NF-kappaB and MAPK signalling pathways. The exact molecular mechanisms contributing to asbestos-mediated carcinogenesis are, however, still to be characterized. METHODS: In this study, gene expression data analyses together with gene annotation data from the Gene Ontology (GO) database were utilized to identify pathways that are differentially regulated in lung and tumour tissues between asbestos-exposed and non-exposed lung cancer patients. Differentially regulated pathways were identified from gene expression data from 14 asbestos-exposed and 14 non-exposed lung cancer patients using custom-made software and Iterative Group Analysis (iGA). Western blotting was used to further characterize the findings, specifically to determine the protein levels of UBA1 and UBA7. RESULTS: Differences between asbestos-related and non-related lung tumours were detected in pathways associated with, e.g., ion transport, NF-kappaB signalling, DNA repair, as well as spliceosome and nucleosome complexes. A notable fraction of the pathways down-regulated in both normal and tumour tissue of the asbestos-exposed patients were related to protein ubiquitination, a versatile process regulating, for instance, DNA repair, cell cycle, and apoptosis, and thus being also a significant contributor of carcinogenesis. Even though UBA1 or UBA7, the early enzymes involved in protein ubiquitination and ubiquitin-like regulation of target proteins, did not underlie the exposure-related deregulation of ubiquitination, a difference was detected in the UBA1 and UBA7 levels between squamous cell carcinomas and respective normal lung tissue (p = 0.02 and p = 0.01) without regard to exposure status. CONCLUSION: Our results indicate alterations in protein ubiquitination related both to cancer type and asbestos. We present for the first time pathway analysis results on asbestos-associated lung cancer, providing important insight into the most relevant targets for future research.

9.
J Mol Med (Berl) ; 86(10): 1139-52, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18600308

RESUMO

Autoimmune polyendocrinopathy-candidiasis-ectodermal dystrophy (APECED) is a monogenic autoimmune disease caused by mutations in the autoimmune regulator (AIRE) gene. AIRE functions as a transcriptional regulator, and it has a central role in the development of immunological tolerance. AIRE regulates the expression of ectopic antigens in epithelial cells of the thymic medulla and has been shown to participate in the development of peripheral tolerance. However, the mechanism of action of AIRE has remained elusive. To further investigate the role of AIRE in host immune functions, we studied the properties and transcript profiles in in vitro monocyte-differentiated dendritic cells (moDCs) obtained from APECED patients and healthy controls. AIRE-deficient monocytes showed typical DC morphology and expressed DC marker proteins cluster of differentiation 86 and human leukocyte antigen class II. APECED patient-derived moDCs were functionally impaired: the transcriptional response of cytokine genes to pathogens was drastically reduced. Interestingly, some changes were observable already at the immature DC stage. Pathway analyses of transcript profiles revealed that the expression of the components of the host cell signaling pathways involved in cell-cell signalling, innate immune responses, and cytokine activity were reduced in APECED moDCs. Our observations support a role for AIRE in peripheral tolerance and are the first ones to show that AIRE has a critical role in DC responses to microbial stimuli in humans.


Assuntos
Células Dendríticas/metabolismo , Poliendocrinopatias Autoimunes/imunologia , Transdução de Sinais/fisiologia , Fatores de Transcrição/fisiologia , Adulto , Candida albicans/crescimento & desenvolvimento , Diferenciação Celular/genética , Diferenciação Celular/fisiologia , Células Cultivadas , Citocinas/genética , Citocinas/metabolismo , Células Dendríticas/microbiologia , Células Dendríticas/virologia , Feminino , Citometria de Fluxo , Perfilação da Expressão Gênica , Humanos , Pessoa de Meia-Idade , Monócitos/citologia , Monócitos/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , Poliendocrinopatias Autoimunes/genética , Poliendocrinopatias Autoimunes/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Vírus Sendai/crescimento & desenvolvimento , Transdução de Sinais/genética , Transdução de Sinais/imunologia , Fatores de Transcrição/genética , Fatores de Transcrição/imunologia , Transcrição Gênica , Proteína AIRE
10.
Cancer Cell ; 13(5): 407-19, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18455124

RESUMO

The Drosophila transcription factor Prospero functions as a tumor suppressor, and it has been suggested that the human counterpart of Prospero, PROX1, acts similarly in human cancers. However, we show here that PROX1 promotes dysplasia in colonic adenomas and colorectal cancer progression. PROX1 expression marks the transition from benign colon adenoma to carcinoma in situ, and its loss inhibits growth of human colorectal tumor xenografts and intestinal adenomas in Apc(min/+) mice, while its transgenic overexpression promotes colorectal tumorigenesis. Furthermore, in intestinal tumors PROX1 is a direct and dose-dependent target of the beta-catenin/TCF signaling pathway, responsible for the neoplastic transformation. Our data underscore the complexity of cancer pathogenesis and implicate PROX1 in malignant tumor progression through the regulation of cell polarity and adhesion.


Assuntos
Adenoma/genética , Neoplasias do Colo/genética , Proteínas de Homeodomínio/genética , Proteínas Supressoras de Tumor/genética , Adenoma/patologia , Carcinoma in Situ/genética , Linhagem Celular Tumoral , Neoplasias do Colo/patologia , Neoplasias Colorretais/genética , Progressão da Doença , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Humanos , Fenótipo , beta Catenina/fisiologia
11.
Proc Natl Acad Sci U S A ; 105(14): 5513-8, 2008 Apr 08.
Artigo em Inglês | MEDLINE | ID: mdl-18391202

RESUMO

A recent study described a recessive ATPase activating germ-line mutation in smooth-muscle myosin (smmhc/myh11) underlying the zebrafish meltdown (mlt) phenotype. The mlt zebrafish develops intestinal abnormalities reminiscent of human Peutz-Jeghers syndrome (PJS) and juvenile polyposis (JP). To examine the role of MYH11 in human intestinal neoplasia, we searched for MYH11 mutations in patients with colorectal cancer (CRC), PJS and JP. We found somatic protein-elongating frameshift mutations in 55% of CRCs displaying microsatellite instability and in the germ-line of one individual with PJS. Additionally, two somatic missense mutations were found in one microsatellite stable CRC. These two missense mutations, R501L and K1044N, and the frameshift mutations were functionally evaluated. All mutations resulted in unregulated molecules displaying constitutive motor activity, similar to the mutant myosin underlying mlt. Thus, MYH11 mutations appear to contribute also to human intestinal neoplasia. Unregulated MYH11 may affect the cellular energy balance or disturb cell lineage decisions in tumor progenitor cells. These data challenge our view on MYH11 as a passive differentiation marker functioning in muscle contraction and add to our understanding of intestinal neoplasia.


Assuntos
Regulação Neoplásica da Expressão Gênica , Neoplasias Intestinais/genética , Mutação , Cadeias Pesadas de Miosina/genética , Miosina Tipo II/genética , Miosinas de Músculo Liso/genética , Polipose Adenomatosa do Colo/genética , Sequência de Aminoácidos , Estudos de Casos e Controles , Neoplasias Colorretais/genética , Análise Mutacional de DNA , Humanos , Dados de Sequência Molecular , Síndrome de Peutz-Jeghers/genética , Miosinas de Músculo Liso/fisiologia
12.
PLoS Med ; 5(3): e51, 2008 Mar 11.
Artigo em Inglês | MEDLINE | ID: mdl-18336063

RESUMO

BACKGROUND: The acquired component of complex traits is difficult to dissect in humans. Obesity represents such a trait, in which the metabolic and molecular consequences emerge from complex interactions of genes and environment. With the substantial morbidity associated with obesity, a deeper understanding of the concurrent metabolic changes is of considerable importance. The goal of this study was to investigate this important acquired component and expose obesity-induced changes in biological pathways in an identical genetic background. METHODS AND FINDINGS: We used a special study design of "clonal controls," rare monozygotic twins discordant for obesity identified through a national registry of 2,453 young, healthy twin pairs. A total of 14 pairs were studied (eight male, six female; white), with a mean +/- standard deviation (SD) age 25.8 +/- 1.4 y and a body mass index (BMI) difference 5.2 +/- 1.8 kg/m(2). Sequence analyses of mitochondrial DNA (mtDNA) in subcutaneous fat and peripheral leukocytes revealed no aberrant heteroplasmy between the co-twins. However, mtDNA copy number was reduced by 47% in the obese co-twin's fat. In addition, novel pathway analyses of the adipose tissue transcription profiles exposed significant down-regulation of mitochondrial branched-chain amino acid (BCAA) catabolism (p < 0.0001). In line with this finding, serum levels of insulin secretion-enhancing BCAAs were increased in obese male co-twins (9% increase, p = 0.025). Lending clinical relevance to the findings, in both sexes the observed aberrations in mitochondrial amino acid metabolism pathways in fat correlated closely with liver fat accumulation, insulin resistance, and hyperinsulinemia, early aberrations of acquired obesity in these healthy young adults. CONCLUSIONS: Our findings emphasize a substantial role of mitochondrial energy- and amino acid metabolism in obesity and development of insulin resistance.


Assuntos
Tecido Adiposo/metabolismo , Índice de Massa Corporal , DNA Mitocondrial/metabolismo , Obesidade/etiologia , Gêmeos Monozigóticos/fisiologia , Tecido Adiposo/citologia , Adulto , Aminoácidos de Cadeia Ramificada/sangue , Diferenciação Celular/fisiologia , Estudos Transversais , Regulação para Baixo , Metabolismo Energético , Feminino , Humanos , Hiperinsulinismo/fisiopatologia , Inflamação/fisiopatologia , Resistência à Insulina/fisiologia , Fígado/anatomia & histologia , Estudos Longitudinais , Masculino , Redes e Vias Metabólicas , Obesidade/metabolismo
13.
BMC Genomics ; 9: 146, 2008 Mar 28.
Artigo em Inglês | MEDLINE | ID: mdl-18371231

RESUMO

BACKGROUND: The neuronal ceroid lipofuscinoses (NCL) are a group of children's inherited neurodegenerative disorders, characterized by blindness, early dementia and pronounced cortical atrophy. The similar pathological and clinical profiles of the different forms of NCL suggest that common disease mechanisms may be involved. To explore the NCL-associated disease pathology and molecular pathways, we have previously produced targeted knock-out mice for Cln1 and Cln5. Both mouse-models replicate the NCL phenotype and neuropathology; the Cln1-/- model presents with early onset, severe neurodegenerative disease, whereas the Cln5-/- model produces a milder disease with a later onset. RESULTS: Here we have performed quantitative gene expression profiling of the cortex from 1 and 4 month old Cln1-/- and Cln5-/- mice. Combined microarray datasets from both mouse models exposed a common affected pathway: genes regulating neuronal growth cone stabilization display similar aberrations in both models. We analyzed locus specific gene expression and showed regional clustering of Cln1 and three major genes of this pathway, further supporting a close functional relationship between the corresponding gene products; adenylate cyclase-associated protein 1 (Cap1), protein tyrosine phosphatase receptor type F (Ptprf) and protein tyrosine phosphatase 4a2 (Ptp4a2). The evidence from the gene expression data, indicating changes in the growth cone assembly, was substantiated by the immunofluorescence staining patterns of Cln1-/- and Cln5-/- cortical neurons. These primary neurons displayed abnormalities in cytoskeleton-associated proteins actin and beta-tubulin as well as abnormal intracellular distribution of growth cone associated proteins GAP-43, synapsin and Rab3. CONCLUSION: Our data provide the first evidence for a common molecular pathogenesis behind neuronal degeneration in INCL and vLINCL. Since CLN1 and CLN5 code for proteins with distinct functional roles these data may have implications for other forms of NCLs as well.


Assuntos
Encéfalo/metabolismo , Regulação da Expressão Gênica/genética , Cones de Crescimento/patologia , Glicoproteínas de Membrana/genética , Lipofuscinoses Ceroides Neuronais/genética , Tioléster Hidrolases/genética , Adenilil Ciclases/genética , Adenilil Ciclases/metabolismo , Animais , Western Blotting , Células Cultivadas , Proteína GAP-43/metabolismo , Perfilação da Expressão Gênica , Genótipo , Cones de Crescimento/metabolismo , Proteínas Imediatamente Precoces/genética , Proteínas Imediatamente Precoces/metabolismo , Imuno-Histoquímica , Proteínas de Membrana Lisossomal , Camundongos , Camundongos Knockout , Lipofuscinoses Ceroides Neuronais/patologia , Proteínas Tirosina Fosfatases/genética , Proteínas Tirosina Fosfatases/metabolismo , Proteínas Tirosina Fosfatases Classe 2 Semelhantes a Receptores/genética , Proteínas Tirosina Fosfatases Classe 2 Semelhantes a Receptores/metabolismo , Sinapsinas/metabolismo , Proteínas rab3 de Ligação ao GTP/metabolismo
14.
Nucleic Acids Res ; 36(Database issue): D830-5, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17932056

RESUMO

The use of genome-wide and high-throughput screening methods on large sample sizes is a well-grounded approach when studying a process as complex and heterogeneous as tumorigenesis. Gene copy number changes are one of the main mechanisms causing cancerous alterations in gene expression and can be detected using array comparative genomic hybridization (aCGH). Microarrays are well suited for the integrative systems biology approach, but none of the existing microarray databases is focusing on copy number changes. We present here CanGEM (Cancer GEnome Mine), which is a public, web-based database for storing quantitative microarray data and relevant metadata about the measurements and samples. CanGEM supports the MIAME standard and in addition, stores clinical information using standardized controlled vocabularies whenever possible. Microarray probes are re-annotated with their physical coordinates in the human genome and aCGH data is analyzed to yield gene-specific copy numbers. Users can build custom datasets by querying for specific clinical sample characteristics or copy number changes of individual genes. Aberration frequencies can be calculated for these datasets, and the data can be visualized on the human genome map with gene annotations. Furthermore, the original data files are available for more detailed analysis. The CanGEM database can be accessed at http://www.cangem.org/.


Assuntos
Bases de Dados Genéticas , Dosagem de Genes , Neoplasias/genética , Análise de Sequência com Séries de Oligonucleotídeos , Mapeamento Cromossômico , Perfilação da Expressão Gênica , Genoma Humano , Humanos , Internet , Interface Usuário-Computador
15.
Mol Immunol ; 45(7): 1847-62, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18083234

RESUMO

AIRE (autoimmune regulator) promotes the establishment of self-tolerance by regulating gene expression in the thymus. Mutations in AIRE lead to an autoimmune disease, APECED. Here we have identified PIAS proteins as novel AIRE interaction partners. Although PIAS proteins function as E3 SUMO ligases, AIRE is not sumoylated. We expressed AIRE, wt PIAS1, and PIAS1 mutants with deleted SP-RING domain or SUMO interaction motif (SIM) in different cell lines and demonstrate that AIRE and PIAS1 localize to adjacent nuclear bodies (NBs). The expression of AIRE enhances the formation of PIAS1 NBs. The ability of PIAS1 to localize into NBs and interconnect with AIRE is neither dependent on the SP-RING domain nor the SIM. Further, we show that PIAS1 is able to attract AIRE into SUMO1-containing complexes and that the process is dependent on the SIM of PIAS1. PIAS1 and AIRE concurrently activate the human insulin promoter, a known target gene of AIRE, and the SP-RING is required for this activation. Moreover, AIRE represses and PIAS1 activates the CSTB promoter, used as a model for a housekeeping promoter, and both the SP-RING and SIM are needed for its activation by PIAS1. Collectively, our data suggest that AIRE and PIAS1 interact functionally to regulate the activities of the target genes of AIRE.


Assuntos
Regulação da Expressão Gênica , Proteínas Inibidoras de STAT Ativados/metabolismo , Fatores de Transcrição/metabolismo , Transcrição Gênica , Motivos de Aminoácidos , Animais , Linhagem Celular , Cistatina B , Cistatinas/genética , Humanos , Insulina/genética , Camundongos , Proteínas Mutantes/metabolismo , Matriz Nuclear/metabolismo , Regiões Promotoras Genéticas/genética , Ligação Proteica , Proteínas Inibidoras de STAT Ativados/química , Processamento de Proteína Pós-Traducional , Estrutura Terciária de Proteína , Transporte Proteico , Deleção de Sequência , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/metabolismo , Frações Subcelulares/metabolismo , Ativação Transcricional/genética , Transfecção , Proteína AIRE
16.
J Mol Med (Berl) ; 85(9): 971-83, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17530208

RESUMO

Rare monogenic dementias have repeatedly exposed novel pathways guiding to details of the molecular pathogenesis behind this complex clinical phenotype. In this paper, we have studied polycystic lipomembranous osteodysplasia with sclerosing leukoencephalopathy (PLOSL), an early onset dementia with bone fractures caused by mutations in TYROBP (DAP12) and TREM2 genes, which encode important signaling molecules in human dendritic cells (DCs). To identify the pathways and biological processes associated with DAP12/TREM2-mediated signaling, we performed genome wide transcript analysis of in vitro differentiated DCs of PLOSL patients representing functional knockouts of either DAP12 or TREM2. Both DAP12- and TREM2-deficient cells differentiated into DCs and responded to pathogenic stimuli. However, the DCs showed morphological differences compared to control cells due to defects in the actin filaments. Not unexpectedly, transcript profiles of the patient DCs showed differential expression of genes involved in immune response. Importantly, significantly diverging transcript levels were also evident for genes earlier associated with other disorders of the central nervous system (CNS) and genes involved in the remodeling of bone, linking these two immunological genes with critical tissue phenotypes of patients. The data underline the functional diversity of the molecules of the innate immune system and implies their significant contribution also in demyelinating CNS disorders, including those resulting in dementia.


Assuntos
Doenças do Sistema Nervoso Central/genética , Doenças Desmielinizantes/genética , Células Dendríticas/metabolismo , Perfilação da Expressão Gênica , Actinas/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Northern Blotting , Diferenciação Celular/genética , Doenças do Sistema Nervoso Central/sangue , Citoesqueleto/metabolismo , Doenças Desmielinizantes/sangue , Células Dendríticas/citologia , Células Dendríticas/imunologia , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Regulação da Expressão Gênica , Humanos , Leucócitos Mononucleares/citologia , Leucócitos Mononucleares/imunologia , Leucócitos Mononucleares/metabolismo , Glicoproteínas de Membrana/genética , Proteínas de Membrana/genética , Microscopia Confocal , Modelos Biológicos , Análise de Sequência com Séries de Oligonucleotídeos , Osteocondrodisplasias/sangue , Osteocondrodisplasias/genética , Receptores Imunológicos/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa
17.
Dev Neurobiol ; 67(3): 270-84, 2007 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-17443787

RESUMO

Precursor cells of the human central nervous system can be cultured in vitro to reveal pathogenesis of diseases or developmental disorders. Here, we have studied the biology of neural precursor cells (NPCs) from patients of lethal congenital contracture syndrome (LCCS), a severe motoneuron disease leading to prenatal death before the 32nd gestational week. LCCS fetuses are immobile because of a motoneuron defect, seen as degeneration of the anterior horn and descending tracts of the developing spinal cord. The genetic defect for the syndrome is unknown. We show that NPCs isolated postmortem from LCCS fetuses grow and are maintained in culture, but display increased cell cycle activity. Global transcript analysis of undifferentiated LCCS precursor cells present with changes in EGF-related signaling when compared with healthy age-matched human controls. Further, we show that LCCS-derived NPCs differentiate into cells of neuronal and glial lineage and that the initial differentiation is not accompanied by overt apoptosis. Cells expressing markers Islet-1 and Hb9 are also generated from the LCCS NPCs, suggesting that the pathogenic mechanism of LCCS does not directly affect the differentiation capacity or survival of the cells, but the absence of motoneurons in LCCS may be caused by a noncell autonomous mechanism.


Assuntos
Diferenciação Celular/fisiologia , Feto/patologia , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Doença dos Neurônios Motores/patologia , Neurônios/citologia , Células-Tronco/fisiologia , Apoptose , Bromodesoxiuridina/metabolismo , Células Cultivadas , Humanos , Análise em Microsséries/métodos , Células-Tronco/patologia
18.
Physiol Genomics ; 28(2): 179-92, 2007 Jan 17.
Artigo em Inglês | MEDLINE | ID: mdl-17234577

RESUMO

The in vivo functions of lymphatic endothelial cells depend on their microenvironment, which cannot be fully reproduced in vitro. Because of technical limitations, gene expression in uncultured, "ex vivo" lymphatic endothelial cells has not been characterized at the molecular level. We combined tissue micropreparation and direct cell isolation with DNA chip experiments to identify 159 genes differentiating human lymphatic endothelial cells from blood vascular endothelial cells ex vivo. The same analysis performed with cultured primary cells revealed that only 19 genes characteristic for lymphatic endothelium ex vivo retained this property upon culture, while 27 marker genes were newly induced. In addition, a set of panendothelial genes could be recognized. The propagation of lymphatic endothelial cells in culture stimulated transcription of genes associated with cell turnover, basic metabolism, and the cytoskeleton. On the other hand, there was downregulation of genes encoding extracellular matrix components, signaling via transmembrane tyrosine kinase pathways and the chemokine (C-C) ligand 21. Direct ex vivo analysis of the lymphatic endothelial cell transcriptome is helpful for the understanding of the physiology of the lymphatic vascular system and of the pathogenesis of its diseases.


Assuntos
Células Endoteliais/metabolismo , Endotélio Linfático/metabolismo , Perfilação da Expressão Gênica/métodos , Transcrição Gênica , Células Cultivadas , Derme/citologia , Derme/metabolismo , Células Endoteliais/citologia , Endotélio Linfático/citologia , Regulação da Expressão Gênica , Humanos , Imuno-Histoquímica , Lectinas Tipo C/genética , Lectinas Tipo C/metabolismo , Receptor de Manose , Lectinas de Ligação a Manose/genética , Lectinas de Ligação a Manose/metabolismo , Glicoproteínas de Membrana/genética , Modelos Genéticos , Modelos Teóricos , Análise de Sequência com Séries de Oligonucleotídeos/métodos , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa
19.
Nature ; 439(7079): 1009-13, 2006 Feb 23.
Artigo em Inglês | MEDLINE | ID: mdl-16496002

RESUMO

Many high-throughput loss-of-function analyses of the eukaryotic cell cycle have relied on the unicellular yeast species Saccharomyces cerevisiae and Schizosaccharomyces pombe. In multicellular organisms, however, additional control mechanisms regulate the cell cycle to specify the size of the organism and its constituent organs. To identify such genes, here we analysed the effect of the loss of function of 70% of Drosophila genes (including 90% of genes conserved in human) on cell-cycle progression of S2 cells using flow cytometry. To address redundancy, we also targeted genes involved in protein phosphorylation simultaneously with their homologues. We identify genes that control cell size, cytokinesis, cell death and/or apoptosis, and the G1 and G2/M phases of the cell cycle. Classification of the genes into pathways by unsupervised hierarchical clustering on the basis of these phenotypes shows that, in addition to classical regulatory mechanisms such as Myc/Max, Cyclin/Cdk and E2F, cell-cycle progression in S2 cells is controlled by vesicular and nuclear transport proteins, COP9 signalosome activity and four extracellular-signal-regulated pathways (Wnt, p38betaMAPK, FRAP/TOR and JAK/STAT). In addition, by simultaneously analysing several phenotypes, we identify a translational regulator, eIF-3p66, that specifically affects the Cyclin/Cdk pathway activity.


Assuntos
Ciclo Celular/genética , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/citologia , Drosophila melanogaster/genética , Interferência de RNA , Transporte Ativo do Núcleo Celular , Animais , Linhagem Celular , Tamanho Celular , Sequência Conservada/genética , Drosophila melanogaster/metabolismo , Biblioteca Gênica , Genes de Insetos/genética , Humanos , Fenótipo , Fosforilação , Transdução de Sinais
20.
Cancer Res ; 66(2): 743-7, 2006 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-16424004

RESUMO

Several candidate genes along androgen pathway have been suggested to affect prostate cancer risk but no single gene seems to be overwhelmingly important for a large fraction of the patients. In this study, we first screened for variants in candidate genes and then chose to explore the association between 18 variants and prostate cancer risk by genotyping DNA samples from unselected (n = 847) and familial (n = 121) prostate cancer patients and population controls (n = 923). We identified a novel single nucleotide polymorphism (SNP) in the CYP19A1 gene, T201M, with a mild significant association with prostate cancer [odds ratio (OR), 2.04; 95% confidence interval (95% CI), 1.03-4.03; P = 0.04]. Stratified analysis revealed that this risk was most apparent in patients with organ-confined (T(1)-T(2)) and low-grade (WHO grade 1) tumors (OR, 5.42; 95% CI, 2.33-12.6; P < 0.0001). In contrast, CYP17A1 -34T>C alteration was associated with moderate to poorly differentiated (WHO grade 2-3) organ-confined disease (OR, 1.42; 95% CI, 1.09-1.83; P = 0.007). We also tested a multigenic model of prostate cancer risk by calculating the joint effect of CYP19A1 T201M with five other common SNPs. Individuals carrying both the CYP19A1 and KLK3 -252A>G variant alleles had a significantly increased risk for prostate cancer (OR, 2.87; 95% CI, 1.10-7.49; P = 0.03). In conclusion, our results suggest that several SNPs along the androgen pathway, especially in CYP19A1 and CYP17A1, may influence prostate cancer development and progression. These genes may have different contributions to distinct clinical subsets as well as combinatorial effects in others illustrating that profiling and joint analysis of several genes along each pathway may be needed to understand genetic contributions to prostate cancer etiology.


Assuntos
Androgênios/biossíntese , Aromatase/genética , Polimorfismo de Nucleotídeo Único , Neoplasias da Próstata/genética , Esteroide 17-alfa-Hidroxilase/genética , Idoso , Predisposição Genética para Doença , Humanos , Masculino , Pessoa de Meia-Idade , Razão de Chances , Neoplasias da Próstata/fisiopatologia , Fatores de Risco
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA