Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cell Rep ; 25(5): 1135-1145.e5, 2018 10 30.
Artigo em Inglês | MEDLINE | ID: mdl-30380406

RESUMO

RNase H2 has two distinct functions: initiation of the ribonucleotide excision repair (RER) pathway by cleaving ribonucleotides (rNMPs) incorporated during DNA replication and processing the RNA portion of an R-loop formed during transcription. An RNase H2 mutant lacking RER activity but supporting R-loop removal revealed that rNMPs in DNA initiate p53-dependent DNA damage response and early embryonic arrest in mouse. However, an RNase H2 AGS-related mutant with residual RER activity develops to birth. Estimations of the number of rNMPs in DNA in these two mutants define a ribonucleotide threshold above which p53 induces apoptosis. Below the threshold, rNMPs in DNA trigger an innate immune response. Compound heterozygous cells, containing both defective enzymes, retain rNMPs above the threshold, indicative of competition for RER substrates between active and inactive enzymes, suggesting that patients with compound heterozygous mutations in RNASEH2 genes may not reflect the properties of recombinantly expressed proteins.


Assuntos
Desenvolvimento Embrionário , Mutação/genética , Ribonuclease H/genética , Ribonucleotídeos/metabolismo , Animais , DNA/metabolismo , Dano ao DNA , Reparo do DNA/efeitos dos fármacos , Perda do Embrião/patologia , Embrião de Mamíferos/anormalidades , Desenvolvimento Embrionário/efeitos dos fármacos , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Interferons/farmacologia , Proteínas de Membrana/metabolismo , Camundongos Knockout , Proteínas Mutantes/metabolismo , Estabilidade de RNA/efeitos dos fármacos , Ribonuclease H/metabolismo , Proteína Supressora de Tumor p53/metabolismo
2.
Mol Biotechnol ; 54(2): 350-60, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22706789

RESUMO

Adrenaline and noradrenaline are important neurotransmitter hormones that mediate physiological stress responses in adult mammals, and are essential for cardiovascular function during a critical period of embryonic/fetal development. In this study, we describe a novel mouse model system for identifying and characterizing adrenergic cells. Specifically, we generated a reporter mouse strain in which a nuclear-localized enhanced green fluorescent protein gene (nEGFP) was inserted into exon 1 of the gene encoding Phenylethanolamine n-methyltransferase (Pnmt), the enzyme responsible for production of adrenaline from noradrenaline. Our analysis demonstrates that this knock-in mutation effectively marks adrenergic cells in embryonic and adult mice. We see expression of nEGFP in Pnmt-expressing cells of the adrenal medulla in adult animals. We also note that nEGFP expression recapitulates the restricted expression of Pnmt in the embryonic heart. Finally, we show that nEGFP and Pnmt expressions are each induced in parallel during the in vitro differentiation of pluripotent mouse embryonic stem cells into beating cardiomyocytes. Thus, this new mouse genetic model should be useful for the identification and functional characterization of adrenergic cells in vitro and in vivo.


Assuntos
Medula Suprarrenal/metabolismo , Genes Reporter/genética , Proteínas de Fluorescência Verde/genética , Medula Suprarrenal/citologia , Animais , Células-Tronco Embrionárias/metabolismo , Epinefrina/genética , Epinefrina/metabolismo , Expressão Gênica , Proteínas de Fluorescência Verde/metabolismo , Camundongos , Mutação , Miócitos Cardíacos/metabolismo , Norepinefrina/genética , Norepinefrina/metabolismo , Feniletanolamina N-Metiltransferase/genética , Feniletanolamina N-Metiltransferase/metabolismo , Células-Tronco Pluripotentes/metabolismo
3.
PLoS One ; 7(2): e31991, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22384122

RESUMO

Pannexin1 (Panx1) forms large nonselective membrane channel that is implicated in paracrine and inflammatory signaling. In vitro experiments suggested that Panx1 could play a key role in ischemic death of hippocampal neurons. Since retinal ganglion cells (RGCs) express high levels of Panx1 and are susceptible to ischemic induced injury, we hypothesized that Panx1 contributes to rapid and selective loss of these neurons in ischemia. To test this hypothesis, we induced experimental retinal ischemia followed by reperfusion in live animals with the Panx1 channel genetically ablated either in the entire mouse (Panx1 KO), or only in neurons using the conditional knockout (Panx1 CKO) technology. Here we report that two distinct neurotoxic processes are induced in RGCs by ischemia in the wild type mice but are inactivated in Panx1KO and Panx1 CKO animals. First, the post-ischemic permeation of RGC plasma membranes is suppressed, as assessed by dye transfer and calcium imaging assays ex vivo and in vitro. Second, the inflammasome-mediated activation of caspase-1 and the production of interleukin-1ß in the Panx1 KO retinas are inhibited. Our findings indicate that post-ischemic neurotoxicity in the retina is mediated by previously uncharacterized pathways, which involve neuronal Panx1 and are intrinsic to RGCs. Thus, our work presents the in vivo evidence for neurotoxicity elicited by neuronal Panx1, and identifies this channel as a new therapeutic target in ischemic pathologies.


Assuntos
Conexinas/genética , Isquemia/patologia , Proteínas do Tecido Nervoso/genética , Neurônios Retinianos/metabolismo , Alelos , Animais , Cálcio/metabolismo , Conexinas/metabolismo , Glucose/química , Hipocampo/metabolismo , Inflamação , Interleucina-1beta/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas do Tecido Nervoso/metabolismo , Oxigênio/química , Traumatismo por Reperfusão , Retina/metabolismo , Células Ganglionares da Retina/citologia
4.
Mol Cell Biol ; 30(5): 1108-15, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20038532

RESUMO

The 2.4-kb H19 imprinting control region (H19ICR) is required to establish parent-of-origin-specific epigenetic marks and expression patterns at the Igf2/H19 locus. H19ICR activity is regulated by DNA methylation. The ICR is methylated in sperm but not in oocytes, and this paternal chromosome-specific methylation is maintained throughout development. We recently showed that the H19ICR can work as an ICR even when inserted into the normally nonimprinted alpha fetoprotein locus. Paternal but not maternal copies of the ICR become methylated in somatic tissue. However, the ectopic ICR remains unmethylated in sperm. To extend these findings and investigate the mechanisms that lead to methylation of the H19ICR in the male germ line, we characterized novel mouse knock-in lines. Our data confirm that the 2.4-kb element is an autonomously acting ICR whose function is not dependent on germ line methylation. Ectopic ICRs become methylated in the male germ line, but the timing of methylation is influenced by the insertion site and by additional genetic information. Our results support the idea that DNA methylation is not the primary genomic imprint and that the H19ICR insertion is sufficient to transmit parent-of-origin-dependent DNA methylation patterns independent of its methylation status in sperm.


Assuntos
Metilação de DNA , Impressão Genômica , RNA não Traduzido/genética , Espermatozoides/metabolismo , Animais , Sequência de Bases , Complexo CD3/genética , Cromossomos Artificiais Bacterianos/genética , Ilhas de CpG , Primers do DNA/genética , Feminino , Fertilização , Fator de Crescimento Insulin-Like II/genética , Masculino , Camundongos , Camundongos Transgênicos , Oócitos , Gravidez , Regiões Promotoras Genéticas , RNA Longo não Codificante , Espermatogênese/genética , alfa-Fetoproteínas/genética
5.
J Clin Invest ; 119(8): 2317-29, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19662682

RESUMO

Psoriasis is a common immune-mediated chronic inflammatory skin disorder, but the mechanisms of pathogenesis are still poorly understood. IL-23 is expressed in psoriatic skin, and IL-23 injection produces IL-22-dependent psoriasiform changes in mouse skin. Th17 cells produce IL-22 and display CCR6, the CCL20 receptor; CCR6+ T cells and CCL20 are abundant in psoriatic skin. We investigated a possible role for CCR6 in recruiting Th17 cells and producing psoriasiform pathology by injecting IL-23 into the skin of WT and Ccr6-/- mice. Unlike for WT mice, IL-23-injected ears of Ccr6-/- mice showed neither substantial epidermal/dermal changes nor increased Il22 mRNA expression. However, injection of IL-22 yielded equivalent psoriasiform changes in WT and Ccr6-/- mice. Surprisingly, IL-23-injected ears of WT and Ccr6-/- mice contained similar numbers of Th cells able to make IL-17A and/or IL-22. Furthermore, in ears of Rag1-/- mice, IL-23 initially induced skin changes and levels of Il22 mRNA that were indistinguishable from WT mice, revealing at least one non-T cell source for IL-22. We conclude that CCR6 is essential in a model of IL-23-induced, IL-22-mediated dermatitis, which develops in sequential T cell-independent and T cell-dependent phases. These findings reveal an expanded role for CCR6 in IL-23-related responses and identify CCR6 as a potential therapeutic target in psoriasis.


Assuntos
Interleucina-23/toxicidade , Psoríase/etiologia , Receptores CCR6/fisiologia , Animais , Linfócitos T CD4-Positivos/fisiologia , Células Dendríticas/fisiologia , Proteínas de Homeodomínio/fisiologia , Interleucina-17/biossíntese , Interleucinas/biossíntese , Camundongos , Camundongos Endogâmicos C57BL , Interleucina 22
6.
Proc Natl Acad Sci U S A ; 104(32): 13182-6, 2007 Aug 07.
Artigo em Inglês | MEDLINE | ID: mdl-17664423

RESUMO

Purkinje cells are one of the major types of neurons that form the neural circuitry in the cerebellum essential for fine control of movement and posture. During development, Purkinje cells also are critically involved in the regulation of proliferation of progenitors of granule cells, the other major type of neurons in the cerebellum. The process that controls differentiation of Purkinje cells from their early precursors is poorly understood. Here we report that two closely related LIM-homeobox genes, Lhx1 and Lhx5, were expressed in the developing Purkinje cells soon after they exited the cell cycle and migrated out of the cerebellar ventricular zone. Double-mutant mice lacking function of both Lhx1 and Lhx5 showed a severe reduction in the number of Purkinje cells. In addition, targeted inactivation of Ldb1, which encodes a cofactor for all LIM-homeodomain proteins, resulted in a similar phenotype. Our studies thus provide evidence that these transcription regulators are essential for controlling Purkinje cell differentiation in the developing mammalian cerebellum.


Assuntos
Cerebelo/embriologia , Proteínas de Ligação a DNA/fisiologia , Proteínas de Homeodomínio/fisiologia , Proteínas do Tecido Nervoso/fisiologia , Células de Purkinje/citologia , Fatores de Transcrição/fisiologia , Animais , Diferenciação Celular , Feminino , Proteínas com Domínio LIM , Proteínas com Homeodomínio LIM , Camundongos , Camundongos Endogâmicos C57BL , Gravidez
7.
J Immunol ; 177(10): 6613-25, 2006 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-17082573

RESUMO

TCR signals drive thymocyte development, but it remains controversial what impact, if any, the intensity of those signals have on T cell differentiation in the thymus. In this study, we assess the impact of CD8 coreceptor signal strength on positive selection and CD4/CD8 lineage choice using novel gene knockin mice in which the endogenous CD8alpha gene has been re-engineered to encode the stronger signaling cytoplasmic tail of CD4, with the re-engineered CD8alpha gene referred to as CD8.4. We found that stronger signaling CD8.4 coreceptors specifically improved the efficiency of CD8-dependent positive selection and quantitatively increased the number of MHC class I (MHC-I)-specific thymocytes signaled to differentiate into CD8+ T cells, even for thymocytes expressing a single, transgenic TCR. Importantly, however, stronger signaling CD8.4 coreceptors did not alter the CD8 lineage choice of any MHC-I-specific thymocytes, even MHC-I-specific thymocytes expressing the high-affinity F5 transgenic TCR. This study documents in a physiologic in vivo model that coreceptor signal strength alters TCR-signaling thresholds for positive selection and so is a major determinant of the CD4:CD8 ratio, but it does not influence CD4/CD8 lineage choice.


Assuntos
Antígenos CD4/fisiologia , Linfócitos T CD4-Positivos/imunologia , Antígenos CD8/fisiologia , Linfócitos T CD8-Positivos/imunologia , Diferenciação Celular/imunologia , Linhagem da Célula/imunologia , Modelos Imunológicos , Transdução de Sinais/imunologia , Animais , Antígenos CD4/biossíntese , Antígenos CD4/genética , Linfócitos T CD4-Positivos/citologia , Linfócitos T CD4-Positivos/metabolismo , Antígenos CD8/biossíntese , Antígenos CD8/genética , Linfócitos T CD8-Positivos/citologia , Linfócitos T CD8-Positivos/metabolismo , Diferenciação Celular/genética , Linhagem Celular , Linhagem da Célula/genética , Feminino , Células Matadoras Naturais/citologia , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Camundongos Transgênicos , Proteínas Recombinantes de Fusão/biossíntese , Proteínas Recombinantes de Fusão/genética , Transdução de Sinais/genética , Linfócitos T Auxiliares-Indutores/citologia , Linfócitos T Auxiliares-Indutores/imunologia , Linfócitos T Auxiliares-Indutores/metabolismo , Regulação para Cima/imunologia
8.
Development ; 133(11): 2149-54, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16672341

RESUMO

The Dkk family of secreted cysteine-rich proteins regulates Wnt/beta-catenin signaling by interacting with the Wnt co-receptor Lrp5/6. Here, we show that Dkk2-mediated repression of the Wnt/beta-catenin pathway is essential to promote differentiation of the corneal epithelial progenitor cells into a non-keratinizing stratified epithelium. Complete transformation of the corneal epithelium into a stratified epithelium that expresses epidermal-specific differentiation markers and develops appendages such as hair follicles is achieved in the absence of the Dkk2 gene function. We show that Dkk2 is a key regulator of the corneal versus epidermal fate of the ocular surface epithelium.


Assuntos
Córnea/crescimento & desenvolvimento , Córnea/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Animais , Biomarcadores , Córnea/embriologia , Epiderme/crescimento & desenvolvimento , Epiderme/metabolismo , Epitélio/embriologia , Epitélio/crescimento & desenvolvimento , Epitélio/metabolismo , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Peptídeos e Proteínas de Sinalização Intercelular/deficiência , Peptídeos e Proteínas de Sinalização Intercelular/genética , Queratinócitos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mutação/genética , Fenótipo
9.
Hum Mol Genet ; 15(6): 839-51, 2006 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-16446309

RESUMO

Smith-Lemli-Opitz syndrome (SLOS) is a genetic syndrome caused by mutations in the 3beta-hydroxysterol Delta(7)-reductase gene (DHCR7). SLOS patients have decreased cholesterol and increased 7-dehydrocholesterol (7-DHC) levels. Dietary cholesterol supplementation improves systemic biochemical abnormalities; however, because of the blood-brain barrier, the central nervous system (CNS) is not treated. Simvastatin therapy has been proposed as a means to treat the CNS. Mice homozygous for a null disruption of Dhcr7, Dhcr7(Delta3-5/Delta3-5), die soon after birth, thus they cannot be used to study postnatal development or therapy. To circumvent this problem, we produced a hypomorphic SLOS mouse model by introducing a mutation corresponding to DHCR7(T93M). Both Dhcr7(T93M/T93M) and Dhcr7(Delta3-5/T93M) mice are viable. Phenotypic findings in Dhcr7(T93M/Delta3-5) mice include CNS ventricular dilatation and two to three syndactyly. Biochemically, both Dhcr7(T93M/T93M) and Dhcr7(T93M/Delta3-5) mice have elevated tissue 7-DHC levels; however, the biochemical defect improved with age. This has not been observed in human patients, and is due to elevated Dhcr7 expression in mouse tissues. Dietary cholesterol therapy improved sterol profiles in peripheral, but not CNS tissues. However, treatment of Dhcr7(T93M/Delta3-5) mice with simvastatin decreased 7-DHC levels in both peripheral and brain tissues. Expression of Dhcr7 increased in Dhcr7(T93M/Delta3-5) tissues after simvastatin therapy, consistent with the hypothesis that simvastatin therapy improves the biochemical phenotype by increasing the expression of a Dhcr7 allele with residual enzymatic activity. We conclude that simvastatin treatment is efficacious in improving the SLOS-associated sterol abnormality found in the brain, and thus has the potential to be an effective therapeutic intervention for behavioral and learning problems associated with SLOS.


Assuntos
Modelos Animais de Doenças , Sinvastatina/uso terapêutico , Síndrome de Smith-Lemli-Opitz/tratamento farmacológico , Síndrome de Smith-Lemli-Opitz/genética , Células 3T3 , Alelos , Substituição de Aminoácidos/genética , Animais , Células Cultivadas , Cruzamentos Genéticos , Células-Tronco Embrionárias/transplante , Feminino , Espectroscopia de Ressonância Magnética , Metionina/genética , Camundongos , Camundongos Endogâmicos C57BL , Mutagênese Sítio-Dirigida , Oxirredutases atuantes sobre Doadores de Grupo CH-CH/genética , Fenótipo , Síndrome de Smith-Lemli-Opitz/enzimologia , Treonina/genética
10.
Mol Cell Biol ; 25(21): 9318-23, 2005 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16227583

RESUMO

The heterotetrameric adaptor protein (AP) complexes AP-1, AP-2, AP-3, and AP-4 play key roles in transport vesicle formation and cargo sorting in post-Golgi trafficking pathways. Studies on cultured mammalian cells have shown that AP-2 mediates rapid endocytosis of a subset of plasma membrane receptors. To determine whether this function is essential in the context of a whole mammalian organism, we carried out targeted disruption of the gene encoding the mu2 subunit of AP-2 in the mouse. We found that mu2 heterozygous mutant mice were viable and had an apparently normal phenotype. In contrast, no mu2 homozygous mutant embryos were identified among blastocysts from intercrossed heterozygotes, indicating that mu2-deficient embryos die before day 3.5 postcoitus (E3.5). These results indicate that AP-2 is indispensable for early embryonic development, which might be due to its requirement for cell viability.


Assuntos
Complexo 2 de Proteínas Adaptadoras/metabolismo , Subunidades mu do Complexo de Proteínas Adaptadoras/metabolismo , Complexo 2 de Proteínas Adaptadoras/genética , Subunidades mu do Complexo de Proteínas Adaptadoras/genética , Animais , Sobrevivência Celular , Células Cultivadas , Embrião de Mamíferos/citologia , Desenvolvimento Embrionário , Endocitose , Fibroblastos/citologia , Fibroblastos/fisiologia , Heterozigoto , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
11.
J Am Soc Nephrol ; 15(12): 3035-43, 2004 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-15579506

RESUMO

Gene targeting has been used to create a variety of lines of mice with Pkd1 mutations that share many common features. Homozygous Pkd1 mutants invariably develop pancreatic and renal cysts if they survive to day 15.5 post coitum and die in either the fetal or the perinatal period. In contrast, mice with heterozygous mutations of Pkd1 are generally normal and have few if any renal cysts. These features have limited the utility of these models as tools to study the pathogenesis of cyst formation and the effect of various therapeutic interventions on disease progression. This report describes a new line of mice with a floxed allele of Pkd1 (Pkd1(cond)) that has an FRT-flanked neomycin cassette inserted into intron 1 and lox P sites inserted into intron 1 and intron 4. The Pkd1(cond) allele is fully functional, and homozygotes are viable and healthy. It is shown that the lox P and FRT sites can be selectively induced to recombine to produce two new alleles, Pkd1(del2-4) and Pkd1(cond-Deltaneo), by crossing to animals that express either the cre or FLPe recombinase, respectively. It is found that Pkd1(del2-4) allele functions as a true null, whereas presence or absence of the neomycin gene has no functional effects. It also is shown that somatic loss of Pkd1 results in renal and hepatic cysts. This new line of mice will be invaluable in the study of Pkd1 biology and serve as a powerful new tool that can be used to study the pathogenesis of autosomal dominant polycystic kidney disease.


Assuntos
Mutagênese Insercional/métodos , Rim Policístico Autossômico Dominante/genética , Rim Policístico Autossômico Dominante/fisiopatologia , Proteínas/genética , Proteínas/metabolismo , Alelos , Animais , Modelos Animais de Doenças , Rim/patologia , Fígado/patologia , Camundongos , Camundongos Mutantes , Neomicina , Rim Policístico Autossômico Dominante/patologia , Canais de Cátion TRPP
12.
Dev Dyn ; 231(4): 849-58, 2004 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-15517585

RESUMO

To evaluate the developmental distribution of adrenergic cells in vivo, we inserted the Cre-recombinase gene into the locus encoding for the epinephrine biosynthetic enzyme phenylethanolamine n-methyltransferase (Pnmt) and crossed these Pnmt-Cre mice with ROSA26 reporter (R26R) mice to activate LacZ (encoding beta-galactosidase) expression in cells that were selectively derived from the adrenergic lineage. Our data show the following: (1) Insertion of Cre-recombinase into the Pnmt locus created a functional knockout of Pnmt expression with concomitant loss of epinephrine in homozygous Pnmt(Cre/Cre) mice; (2) Despite the reduction in Pnmt expression and epinephrine production in Pnmt(Cre/Cre) mice, these mice were viable and fertile, with no apparent developmental defects; (3) When crossed with R26R mice, Pnmt-Cre activation of LacZ expression faithfully recapitulated Pnmt expression in vivo; and (4) LacZ expression was activated in substantial numbers of pacemaking, conduction, and working cardiomyocytes.


Assuntos
Glândulas Suprarrenais/embriologia , Epinefrina/fisiologia , Integrases/genética , Feniletanolamina N-Metiltransferase/genética , Glândulas Suprarrenais/citologia , Glândulas Suprarrenais/fisiologia , Animais , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Regulação Enzimológica da Expressão Gênica , Genes Reporter , Óperon Lac , Masculino , Camundongos , Camundongos Knockout , Modelos Animais , Mutagênese Insercional , Gravidez
13.
Genomics ; 84(3): 555-64, 2004 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-15498462

RESUMO

Inherited long QT syndrome is most frequently associated with mutations in KCNQ1, which encodes the primary subunit of a potassium channel. Patients with mutations in KCNQ1 may show only the cardiac defect (Romano-Ward syndrome or RWS) or may also have severe deafness (Jervell and Lange-Nielsen syndrome or JLNS). Targeted disruption of mouse Kcnq1 models JLNS in that mice are deaf and show abnormal ECGs. However, the phenotype is broader than that seen in patients. Most dramatically, the inner ear defects result in a severe hyperactivity/circling behavior, which may influence cardiac function. To understand the etiology of the cardiac phenotype in these mice and to generate a potentially more useful model system, we generated new mouse lines by introducing point mutations associated with RWS. The A340E line phenocopies RWS: the repolarization phenotype is inherited in a dominant manner and is observed independent of any inner ear defect. The T311I line phenocopies JLNS, with deafness associated with inner hair cell malfunction.


Assuntos
Modelos Animais de Doenças , Camundongos/genética , Fenótipo , Canais de Potássio de Abertura Dependente da Tensão da Membrana/genética , Síndrome de Romano-Ward/genética , Animais , Northern Blotting , Primers do DNA , Surdez/genética , Eletrocardiografia , Potenciais Evocados Auditivos do Tronco Encefálico , Células Ciliadas Auditivas Internas/patologia , Canais de Potássio KCNQ , Canal de Potássio KCNQ1 , Mutagênese Sítio-Dirigida
14.
Mol Cell Biol ; 24(9): 3588-95, 2004 May.
Artigo em Inglês | MEDLINE | ID: mdl-15082756

RESUMO

Igf2 and H19 are coordinately regulated imprinted genes physically linked on the distal end of mouse chromosome 7. Genetic analyses demonstrate that the differentially methylated region (DMR) upstream of the H19 gene is necessary for three distinct functions: transcriptional insulation of the maternal Igf2 allele, transcriptional silencing of paternal H19 allele, and marking of the parental origin of the two chromosomes. To test the sufficiency of the DMR for the third function, we inserted DMR at two heterologous positions in the genome, downstream of H19 and at the alpha-fetoprotein locus on chromosome 5. Our results demonstrate that the DMR alone is sufficient to act as a mark of parental origin. Moreover, this activity is not dependent on germ line differences in DMR methylation. Thus, the DMR can mark its parental origin by a mechanism independent of its own DNA methylation.


Assuntos
Cromossomos/metabolismo , Metilação de DNA , Impressão Genômica , Proteínas/genética , RNA não Traduzido/genética , Animais , Cromossomos/genética , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Inativação Gênica , Humanos , Masculino , Camundongos , Biossíntese de Proteínas , Proteínas/metabolismo , RNA Longo não Codificante
15.
Proc Natl Acad Sci U S A ; 100(16): 9566-71, 2003 Aug 05.
Artigo em Inglês | MEDLINE | ID: mdl-12886012

RESUMO

Neural progenitor cells are widespread throughout the adult central nervous system but only give rise to neurons in specific loci. Negative regulators of neurogenesis have therefore been postulated, but none have yet been identified as subserving a significant role in the adult brain. Here we report that nitric oxide (NO) acts as an important negative regulator of cell proliferation in the adult mammalian brain. We used two independent approaches to examine the function of NO in adult neurogenesis. In a pharmacological approach, we suppressed NO production in the rat brain by intraventricular infusion of an NO synthase inhibitor. In a genetic approach, we generated a null mutant neuronal NO synthase knockout mouse line by targeting the exon encoding active center of the enzyme. In both models, the number of new cells generated in neurogenic areas of the adult brain, the olfactory subependyma and the dentate gyrus, was strongly augmented, which indicates that division of neural stem cells in the adult brain is controlled by NO and suggests a strategy for enhancing neurogenesis in the adult central nervous system.


Assuntos
Neurônios/metabolismo , Neurônios/fisiologia , Óxido Nítrico/metabolismo , Animais , Encéfalo/metabolismo , Bromodesoxiuridina/farmacologia , Divisão Celular , Sistema Nervoso Central/metabolismo , Éxons , Marcação In Situ das Extremidades Cortadas , Camundongos , Camundongos Knockout , Microscopia Confocal , Modelos Genéticos , Dados de Sequência Molecular , NG-Nitroarginina Metil Éster/farmacologia , Óxido Nítrico Sintase/metabolismo , Fases de Leitura Aberta , Ratos , Recombinação Genética
16.
Hum Mol Genet ; 12(13): 1631-41, 2003 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-12812989

RESUMO

Lathosterol 5-desaturase catalyzes the conversion of lathosterol to 7-dehydrocholesterol in the next to last step of cholesterol synthesis. Inborn errors of cholesterol synthesis underlie a group of human malformation syndromes including Smith-Lemli-Opitz syndrome, desmosterolosis, CHILD syndrome, CDPX2 and lathosterolosis. We disrupted the lathosterol 5-desaturase gene (Sc5d ) in order to further our understanding of the pathophysiological processes underlying these disorders and to gain insight into the corresponding human disorder. Sc5d (-/-) pups were stillborn, had elevated lathosterol and decreased cholesterol levels, had craniofacial defects including cleft palate and micrognathia, and limb patterning defects. Many of the malformations found in Sc5d (-/-) mice are consistent with impaired hedgehog signaling, and appear to be a result of decreased cholesterol rather than increased lathosterol. A patient initially described as atypical SLOS with mucolipidosis was shown to have lathosterolosis by biochemical and molecular analysis. We identified a homozygous mutation of SC5D (137A>C, Y46S) in this patient. An unique aspect of the lathosterolosis phenotype is the combination of a malformation syndrome with an intracellular storage defect.


Assuntos
Erros Inatos do Metabolismo Lipídico/genética , Oxirredutases atuantes sobre Doadores de Grupo CH-CH/deficiência , Oxirredutases atuantes sobre Doadores de Grupo CH-CH/genética , Síndrome de Smith-Lemli-Opitz/genética , Sequência de Aminoácidos , Animais , Colesterol/metabolismo , Fibroblastos/metabolismo , Fibroblastos/ultraestrutura , Genótipo , Homozigoto , Humanos , Recém-Nascido , Camundongos , Camundongos Transgênicos , Modelos Químicos , Modelos Genéticos , Dados de Sequência Molecular , Mutação , Fenótipo , Homologia de Sequência de Aminoácidos , Pele/patologia , Esteróis/metabolismo , Fatores de Tempo
17.
Mol Cell ; 11(3): 807-15, 2003 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-12667461

RESUMO

Although ribonucleases H (RNases H) have long been implicated in DNA metabolism, they are not required for viability in prokaryotes or unicellular eukaryotes. We generated Rnaseh1(-/-) mice to investigate the role of RNase H1 in mammals and observed developmental arrest at E8.5 in null embryos. A fraction of the mainly nuclear RNase H1 was targeted to mitochondria, and its absence in embryos resulted in a significant decrease in mitochondrial DNA content, leading to apoptotic cell death. This report links RNase H1 to generation of mitochondrial DNA, providing direct support for the strand-coupled mechanism of mitochondrial DNA replication. These findings also have important implications for therapy of mitochondrial dysfunctions and drug development for the structurally related RNase H of HIV.


Assuntos
DNA Mitocondrial/fisiologia , Ribonuclease H/genética , Ribonuclease H/fisiologia , Alelos , Sequência de Aminoácidos , Animais , Apoptose , Southern Blotting , Núcleo Celular/metabolismo , DNA/biossíntese , DNA/metabolismo , Transporte de Elétrons , Vetores Genéticos , Genótipo , Homozigoto , Marcação In Situ das Extremidades Cortadas , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Modelos Genéticos , Dados de Sequência Molecular , Plasmídeos/metabolismo , Reação em Cadeia da Polimerase , Ribonuclease H/química , Ribonuclease H/metabolismo , Fatores de Tempo
18.
Development ; 130(3): 495-505, 2003 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-12490556

RESUMO

The LIM domain-binding protein 1 (Ldb1) is found in multi-protein complexes containing various combinations of LIM-homeodomain, LIM-only, bHLH, GATA and Otx transcription factors. These proteins exert key functions during embryogenesis. Here we show that targeted deletion of the Ldb1 gene in mice results in a pleiotropic phenotype. There is no heart anlage and head structures are truncated anterior to the hindbrain. In about 40% of the mutants, posterior axis duplication is observed. There are also severe defects in mesoderm-derived extraembryonic structures, including the allantois, blood islands of the yolk sack, primordial germ cells and the amnion. Abnormal organizer gene expression during gastrulation may account for the observed axis defects in Ldb1 mutant embryos. The expression of several Wnt inhibitors is curtailed in the mutant, suggesting that Wnt pathways may be involved in axial patterning regulated by Ldb1.


Assuntos
Padronização Corporal/genética , Proteínas de Ligação a DNA/deficiência , Proteínas de Ligação a DNA/genética , Gástrula/citologia , Proteínas de Peixe-Zebra , Animais , Sequência de Bases , Padronização Corporal/fisiologia , DNA/genética , Proteínas de Ligação a DNA/fisiologia , Feminino , Coração Fetal/embriologia , Gástrula/metabolismo , Marcação de Genes , Proteínas com Domínio LIM , Masculino , Mesoderma/citologia , Camundongos , Camundongos Knockout , Oócitos/metabolismo , Fenótipo , Gravidez , Proteínas Proto-Oncogênicas/antagonistas & inibidores , Proteínas Proto-Oncogênicas/fisiologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Proteínas Wnt
19.
J Biol Chem ; 278(8): 5977-83, 2003 Feb 21.
Artigo em Inglês | MEDLINE | ID: mdl-12270940

RESUMO

Understanding the molecular basis of monoallelic expression as observed at imprinted loci is helpful in understanding the mechanisms underlying epigenetic regulation. Genomic imprinting begins during gametogenesis with the establishment of epigenetic marks on the chromosomes such that paternal and maternal chromosomes are rendered distinct. During embryonic development, the primary imprint can lead to generation of secondary epigenetic modifications (secondary imprints) of the chromosomes. Eventually, either the primary imprints or the secondary imprints interfere with transcription, leading to parent-of-origin-dependent silencing of one of the two alleles. Here we investigated several aspects pertaining to the generation and functional necessity of secondary methylation imprints at the Igf2/H19 locus. At the H19 locus, these secondary imprints are, in fact, the signals mediating paternal chromosome-specific silencing of that gene. We first demonstrated that the H19 secondary methylation imprints are entirely stable through multiple cell divisions, even in the absence of the primary imprint. Second, we generated mouse mutations to determine which DNA sequences are important in mediating establishment and maintenance of the silent state of the paternal H19 allele. Finally, we analyzed the dependence of the methylation of Igf2DMR1 region on the primary methylation imprint about 90 kilobases away.


Assuntos
Impressão Genômica , Fator de Crescimento Insulin-Like II/genética , Proteínas/genética , Animais , Sequência de Bases , Antígenos CD2/genética , Mapeamento Cromossômico , Metilação de DNA , Primers do DNA , Desenvolvimento Embrionário e Fetal , Éxons , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Ativação Linfocitária , Masculino , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Reação em Cadeia da Polimerase , RNA Longo não Codificante , RNA não Traduzido/genética , Mapeamento por Restrição , Linfócitos T/imunologia
20.
J Neurosci ; 22(24): 10801-10, 2002 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-12486173

RESUMO

Dopamine is an important transmitter in the CNS and PNS, critically regulating numerous neuropsychiatric and physiological functions. These actions of dopamine are mediated by five distinct receptor subtypes. Of these receptors, probably the least understood in terms of physiological functions is the D5 receptor subtype. To better understand the role of the D5 dopamine receptor (DAR) in normal physiology and behavior, we have now used gene-targeting technology to create mice that lack this receptor subtype. We find that the D5 receptor-deficient mice are viable and fertile and appear to develop normally. No compensatory alterations in other dopamine receptor subtypes were observed. We find, however, that the mutant mice develop hypertension and exhibit significantly elevated blood pressure (BP) by 3 months of age. This hypertension appears to be caused by increased sympathetic tone, primarily attributable to a CNS defect. Our data further suggest that this defect involves an oxytocin-dependent sensitization of V1 vasopressin and non-NMDA glutamatergic receptor-mediated pathways, potentially within the medulla, leading to increased sympathetic outflow. These results indicate that D5 dopamine receptors modulate neuronal pathways regulating blood pressure responses and may provide new insights into mechanisms for some forms of essential hypertension in humans, a disease that afflicts up to 25% of the aged adult population in industrialized societies.


Assuntos
Hipertensão/etiologia , Receptores de Dopamina D1/fisiologia , Sistema Nervoso Simpático/fisiopatologia , Glândulas Suprarrenais/química , Animais , Pressão Sanguínea , Encéfalo/metabolismo , Química Encefálica , Epinefrina/análise , Marcação de Genes , Hipertensão/metabolismo , Hipertensão/fisiopatologia , Imuno-Histoquímica , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Norepinefrina/análise , Ocitocina/genética , RNA Mensageiro/análise , Receptores de Dopamina D1/análise , Receptores de Dopamina D1/genética , Receptores de Dopamina D2/metabolismo , Receptores de Dopamina D5 , Receptores de Ocitocina/análise , Receptores de Vasopressinas/análise , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Vasopressinas/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...