Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Methods Enzymol ; 661: 95-120, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34776225

RESUMO

Maintenance and surveillance of genome integrity is crucial during the very early steps of embryonic development, since de novo mutations generated during this stage can be propagated in differentiated adult cells and may lead to predisposition to diseases including cancer. Surprisingly, early embryos are characterized by a relaxed control of genome integrity, reminiscent of that observed in cancer cells. How embryos manage to produce healthy adult individuals in such conditions remains still unclear. Here, we describe protocols and methods to study and analyze the DNA damage response and genome integrity in two embryonic experimental systems, early Xenopus laevis embryos and mouse embryonic stem cells. We describe methods to study gene functions in the DNA damage response by mRNA microinjection in Xenopus embryos generated by in vitro fertilization, mutagenesis and developmental regulation of the DNA damage response. We also describe methods to analyze the DNA damage response in mESCs, including synchronization experiments that allow studying the DNA damage response at different cell cycle stages. Analysis of genome integrity in these systems may also help to shed light on the molecular mechanisms that preserve genome integrity and become dysregulated in cancer cells.


Assuntos
Desenvolvimento Embrionário , Genoma , Animais , Dano ao DNA , Desenvolvimento Embrionário/genética , Feminino , Camundongos , Células-Tronco Embrionárias Murinas , Gravidez , Xenopus laevis/genética
2.
Pharmacol Ther ; 218: 107681, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-32961263

RESUMO

In the brain of patients with Alzheimer's disease (AD), the number and length of microtubules (MTs) are significantly and selectively reduced. MTs are involved in a wide range of cellular functions, and defects of the microtubular system have emerged as a unifying hypothesis for the heterogeneous and variable clinical presentations of AD. MTs orchestrate their numerous functions through the spatiotemporal regulation of the binding of specialised microtubule-associated proteins (MAPs) and molecular motors. Covalent posttranslational modifications (PTMs) on the tubulin C-termini that protrude at the surface of MTs regulate the binding of these effectors. In neurons, MAP tau is highly abundant and its abnormal dissociation from MTs in the axon, cellular mislocalization and hyperphosphorylation, are primary events leading to neuronal death. Consequently, compounds targeting tau phosphorylation or aggregation are currently evaluated but their clinical significance has not been demonstrated yet. In this review, we discuss the emerging link between tubulin PTMs and tau dysfunction. In neurons, high levels of glutamylation and detyrosination profoundly impact the physicochemical properties at the surface of MTs. Moreover, in patients with early-onset progressive neurodegeneration, deleterious mutations in enzymes involved in modifying MTs at the surface have recently been identified, underscoring the importance of this enzymatic machinery in neurology. We postulate that pharmacologically targeting the tubulin-modifying enzymes holds promise as therapeutic approach for the treatment of neurodegenerative diseases.


Assuntos
Doença de Alzheimer , Tubulina (Proteína) , Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/enzimologia , Humanos , Microtúbulos , Neurônios , Tubulina (Proteína)/metabolismo , Proteínas tau
3.
Cell Rep ; 29(12): 4159-4171.e6, 2019 12 17.
Artigo em Inglês | MEDLINE | ID: mdl-31851940

RESUMO

The two related members of the vasohibin family, VASH1 and VASH2, encode human tubulin detyrosinases. Here we demonstrate that, in contrast to VASH1, which requires binding of small vasohibin binding protein (SVBP), VASH2 has autonomous tubulin detyrosinating activity. Moreover, we demonstrate that SVBP acts as a bona fide activator of both enzymes. Phylogenetic analysis of the vasohibin family revealed that regulatory diversification of VASH-mediated tubulin detyrosination coincided with early vertebrate evolution. Thus, as a model organism for functional analysis, we used Trypanosoma brucei (Tb), an evolutionarily early-branched eukaryote that possesses a single VASH and encodes a terminal tyrosine on both α- and ß-tubulin tails, both subject to removal. Remarkably, although detyrosination levels are high in the flagellum, TbVASH knockout parasites did not present any noticeable flagellar abnormalities. In contrast, we observed reduced proliferation associated with profound morphological and mitotic defects, underscoring the importance of tubulin detyrosination in cell division.


Assuntos
Proteínas Angiogênicas/metabolismo , Evolução Biológica , Proteínas de Transporte/metabolismo , Proteínas de Ciclo Celular/metabolismo , Trypanosoma brucei brucei/metabolismo , Tirosina/metabolismo , Proteínas Angiogênicas/química , Proteínas Angiogênicas/genética , Proteínas de Transporte/química , Proteínas de Transporte/genética , Proteínas de Ciclo Celular/química , Proteínas de Ciclo Celular/genética , Cristalografia por Raios X , Flagelos/metabolismo , Células HEK293 , Humanos , Microtúbulos/metabolismo , Mitose , Filogenia , Conformação Proteica , Processamento de Proteína Pós-Traducional , Trypanosoma brucei brucei/genética , Trypanosoma brucei brucei/crescimento & desenvolvimento , Tirosina/química , Tirosina/genética
5.
Cell Rep ; 25(10): 2866-2877.e5, 2018 12 04.
Artigo em Inglês | MEDLINE | ID: mdl-30517872

RESUMO

Tubulin glutamylation is a reversible posttranslational modification that accumulates on stable microtubules (MTs). While abnormally high levels of this modification lead to a number of disorders such as male sterility, retinal degeneration, and neurodegeneration, very little is known about the molecular mechanisms underlying the regulation of glutamylase activity. Here, we found that CSAP forms a complex with TTLL5, and we demonstrate that the two proteins regulate their reciprocal abundance. Moreover, we show that CSAP increases TTLL5-mediated glutamylation and identify the TTLL5-interacting domain. Deletion of this domain leads to complete loss of CSAP activating function without impacting its MT binding. Binding of CSAP to TTLL5 promotes relocalization of TTLL5 toward MTs. Finally, we show that CSAP binds and activates all of the remaining autonomously active TTLL glutamylases. As such, we present CSAP as a major regulator of tubulin glutamylation and associated functions.


Assuntos
Proteínas de Transporte/metabolismo , Glutamatos/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo , Microtúbulos/metabolismo , Linhagem Celular , Humanos , Ligação Proteica , Tubulina (Proteína)/metabolismo
6.
Drug Discov Today ; 22(7): 1056-1063, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28188894

RESUMO

Unanticipated adverse drug reactions (ADRs) on the central nervous system are a major cause of clinical attrition and market withdrawal. Current practices for their prospective assessment still lean on extensive analysis of rodent behaviour despite their highly controversial predictive value. Human-derived in vitro models that objectively quantify mechanism-related biomarkers can greatly contribute to better ADR prediction at early developmental stages. Adenosine-to-inosine RNA editing constitutes a physiological cellular process that translates environmental cues by regulating protein function at the synaptic level in health and disease. Robust solutions based on NGS-based quantification of RNA editing biomarkers have emerged to predict the likelihood of treatment-related suicidal ideation and behaviour allowing cost-effective high-throughput drug screening as a strategy for risk mitigation.


Assuntos
Descoberta de Drogas , Edição de RNA , Animais , Biomarcadores , Humanos , Ideação Suicida
7.
Dev Cell ; 34(3): 364-72, 2015 Aug 10.
Artigo em Inglês | MEDLINE | ID: mdl-26212134

RESUMO

In early embryos, the DNA damage checkpoint is silent until the midblastula transition (MBT) because of maternal limiting factors of unknown identity. Here we identify the RAD18 ubiquitin ligase as one such factor in Xenopus. We show, in vitro and in vivo, that inactivation of RAD18 function leads to DNA damage-dependent checkpoint activation, monitored by CHK1 phosphorylation. Moreover, we show that the abundance of both RAD18 and PCNA monoubiquitylated (mUb) are developmentally regulated. Increased DNA abundance limits the availability of RAD18 close to the MBT, thereby reducing PCNA(mUb) and inducing checkpoint derepression. Furthermore, we show that this embryonic-like regulation can be reactivated in somatic mammalian cells by ectopic RAD18 expression, therefore conferring resistance to DNA damage. Finally, we find high RAD18 expression in cancer stem cells highly resistant to DNA damage. Together, these data propose RAD18 as a critical embryonic checkpoint-inhibiting factor and suggest that RAD18 deregulation may have unexpected oncogenic potential.


Assuntos
Dano ao DNA/genética , Reparo do DNA/genética , Proteínas de Ligação a DNA/metabolismo , DNA Polimerase Dirigida por DNA/metabolismo , Antígeno Nuclear de Célula em Proliferação/metabolismo , Proteínas de Xenopus/metabolismo , Xenopus laevis/embriologia , Animais , Proteínas de Ciclo Celular , Células Cultivadas , Quinase 1 do Ponto de Checagem , Proteínas de Ligação a DNA/biossíntese , Proteínas de Ligação a DNA/genética , Embrião não Mamífero , Dados de Sequência Molecular , Células-Tronco Neoplásicas/metabolismo , Óvulo/citologia , Fosforilação , Proteínas Quinases/metabolismo , Interferência de RNA , RNA Interferente Pequeno , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitinação , Proteínas de Xenopus/biossíntese , Proteínas de Xenopus/genética
8.
Genes (Basel) ; 5(1): 147-75, 2014 Mar 06.
Artigo em Inglês | MEDLINE | ID: mdl-24705291

RESUMO

The major challenge of the cell cycle is to deliver an intact, and fully duplicated, genetic material to the daughter cells. To this end, progression of DNA synthesis is monitored by a feedback mechanism known as replication checkpoint that is untimely linked to DNA replication. This signaling pathway ensures coordination of DNA synthesis with cell cycle progression. Failure to activate this checkpoint in response to perturbation of DNA synthesis (replication stress) results in forced cell division leading to chromosome fragmentation, aneuploidy, and genomic instability. In this review, we will describe current knowledge of the molecular determinants of the DNA replication checkpoint in eukaryotic cells and discuss a model of activation of this signaling pathway crucial for maintenance of genomic stability.

9.
PLoS One ; 9(4): e93663, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24695638

RESUMO

Pluripotency of embryonic stem cells (ESC) is tightly regulated by a network of transcription factors among which the estrogen-related receptor ß (Esrrb). Esrrb contributes to the relaxation of the G1 to S-phase (G1/S) checkpoint in mouse ESCs by transcriptional control of the deubiquitylase Dub3 gene, contributing to Cdc25A persistence after DNA damage. We show that in mESCs, Dub3 gene expression is cell cycle regulated and is maximal prior G1/S transition. In addition, following UV-induced DNA damage in G1, Dub3 expression markedly increases in S-phase also suggesting a role in checkpoint recovery. Unexpectedly, we also observed cell cycle-regulation of Nanog expression, and not Oct4, reaching high levels prior to G1/S transition, finely mirroring Cyclin E1 fluctuations. Curiously, while Esrrb showed only limited cell-cycle oscillations, transcript levels of the p160 family of nuclear receptor coactivators (NCoAs) displayed strong cell cycle-dependent fluctuations. Since NCoAs function in concert with Esrrb in transcriptional activation, we focussed on NCoA1 whose levels specifically increase prior onset of Dub3 transcription. Using a reporter assay, we show that NCoA1 potentiates Esrrb-mediated transcription of Dub3 and we present evidence of protein interaction between the SRC1 splice variant NCoA1 and Esrrb. Finally, we show a differential developmental regulation of all members of the p160 family during neural conversion of mESCs. These findings suggest that in mouse ESCs, changes in the relative concentration of a coactivator at a given cell cycle phase, may contribute to modulation of the transcriptional activity of the core transcription factors of the pluripotent network and be implicated in cell fate decisions upon onset of differentiation.


Assuntos
Ciclo Celular , Células-Tronco Embrionárias/metabolismo , Endopeptidases/metabolismo , Proteínas de Homeodomínio/metabolismo , Quinases Associadas a rho/metabolismo , Animais , Linhagem Celular , Dano ao DNA , Células-Tronco Embrionárias/citologia , Endopeptidases/genética , Endopeptidases/fisiologia , Regulação da Expressão Gênica/fisiologia , Haplorrinos , Camundongos , Proteína Homeobox Nanog , Regiões Promotoras Genéticas , Reação em Cadeia da Polimerase em Tempo Real , Transcrição Gênica/fisiologia
11.
Nucleic Acids Res ; 42(6): 3692-706, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24423875

RESUMO

Proliferating cell nuclear antigen (PCNA) is a well-known scaffold for many DNA replication and repair proteins, but how the switch between partners is regulated is currently unclear. Interaction with PCNA occurs via a domain known as a PCNA-Interacting Protein motif (PIP box). More recently, an additional specialized PIP box has been described, the « PIP degron ¼, that targets PCNA-interacting proteins for proteasomal degradation via the E3 ubiquitin ligase CRL4(Cdt2). Here we provide evidence that CRL4(Cdt2)-dependent degradation of PIP degron proteins plays a role in the switch of PCNA partners during the DNA damage response by facilitating accumulation of translesion synthesis DNA polymerases into nuclear foci. We show that expression of a nondegradable PIP degron (Cdt1) impairs both Pol η and Pol κ focus formation on ultraviolet irradiation and reduces cell viability, while canonical PIP box-containing proteins have no effect. Furthermore, we identify PIP degron-containing peptides from several substrates of CRL4(Cdt2) as efficient inhibitors of Pol η foci formation. By site-directed mutagenesis we show that inhibition depends on a conserved threonine residue that confers high affinity for PCNA-binding. Altogether these findings reveal an important regulative role for the CRL4(Cdt2) pathway in the switch of PCNA partners on DNA damage.


Assuntos
Dano ao DNA , Reparo do DNA , DNA Polimerase Dirigida por DNA/metabolismo , Proteínas Nucleares/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Animais , Proteínas de Ciclo Celular/química , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Morte Celular , Linhagem Celular , Inibidor de Quinase Dependente de Ciclina p21/química , Histona-Lisina N-Metiltransferase/química , Humanos , Camundongos , Células NIH 3T3 , Domínios e Motivos de Interação entre Proteínas , Proteólise , Raios Ultravioleta
12.
Mol Cell ; 52(3): 366-79, 2013 Nov 07.
Artigo em Inglês | MEDLINE | ID: mdl-24207026

RESUMO

The molecular mechanism underlying G1/S checkpoint bypass in mouse embryonic stem cells (ESCs) remains unknown. DNA damage blocks S phase entry by inhibiting the CDK2 kinase through destruction of its activator, the Cdc25A phosphatase. We observed high Cdc25A levels in G1 that persist even after DNA damage in mouse ESCs. We also found higher expression of Dub3, a deubiquitylase that controls Cdc25A protein abundance. Moreover, we demonstrate that the Dub3 gene is a direct target of Esrrb, a key transcription factor of the self-renewal machinery. We show that Dub3 expression is strongly downregulated during neural conversion and precedes Cdc25A destabilization, while forced Dub3 expression in ESCs becomes lethal upon differentiation, concomitant to cell-cycle remodeling and lineage commitment. Finally, knockdown of either Dub3 or Cdc25A induced spontaneous differentiation of ESCs. Altogether, these findings couple the self-renewal machinery to cell-cycle control through a deubiquitylase in ESCs.


Assuntos
Diferenciação Celular/genética , Células-Tronco Embrionárias/metabolismo , Endopeptidases/genética , Células-Tronco Pluripotentes/metabolismo , Proteases Específicas de Ubiquitina/metabolismo , Animais , Quinase 2 Dependente de Ciclina/metabolismo , Dano ao DNA/genética , Células-Tronco Embrionárias/citologia , Pontos de Checagem da Fase G1 do Ciclo Celular/genética , Regulação da Expressão Gênica no Desenvolvimento , Camundongos , Células-Tronco Pluripotentes/citologia , Receptores de Estrogênio/metabolismo , Proteases Específicas de Ubiquitina/genética , Fosfatases cdc25/metabolismo
13.
EMBO J ; 32(15): 2172-85, 2013 Jul 31.
Artigo em Inglês | MEDLINE | ID: mdl-23799366

RESUMO

Formation of primed single-stranded DNA at stalled replication forks triggers activation of the replication checkpoint signalling cascade resulting in the ATR-mediated phosphorylation of the Chk1 protein kinase, thus preventing genomic instability. By using siRNA-mediated depletion in human cells and immunodepletion and reconstitution experiments in Xenopus egg extracts, we report that the Y-family translesion (TLS) DNA polymerase kappa (Pol κ) contributes to the replication checkpoint response and is required for recovery after replication stress. We found that Pol κ is implicated in the synthesis of short DNA intermediates at stalled forks, facilitating the recruitment of the 9-1-1 checkpoint clamp. Furthermore, we show that Pol κ interacts with the Rad9 subunit of the 9-1-1 complex. Finally, we show that this novel checkpoint function of Pol κ is required for the maintenance of genomic stability and cell proliferation in unstressed human cells.


Assuntos
Replicação do DNA/fisiologia , DNA Polimerase Dirigida por DNA/metabolismo , Instabilidade Genômica/fisiologia , Proteínas Quinases/metabolismo , Proteínas de Xenopus/metabolismo , Animais , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Proliferação de Células , Quinase 1 do Ponto de Checagem , DNA Polimerase Dirigida por DNA/genética , Células HeLa , Humanos , Proteínas Quinases/genética , Proteínas de Xenopus/genética , Xenopus laevis
14.
Nucleic Acids Res ; 40(8): 3431-42, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22187152

RESUMO

Uncoupling between DNA polymerases and helicase activities at replication forks, induced by diverse DNA lesions or replication inhibitors, generate long stretches of primed single-stranded DNA that is implicated in activation of the S-phase checkpoint. It is currently unclear whether nucleation of the essential replication factor RPA onto this substrate stimulates the ATR-dependent checkpoint response independently of its role in DNA synthesis. Using Xenopus egg extracts to investigate the role of RPA recruitment at uncoupled forks in checkpoint activation we have surprisingly found that in conditions in which DNA synthesis occurs, RPA accumulation at forks stalled by either replication stress or UV irradiation is dispensable for Chk1 phosphorylation. In contrast, when both replication fork uncoupling and RPA hyperloading are suppressed, Chk1 phosphorylation is inhibited. Moreover, we show that extracts containing reduced levels of RPA accumulate ssDNA and induce spontaneous, caffeine-sensitive, Chk1 phosphorylation in S-phase. These results strongly suggest that disturbance of enzymatic activities of replication forks, rather than RPA hyperloading at stalled forks, is a critical determinant of ATR activation.


Assuntos
Proteína de Replicação A/fisiologia , Pontos de Checagem da Fase S do Ciclo Celular , Animais , Cafeína/farmacologia , Extratos Celulares , Quinase 1 do Ponto de Checagem , Replicação do DNA , Óvulo/enzimologia , Óvulo/metabolismo , Fosforilação/efeitos dos fármacos , Proteínas Quinases/metabolismo , Proteína de Replicação A/metabolismo , Estresse Fisiológico/genética , Xenopus , Proteínas de Xenopus
15.
Endocrinology ; 150(10): 4615-24, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19589863

RESUMO

Stress and corticosterone affect, via glucocorticoid receptors, cellular physiology in the rodent brain. A well-documented example concerns corticosteroid effects on high-voltage activated (L type) calcium currents in the hippocampal CA1 area. We tested whether corticosterone also affects calcium currents in another hippocampal area that highly expresses glucocorticoid receptors, i.e. the dentate gyrus (DG). Remarkably, corticosterone (100 nm, given for 20 min, 1-4.5 hr before recording) did not change high-voltage activated calcium currents in the DG, whereas currents in the CA1 area of the same rats were increased. Follow-up studies revealed that no apparent dissociation between the two areas was observed with respect to transcriptional regulation of calcium channel subunits; thus, in both areas corticosterone increased mRNA levels of the calcium channel-beta4 but not the (alpha) Ca(v)1.2 subunit. At the protein level, however, beta4 and Ca(v)1.2 levels were significantly up-regulated by corticosterone in the CA1 but not the DG area. These data suggest that stress-induced elevations in the level of corticosterone result in a regionally differentiated physiological response that is not simply determined by the glucocorticoid receptor distribution and that the observed regional differentiation may be caused by a gene involved in the translational machinery or in mechanisms regulating mRNA or protein stability.


Assuntos
Canais de Cálcio Tipo L/metabolismo , Corticosterona/farmacologia , Giro Denteado/metabolismo , Animais , Western Blotting , Eletrofisiologia , Hibridização In Situ , Técnicas In Vitro , Masculino , Ratos , Ratos Wistar
16.
Proc Natl Acad Sci U S A ; 106(19): 8038-42, 2009 May 12.
Artigo em Inglês | MEDLINE | ID: mdl-19416907

RESUMO

Adaptation to stress in vertebrates occurs via activation of hormonal and neuronal signaling cascades in which corticotropin-releasing hormone (CRH) plays a central role. Expression of brain CRH is subject to strong, brain-region specific regulation by glucocorticoid hormones and neurogenic intracellular signals. We hypothesized that Steroid Receptor Coactivator 1 (SRC-1), a transcriptional coregulator of the glucocorticoid receptor, is involved in the sensitivity of CRH regulation by stress-related factors. In the brains of SRC-1 knockout mice we found basal CRH mRNA levels to be lower in the central nucleus of the amygdala. Hypothalamic CRH up-regulation after chronic (but not acute) stress, as well as region-dependent up- and down-regulation induced by synthetic glucocorticoids, were significantly attenuated compared with wild type. The impaired induction of the crh gene by neurogenic signals was corroborated in AtT-20 cells, where siRNA and overexpression experiments showed that SRC-1 is necessary for full induction of a CRH promoter reporter gene by forskolin, suggestive of involvement of transcription factor CREB. In conclusion, SRC-1 is involved in positive and negative regulation of the crh gene, and an important factor for the adaptive capacity of stress.


Assuntos
Hormônio Liberador da Corticotropina/metabolismo , Regulação da Expressão Gênica , Glucocorticoides/metabolismo , Histona Acetiltransferases/fisiologia , Estresse Psicológico , Fatores de Transcrição/fisiologia , Tonsila do Cerebelo/metabolismo , Animais , Colforsina/farmacologia , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Genótipo , Hipotálamo/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Coativador 1 de Receptor Nuclear , Regiões Promotoras Genéticas
17.
PLoS One ; 4(1): e4327, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19177170

RESUMO

Glucocorticoid negative feedback of the hypothalamus-pituitary-adrenal axis is mediated in part by direct repression of gene transcription in glucocorticoid receptor (GR) expressing cells. We have investigated the cross talk between the two main signaling pathways involved in activation and repression of corticotrophin releasing hormone (CRH) mRNA expression: cyclic AMP (cAMP) and GR. We report that in the At-T20 cell-line the glucocorticoid-mediated repression of the cAMP-induced human CRH proximal promoter activity depends on the relative timing of activation of both signaling pathways. Activation of the GR prior to or in conjunction with cAMP signaling results in an effective repression of the cAMP-induced transcription of the CRH gene. In contrast, activation of the GR 10 minutes after onset of cAMP treatment, results in a significant loss of GR-mediated repression. In addition, translocation of ligand-activated GR to the nucleus was found as early as 10 minutes after glucocorticoid treatment. Interestingly, while both signaling cascades counteract each other on the CRH proximal promoter, they synergize on a synthetic promoter containing 'positive' response elements. Since the order of activation of both signaling pathways may vary considerably in vivo, we conclude that a critical time-window exists for effective repression of the CRH gene by glucocorticoids.


Assuntos
Hormônio Liberador da Corticotropina/genética , AMP Cíclico/farmacologia , Regulação para Baixo/efeitos dos fármacos , Receptores de Glucocorticoides/metabolismo , Regulação para Cima/efeitos dos fármacos , Colforsina/farmacologia , Hormônio Liberador da Corticotropina/metabolismo , Dexametasona/farmacologia , Humanos , Luciferases/metabolismo , Regiões Promotoras Genéticas/genética , Transporte Proteico/efeitos dos fármacos , Fatores de Tempo
18.
J Neurochem ; 106(6): 2515-23, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18643788

RESUMO

While the actions of glucocorticoids on brain functions have been comprehensively studied, the underlying genomic mechanisms are poorly understood. In this study, we show that glucocorticoid-induced leucine zipper (GILZ) mRNA is strongly and ubiquitously induced in rat brain. To decipher the molecular mechanisms underlying these genomic effects, it is of interest to identify the regulatory sites in the promoter region. Alignment of the rat GILZ promoter with the well-characterized human promoter resulted in poor sequence homology. Consequently, we analyzed the rat 5' flanking sequence by Matrix REDUCE and identified two high-affinity glucocorticoid response elements (GRE) located 2 kb upstream of the transcription start site. These findings were corroborated using the glucocorticoid receptor (GR) expressing Ns-1 PC12 rat cell-line. In these cells, dexamethasone treatment leads to a progressive increase of GILZ mRNA expression levels via a GR-dependent mechanism. Subsequently, using chromatin immunoprecipitation assays we show that the two high-affinity GREs are located within the GR-binding regions. Lastly, we demonstrate using multiple tissue in situ hybridization a marked increase in mRNA expression levels in spleen, thymus, heart, lung, liver, muscle, testis, kidney, colon, ileum, as well as in brain and conclude that the GILZ gene can be used to study glucocorticoid effects in many additional rodent tissues.


Assuntos
Encéfalo/metabolismo , Regulação da Expressão Gênica/genética , Glucocorticoides/metabolismo , Regiões Promotoras Genéticas/genética , Receptores de Glucocorticoides/metabolismo , Fatores de Transcrição/genética , Região 5'-Flanqueadora/genética , Animais , Sítios de Ligação/efeitos dos fármacos , Sítios de Ligação/genética , Imunoprecipitação da Cromatina/métodos , Dexametasona/metabolismo , Dexametasona/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Glucocorticoides/farmacologia , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Biologia Molecular/métodos , Células PC12 , Regiões Promotoras Genéticas/efeitos dos fármacos , RNA Mensageiro/efeitos dos fármacos , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Receptores de Glucocorticoides/efeitos dos fármacos , Receptores de Glucocorticoides/genética , Elementos de Resposta/genética , Fatores de Transcrição/metabolismo , Vísceras/metabolismo
19.
Eur J Pharmacol ; 585(2-3): 483-91, 2008 May 13.
Artigo em Inglês | MEDLINE | ID: mdl-18410913

RESUMO

Glucocorticoid hormones are important regulators of homeostasis. They are used clinically as highly effective anti-inflammatory compounds and have been prescribed for more than fifty years for a variety of conditions. They mediate their genomic actions by binding to two different intracellular receptors in target cells. The pharmacology of glucocorticoids largely depends on ligand concentration and receptor expression levels in target tissue. However, their genomic actions also critically depend on coactivators and corepressors recruitment. We discuss how various non-receptor factors affect glucocorticoid potency and efficacy with respect to their genomic effects. Differential recruitment of coregulators may account for many ligand- and cell-specific effects of glucocorticoids. This is best illustrated by the recent identification of selective glucocorticoid receptor agonists that induce distinct conformational changes to the receptors resulting in altered protein-protein interactions and consequently different regulation of gene expression. We conclude that these new molecular insights will contribute to the design of safer glucocorticoids that retain full pharmacological properties with reduced side-effects.


Assuntos
Glucocorticoides/fisiologia , Receptores de Glucocorticoides/fisiologia , Animais , Glucocorticoides/genética , Glucocorticoides/farmacologia , Humanos , Ligantes , Receptores de Glucocorticoides/efeitos dos fármacos , Receptores de Glucocorticoides/genética , Transdução de Sinais/efeitos dos fármacos
20.
Neuropharmacology ; 48(1): 80-92, 2005 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-15617730

RESUMO

The aim of the present study was to investigate if different levels of circulating corticosterone (CORT) modulate the effect of nicotine on prepulse inhibition (PPI), a measure of sensorimotor gating that is disrupted in schizophrenia and other mental illnesses. Four groups of mice were investigated: sham-operated, adrenalectomized (ADX) and implanted with a cholesterol pellet, ADX and implanted with a 10 mg CORT pellet, or ADX and 50 mg of CORT. Different CORT levels or doses of nicotine did not significantly affect startle responses. Baseline PPI was significantly reduced in mice implanted with the highest dose of CORT. In ADX mice implanted with cholesterol, nicotine treatment influenced PPI depending on the prepulse intensity. In ADX mice implanted with 50 mg of CORT, treatment with 10 mg/kg of nicotine caused a significant increase in PPI at all prepulse intensities. Binding studies showed that corticosterone treatment had significantly affected nicotinic acetylcholine receptor (nAChR) density in the mouse brain. Treatment with 50 mg CORT decreased 125I-epibatidine binding in the globus pallidus and 125I-alpha-bungarotoxin binding in the claustrum. These results suggest a possible interaction of corticosterone and nicotine at the level of the alpha4- and alpha7-type nAChR in the regulation of PPI. In situations of high circulating levels of corticosterone, nicotine may be beneficial to restore disruption of PPI.


Assuntos
Condicionamento Clássico/efeitos dos fármacos , Corticosterona/farmacologia , Nicotina/farmacologia , Agonistas Nicotínicos/farmacologia , Reflexo de Sobressalto/efeitos dos fármacos , Adrenalectomia/métodos , Análise de Variância , Animais , Autorradiografia/métodos , Comportamento Animal/efeitos dos fármacos , Peso Corporal/efeitos dos fármacos , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Compostos Bicíclicos Heterocíclicos com Pontes/farmacocinética , Bungarotoxinas/farmacocinética , Relação Dose-Resposta a Droga , Interações Medicamentosas , Isótopos de Iodo/farmacocinética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Antagonistas Nicotínicos/farmacocinética , Tamanho do Órgão/efeitos dos fármacos , Piridinas/farmacocinética , Reflexo de Sobressalto/fisiologia , Baço/efeitos dos fármacos , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA