Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Exp Med ; 217(12)2020 12 07.
Artigo em Inglês | MEDLINE | ID: mdl-32845958

RESUMO

Failure of neural tube closure during embryonic development can result in anencephaly, one of the most common birth defects in humans. A family with recurrent anencephalic fetuses was investigated to understand its etiology and pathogenesis. Exome sequencing revealed a recessive germline 21-bp in-frame deletion in NUAK2 segregating with the disease. In vitro kinase assays demonstrated that the 7-amino acid truncation in NUAK2, a serine/threonine kinase, completely abrogated its catalytic activity. Patient-derived disease models including neural progenitor cells and cerebral organoids showed that loss of NUAK2 activity led to decreased Hippo signaling via cytoplasmic YAP retention. In neural tube-like structures, endogenous NUAK2 colocalized apically with the actomyosin network, which was disrupted in patient cells, causing impaired nucleokinesis and apical constriction. Our results establish NUAK2 as an indispensable kinase for brain development in humans and suggest that a NUAK2-Hippo signaling axis regulates cytoskeletal processes that govern cell shape during neural tube closure.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Anencefalia/genética , Mutação com Perda de Função/genética , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Fatores de Transcrição/metabolismo , Actinas/metabolismo , Actomiosina/metabolismo , Sequência de Aminoácidos , Sequência de Bases , Agregação Celular , Consanguinidade , Regulação para Baixo/genética , Feminino , Feto/patologia , Genes Recessivos , Via de Sinalização Hippo , Humanos , Masculino , Células-Tronco Neurais/metabolismo , Tubo Neural/patologia , Organoides/patologia , Linhagem , Domínios Proteicos , Proteínas Serina-Treonina Quinases/química , Transdução de Sinais , Transcrição Gênica , Turquia , Proteínas de Sinalização YAP
2.
J Biol Chem ; 293(44): 17208-17217, 2018 11 02.
Artigo em Inglês | MEDLINE | ID: mdl-30232152

RESUMO

Mitochondrial reactive oxygen species (ROS) production is a tightly regulated redox signal that transmits information from the organelle to the cell. Other mitochondrial signals, such as ATP, are sensed by enzymes, including the key metabolic sensor and regulator, AMP-activated protein kinase (AMPK). AMPK responds to the cellular ATP/AMP and ATP/ADP ratios by matching mitochondrial ATP production to demand. Previous reports proposed that AMPK activity also responds to ROS, by ROS acting on redox-sensitive cysteine residues (Cys-299/Cys-304) on the AMPK α subunit. This suggests an appealing model in which mitochondria fine-tune AMPK activity by both adenine nucleotide-dependent mechanisms and by redox signals. Here we assessed whether physiological levels of ROS directly alter AMPK activity. To this end we added exogenous hydrogen peroxide (H2O2) to cells and utilized the mitochondria-targeted redox cycler MitoParaquat to generate ROS within mitochondria without disrupting oxidative phosphorylation. Mitochondrial and cytosolic thiol oxidation was assessed by measuring peroxiredoxin dimerization and by redox-sensitive fluorescent proteins. Replacing the putative redox-active cysteine residues on AMPK α1 with alanines did not alter the response of AMPK to H2O2 In parallel with measurements of AMPK activity, we measured the cell ATP/ADP ratio. This allowed us to separate the effects on AMPK activity due to ROS production from those caused by changes in this ratio. We conclude that AMPK activity in response to redox changes is not due to direct action on AMPK itself, but is a secondary consequence of redox effects on other processes, such as mitochondrial ATP production.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Mitocôndrias/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Proteínas Quinases Ativadas por AMP/genética , Difosfato de Adenosina/metabolismo , Trifosfato de Adenosina/metabolismo , Animais , Linhagem Celular , Ativação Enzimática , Humanos , Peróxido de Hidrogênio/metabolismo , Camundongos , Mitocôndrias/genética , Fibras Musculares Esqueléticas/enzimologia , Fibras Musculares Esqueléticas/metabolismo , Oxirredução
3.
Nucleic Acids Res ; 45(19): 11056-11069, 2017 Nov 02.
Artigo em Inglês | MEDLINE | ID: mdl-28977491

RESUMO

Cancer genome sequencing has implicated the cytosine deaminase activity of apolipoprotein B mRNA editing enzyme catalytic polypeptide-like (APOBEC) genes as an important source of mutations in diverse cancers, with APOBEC3B (A3B) expression especially correlated with such cancer mutations. To better understand the processes directing A3B over-expression in cancer, and possible therapeutic avenues for targeting A3B, we have investigated the regulation of A3B gene expression. Here, we show that A3B expression is inversely related to p53 status in different cancer types and demonstrate that this is due to a direct and pivotal role for p53 in repressing A3B expression. This occurs through the induction of p21 (CDKN1A) and the recruitment of the repressive DREAM complex to the A3B gene promoter, such that loss of p53 through mutation, or human papilloma virus-mediated inhibition, prevents recruitment of the complex, thereby causing elevated A3B expression and cytosine deaminase activity in cancer cells. As p53 is frequently mutated in cancer, our findings provide a mechanism by which p53 loss can promote cancer mutagenesis.


Assuntos
Citidina Desaminase/genética , Regulação Neoplásica da Expressão Gênica , Antígenos de Histocompatibilidade Menor/genética , Proteína Supressora de Tumor p53/genética , Linhagem Celular , Inibidor de Quinase Dependente de Ciclina p21/genética , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Citidina Desaminase/metabolismo , Células HCT116 , Humanos , Immunoblotting , Antígenos de Histocompatibilidade Menor/metabolismo , Mutação , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/patologia , Interferência de RNA , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Proteína Supressora de Tumor p53/metabolismo
4.
Biochem J ; 474(10): 1741-1754, 2017 05 09.
Artigo em Inglês | MEDLINE | ID: mdl-28302767

RESUMO

AMP-activated protein kinase (AMPK) plays a key role in integrating metabolic pathways in response to energy demand. AMPK activation results in a wide range of downstream responses, many of which are associated with improved metabolic outcome, making AMPK an attractive target for the treatment of metabolic diseases. AMPK is a heterotrimeric complex consisting of a catalytic subunit (α) and two regulatory subunits (ß and γ). The γ-subunit harbours the nucleotide-binding sites and plays an important role in AMPK regulation in response to cellular energy levels. In mammals, there are three isoforms of the γ-subunit and these respond differently to regulation by nucleotides, but there is limited information regarding their role in activation by small molecules. Here, we determined the effect of different γ-isoforms on AMPK by a direct activator, 991. In cells, 991 led to a greater activation of γ2-containing AMPK complexes compared with either γ1 or γ3. This effect was dependent on the long N-terminal region of the γ2-isoform. We were able to rule out an effect of Ser108 phosphorylation, since mutation of Ser108 to alanine in the ß2-isoform had no effect on activation of AMPK by 991 in either γ1- or γ2-complexes. The rate of dephosphorylation of Thr172 was slower for γ2- compared with γ1-complexes, both in the absence and presence of 991. Our studies show that activation of AMPK by 991 depends on the nature of the γ-isoform. This finding may have implications for the design of isoform-selective AMPK activators.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Proteínas Quinases Ativadas por AMP/antagonistas & inibidores , Proteínas Quinases Ativadas por AMP/química , Proteínas Quinases Ativadas por AMP/genética , Regulação Alostérica/efeitos dos fármacos , Substituição de Aminoácidos , Aminopiridinas/farmacologia , Benzimidazóis/farmacologia , Benzoatos/farmacologia , Sítios de Ligação , Sistemas CRISPR-Cas , Ativação Enzimática/efeitos dos fármacos , Ativadores de Enzimas/farmacologia , Células HEK293 , Humanos , Indóis/farmacologia , Isoenzimas/antagonistas & inibidores , Isoenzimas/química , Isoenzimas/genética , Isoenzimas/metabolismo , Ligantes , Mutação , Fosforilação/efeitos dos fármacos , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Treonina/metabolismo
5.
Oncotarget ; 7(24): 37103-37120, 2016 Jun 14.
Artigo em Inglês | MEDLINE | ID: mdl-27206796

RESUMO

The glycerophospholipid phosphatidylcholine is the most abundant phospholipid species of eukaryotic membranes and essential for structural integrity and signaling function of cell membranes required for cancer cell growth. Inhibition of choline kinase alpha (CHKA), the first committed step to phosphatidylcholine synthesis, by the selective small-molecule ICL-CCIC-0019, potently suppressed growth of a panel of 60 cancer cell lines with median GI50 of 1.12 µM and inhibited tumor xenograft growth in mice. ICL-CCIC-0019 decreased phosphocholine levels and the fraction of labeled choline in lipids, and induced G1 arrest, endoplasmic reticulum stress and apoptosis. Changes in phosphocholine cellular levels following treatment could be detected non-invasively in tumor xenografts by [18F]-fluoromethyl-[1,2-2H4]-choline positron emission tomography. Herein, we reveal a previously unappreciated effect of choline metabolism on mitochondria function. Comparative metabolomics demonstrated that phosphatidylcholine pathway inhibition leads to a metabolically stressed phenotype analogous to mitochondria toxin treatment but without reactive oxygen species activation. Drug treatment decreased mitochondria function with associated reduction of citrate synthase expression and AMPK activation. Glucose and acetate uptake were increased in an attempt to overcome the metabolic stress. This study indicates that choline pathway pharmacological inhibition critically affects the metabolic function of the cell beyond reduced synthesis of phospholipids.


Assuntos
Aminopiridinas/farmacologia , Transformação Celular Neoplásica/efeitos dos fármacos , Colina Quinase/antagonistas & inibidores , Pontos de Checagem da Fase G1 do Ciclo Celular/efeitos dos fármacos , Mitocôndrias/efeitos dos fármacos , Fosfatidilcolinas/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Compostos de Piridínio/farmacologia , Transdução de Sinais/efeitos dos fármacos , Animais , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Colina/metabolismo , Citrato (si)-Sintase/metabolismo , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Feminino , Imunofluorescência , Humanos , Metabolômica , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Mitocôndrias/metabolismo , Tomografia por Emissão de Pósitrons , Espécies Reativas de Oxigênio/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
6.
Immunol Cell Biol ; 94(7): 689-700, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-26987686

RESUMO

APOBEC3G (apolipoprotein B mRNA editing enzyme catalytic polypeptide-like 3G; A3G) is an innate defense protein showing activity against retroviruses and retrotransposons. Activated CD4(+) T cells are highly permissive for HIV-1 replication, whereas resting CD4(+) T cells are refractory. Dendritic cells (DCs), especially mature DCs, are also refractory. We investigated whether these differences could be related to a differential A3G expression and/or subcellular distribution. We found that A3G mRNA and protein expression is very low in resting CD4(+) T cells and immature DCs, but increases strongly following T-cell activation and DC maturation. The Apo-7 anti-A3G monoclonal antibody (mAb), which was specifically developed, confirmed these differences at the protein level and disclosed that A3G is mainly cytoplasmic in resting CD4(+) T cells and immature DCs. Nevertheless, A3G translocates to the nucleus in activated-proliferating CD4(+) T cells, yet remaining cytoplasmic in matured DCs, a finding confirmed by immunoblotting analysis of cytoplasmic and nuclear fractions. Apo-7 mAb was able to immunoprecipitate endogenous A3G allowing to detect complexes with numerous proteins in activated-proliferating but not in resting CD4(+) T cells. The results show for the first time the nuclear translocation of A3G in activated-proliferating CD4(+) T cells.


Assuntos
Desaminase APOBEC-3G/metabolismo , Linfócitos T CD4-Positivos/imunologia , Diferenciação Celular , Células Dendríticas/citologia , Ativação Linfocitária/imunologia , Desaminase APOBEC-3G/genética , Desaminase APOBEC-3G/imunologia , Animais , Anticorpos Monoclonais/metabolismo , Linhagem Celular , Núcleo Celular/metabolismo , Humanos , Imunoprecipitação , Camundongos Endogâmicos BALB C , Peso Molecular , Monócitos/citologia , Ligação Proteica , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Frações Subcelulares/enzimologia , Regulação para Cima/genética
7.
Cell Rep ; 13(1): 108-121, 2015 Oct 06.
Artigo em Inglês | MEDLINE | ID: mdl-26411678

RESUMO

Estrogen receptor α (ERα) is the key transcriptional driver in a large proportion of breast cancers. We report that APOBEC3B (A3B) is required for regulation of gene expression by ER and acts by causing C-to-U deamination at ER binding regions. We show that these C-to-U changes lead to the generation of DNA strand breaks through activation of base excision repair (BER) and to repair by non-homologous end-joining (NHEJ) pathways. We provide evidence that transient cytidine deamination by A3B aids chromatin modification and remodelling at the regulatory regions of ER target genes that promotes their expression. A3B expression is associated with poor patient survival in ER+ breast cancer, reinforcing the physiological significance of A3B for ER action.


Assuntos
Neoplasias da Mama/genética , Citidina Desaminase/genética , Citidina/metabolismo , Reparo do DNA por Junção de Extremidades , Receptor alfa de Estrogênio/genética , Regulação Neoplásica da Expressão Gênica , Sítios de Ligação , Neoplasias da Mama/metabolismo , Neoplasias da Mama/mortalidade , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Proliferação de Células , Citidina Desaminase/antagonistas & inibidores , Citidina Desaminase/metabolismo , DNA/genética , DNA/metabolismo , Dano ao DNA , Desaminação , Receptor alfa de Estrogênio/metabolismo , Feminino , Humanos , Antígenos de Histocompatibilidade Menor , Prognóstico , Ligação Proteica , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Transdução de Sinais , Análise de Sobrevida , Transcrição Gênica , Fator Trefoil-1 , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/metabolismo
8.
Cancer Res ; 73(16): 5253-65, 2013 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-23774215

RESUMO

Oncogene-induced senescence (OIS) constitutes a failsafe program that restricts tumor development. However, the mechanisms that link oncogenesis to senescence are not completely understood. We carried out a loss-of-function genetic screen that identified the potassium channel KCNA1 as a determinant of OIS escape that can license tumor growth. Oncogenic stress triggers an increase in KCNA1 expression and its relocation from the cytoplasm to the membrane. Mechanistically, this relocation is due to a loss of protein kinase A (PKA)-induced phosphorylation at residue S446 of KCNA1. Accordingly, sustaining PKA activity or expressing a KCNA1 phosphomimetic mutant maintained KCNA1 in the cytoplasm and caused escape from OIS. KCNA1 relocation to the membrane induced a change in membrane potential that invariably resulted in cellular senescence. Restoring KCNA1 expression in transformation-competent cells triggered variation in membrane potential and blocked RAS-induced transformation, and PKA activation suppressed both effects. Furthermore, KCNA1 expression was reduced in human cancers, and this decrease correlated with an increase in breast cancer aggressiveness. Taken together, our results identify a novel pathway that restricts oncogenesis through a potassium channel-dependent senescence pathway.


Assuntos
Transformação Celular Neoplásica/genética , Canal de Potássio Kv1.1/genética , Canal de Potássio Kv1.1/metabolismo , Animais , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Processos de Crescimento Celular/fisiologia , Linhagem Celular , Membrana Celular/genética , Membrana Celular/metabolismo , Senescência Celular/genética , Proteínas Quinases Dependentes de AMP Cíclico/genética , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Citoplasma/genética , Citoplasma/metabolismo , Regulação para Baixo , Humanos , Glândulas Mamárias Humanas/metabolismo , Glândulas Mamárias Humanas/patologia , Potenciais da Membrana/genética , Camundongos , Células NIH 3T3 , Fosforilação/genética , Transdução de Sinais/genética
9.
BMC Mol Biol ; 14: 5, 2013 Feb 07.
Artigo em Inglês | MEDLINE | ID: mdl-23391187

RESUMO

BACKGROUND: Apolipoprotein B (APOB) is an integral component of the chylomicron and the atherogenic lipoproteins LDL and Lp(a). Exon 26 of the APOB pre-mRNA is unusually long at 7,572 nt and is constitutively spliced. It is also subject to RNA editing in the intestine, which generates a shortened isoform, APOB48, assembled exclusively into chylomicrons. Due to its length, exon 26 contains multiple pseudo splice sites which are not spliced, but which conform to the degenerate splice site consensus. RESULTS: We demonstrate that these pseudo splice sites are repressed by multiple, tandem splicing silencers distributed along the length of exon 26. The distribution of these elements appears to be heterogeneous, with a greater frequency in the middle 4,800 nt of the exon. CONCLUSION: Repression of these splice sites is key to maintaining the integrity of exon 26 during RNA splicing and therefore the correct expression of both isoforms of APOB.


Assuntos
Processamento Alternativo , Apolipoproteínas B/genética , Regulação para Baixo , Elementos Silenciadores Transcricionais , Apolipoproteínas B/química , Apolipoproteínas B/metabolismo , Sequência de Bases , Éxons , Humanos , Dados de Sequência Molecular , Sítios de Splice de RNA
10.
Biochem Pharmacol ; 79(6): 853-63, 2010 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-19896469

RESUMO

The tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) is a TNF superfamily member that is being considered as a new strategy in anticancer therapy because of its ability to induce apoptosis, alone or in combination with other stimuli, in many cancer cells. AMP-activated protein kinase (AMPK) is an evolutionarily conserved key regulator of cellular energy homeostasis that protects the cell from energy depletion and stress by activating several biochemical pathways that lead to the conservation, as well as generation, of ATP. Here we report that a number of AMPK activators, including the small molecule activator A-769662, markedly sensitize TRAIL-resistant breast cancer cells to TRAIL-induced apoptosis. However, silencing AMPKalpha1 expression with siRNA or over-expression of DN-AMPKalpha1 does not inhibit AICAR, glucose deprivation, phenformin or A-769662-induced sensitization to TRAIL. Furthermore, the expression of constitutively active AMPK subunits does not sensitize resistant breast cancer cells to TRAIL-induced apoptosis. The cellular FLICE-inhibitory proteins (cFLIP(L) and cFLIP(S)) were significantly down-regulated following exposure to AMPK activators through an AMPK-independent mechanism. Furthermore, in cells over-expressing cFLIP(L), sensitization to TRAIL by AMPK activators was markedly reduced. In summary, our results indicate that AMPK activators facilitate the activation by TRAIL of an apoptotic cell death program through a mechanism independent of AMPK and dependent on the down-regulation of cFLIP levels.


Assuntos
Adenilato Quinase/metabolismo , Neoplasias da Mama/tratamento farmacológico , Proteína Reguladora de Apoptosis Semelhante a CASP8 e FADD/metabolismo , Regulação para Baixo , Ativadores de Enzimas/farmacologia , Ligante Indutor de Apoptose Relacionado a TNF/farmacologia , Proteínas Quinases Ativadas por AMP/genética , Proteínas Quinases Ativadas por AMP/metabolismo , Apoptose , Proteína Reguladora de Apoptosis Semelhante a CASP8 e FADD/genética , Linhagem Celular Tumoral , Feminino , Regulação Neoplásica da Expressão Gênica/fisiologia , Inativação Gênica , Humanos , Subunidades Proteicas
11.
EMBO J ; 29(2): 376-86, 2010 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-19927127

RESUMO

Senescence is an irreversible cell-cycle arrest that is elicited by a wide range of factors, including replicative exhaustion. Emerging evidences suggest that cellular senescence contributes to ageing and acts as a tumour suppressor mechanism. To identify novel genes regulating senescence, we performed a loss-of-function screen on normal human diploid fibroblasts. We show that downregulation of the AMPK-related protein kinase 5 (ARK5 or NUAK1) results in extension of the cellular replicative lifespan. Interestingly, the levels of NUAK1 are upregulated during senescence whereas its ectopic expression triggers a premature senescence. Cells that constitutively express NUAK1 suffer gross aneuploidies and show diminished expression of the genomic stability regulator LATS1, whereas depletion of NUAK1 with shRNA exerts opposite effects. Interestingly, a dominant-negative form of LATS1 phenocopies NUAK1 effects. Moreover, we show that NUAK1 phosphorylates LATS1 at S464 and this has a role in controlling its stability. In summary, our work highlights a novel role for NUAK1 in the control of cellular senescence and cellular ploidy.


Assuntos
Senescência Celular , Fibroblastos/citologia , Ploidias , Proteínas Quinases/metabolismo , Proteínas Repressoras/metabolismo , Quinases Proteína-Quinases Ativadas por AMP , Linhagem Celular , Fibroblastos/metabolismo , Regulação da Expressão Gênica , Humanos , Fosforilação , Proteínas Quinases/genética , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Repressoras/genética
12.
Antiviral Res ; 78(3): 260-7, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18313151

RESUMO

BACKGROUND/AIM: Interferon-gamma (IFN-gamma) produced by activated T-cells is the principle mediator of non-cytolytic Hepatitis B virus (HBV) inactivation; however the intracellular pathways responsible are poorly defined. We investigated the role of IFN-gamma-inducible nitric oxide synthase (iNOS) and APOBEC3 (A3) enzyme family in the inhibition of HBV replication by IFN-gamma. METHODS: Hepatoma-cell lines transfected with HBV DNA were treated with IFN-gamma. Viral replication, iNOS and A3 mRNAs were quantitated by TaqManPCR and the direct nitric oxide (NO) effect on HBV replication was investigated using an NO-donor. A3G antiviral activity was verified by co-transfection with its inhibitor, human immunodeficiency virus (HIV)-associated virion infectivity factor (Vif). RESULTS: IFN-gamma caused a dose-dependent reduction (>50%) of HBV DNA in the absence of cytotoxicity. Although iNOS mRNA increased 45-fold in IFN-gamma treated cells, NO2- was not detectable in supernatants and the use of an NO-donor did not inhibit HBV replication. A3 enzyme mRNAs varied between cells and were >10-fold higher in lymphocytes than in liver tissue. IFN-gamma up-regulated A3G mRNA by three-fold, associated with significant HBV DNA decrease. However, A3G degradation by Vif did not abolish the antiviral effect of IFN-gamma against HBV. CONCLUSIONS: IFN-gamma inhibits HBV replication and up-regulates both iNOS and A3G. However, other pathways appear to have a greater role in IFN-gamma-induced HBV inactivation in the liver.


Assuntos
Antivirais/farmacologia , Citosina Desaminase/metabolismo , Vírus da Hepatite B/efeitos dos fármacos , Interferon gama/farmacologia , Óxido Nítrico Sintase Tipo II/metabolismo , Desaminases APOBEC , Linhagem Celular Tumoral , Citidina Desaminase , Citosina Desaminase/genética , DNA Viral/metabolismo , Vírus da Hepatite B/genética , Vírus da Hepatite B/metabolismo , Humanos , Fígado/virologia , Óxido Nítrico Sintase Tipo II/genética , Proteínas Recombinantes , Replicação Viral/efeitos dos fármacos
13.
PLoS Biol ; 5(3): e67, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17341133

RESUMO

Regulation of transforming growth factor-beta (TGF-beta) signaling is critical in vertebrate development, as several members of the TGF-beta family have been shown to act as morphogens, controlling a variety of cell fate decisions depending on concentration. Little is known about the role of intracellular regulation of the TGF-beta pathway in development. E3 ubiquitin ligases target specific protein substrates for proteasome-mediated degradation, and several are implicated in signaling. We have shown that Arkadia, a nuclear RING-domain E3 ubiquitin ligase, is essential for a subset of Nodal functions in the embryo, but the molecular mechanism of its action in embryonic cells had not been addressed. Here, we find that Arkadia facilitates Nodal signaling broadly in the embryo, and that it is indispensable for cell fates that depend on maximum signaling. Loss of Arkadia in embryonic cells causes nuclear accumulation of phospho-Smad2/3 (P-Smad2/3), the effectors of Nodal signaling; however, these must be repressed or hypoactive as the expression of their direct target genes is reduced or lost. Molecular and functional analysis shows that Arkadia interacts with and ubiquitinates P-Smad2/3 causing their degradation, and that this is via the same domains required for enhancing their activity. Consistent with this dual function, introduction of Arkadia in homozygous null (-/-) embryonic stem cells activates the accumulated and hypoactive P-Smad2/3 at the expense of their abundance. Arkadia-/- cells, like Smad2-/- cells, cannot form foregut and prechordal plate in chimeras, confirming this functional interaction in vivo. As Arkadia overexpression never represses, and in some cells enhances signaling, the degradation of P-Smad2/3 by Arkadia cannot occur prior to their activation in the nucleus. Therefore, Arkadia provides a mechanism for signaling termination at the end of the cascade by coupling degradation of P-Smad2/3 with the activation of target gene transcription. This mechanism can account for achieving efficient and maximum Nodal signaling during embryogenesis and for rapid resetting of target gene promoters allowing cells to respond to dynamic changes in extracellular signals.


Assuntos
Transdução de Sinais , Proteína Smad2/metabolismo , Proteína Smad3/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Ubiquitina/fisiologia , Animais , Sequência de Bases , Quimera , Primers do DNA , Camundongos , Ubiquitina-Proteína Ligases
14.
Int J Hematol ; 83(3): 195-200, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16720547

RESUMO

Enzymes that deaminate cytidine to uridine play an important role in a variety of pathways from bacteria to man. Ancestral members of this family were able to deaminate cytidine only in a mononucleotide or nucleoside context. Recently, a family of enzymes has been discovered with the ability to deaminate cytidines on RNA or DNA. The first member of this new family is APOBEC1, which deaminates apolipoprotein B messenger RNA to generate a premature stop codon. APOBEC1 has the conserved active site motif found in Escherichia coli cytidine deaminase. In addition, APOBEC1 has a unique motif containing 2 phenylalanine residues and an insert of 4 amino acid residues across the active site motif. This motif is present in APOBEC family members including activation-induced cytidine deaminase (AID), APOBEC2, and APOBEC3A through APOBEC3G. AID is essential for initiating class-switch recombination, somatic hypermutation, and gene conversion. The APOBEC3 family is unique to primates. APOBEC3G is able to protect cells from human immunodeficiency virus and other viral infections. This function is not unique to APOBEC3G; other APOBEC3 family members also have this ability. Overexpression of enzymes in this family can cause cancer, suggesting that the genes for the APOBEC family of proteins are proto-oncogenes. Recent advances in the understanding of the mechanism of action of this family are summarized in this review.


Assuntos
Citidina Desaminase/metabolismo , Citosina Desaminase/metabolismo , Regulação Enzimológica da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Neoplasias/enzimologia , Proteínas Oncogênicas/metabolismo , Viroses/enzimologia , Desaminases APOBEC , Desaminase APOBEC-1 , Animais , Sítios de Ligação , Citidina Desaminase/genética , Citosina Desaminase/genética , Ativação Enzimática , Escherichia coli/enzimologia , Escherichia coli/genética , Humanos , Neoplasias/genética , Proteínas Oncogênicas/genética , Edição de RNA/genética , Viroses/genética
15.
RNA ; 11(2): 173-86, 2005 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-15659357

RESUMO

We have solved the NMR structure of the 31-nucleotide (nt) apoB mRNA stem-loop, a substrate of the cytidine deaminase APOBEC1. We found that the edited base located at the 5' end of the octa-loop is stacked between two adenosines in both the unedited (cytidine 6666) and the edited (uridine 6666) forms and that the rest of the loop is unstructured. The 11-nt "mooring" sequence essential for editing is partially flexible although it is mostly in the stem of the RNA. The octa-loop and the internal loop in the middle of the stem confer this flexibility. These findings shed light on why APOBEC1 alone cannot edit efficiently the cytidine 6666 under physiological conditions, the editing base being buried in the loop and not directly accessible. We also show that APOBEC1 does not specifically bind apoB mRNA and requires the auxiliary factor, APOBEC1 complementary factor (ACF), to edit specifically cytidine 6666. The binding of ACF to both the mooring sequence and APOBEC1 explains the specificity of the reaction. Our NMR study lead us to propose a mechanism in which ACF recognizes first the flexible nucleotides of the mooring sequence (the internal loop and the 3' end octa-loop) and subsequently melts the stem-loop, exposing the amino group of the cytidine 6666 to APOBEC1. Thus, the flexibility of the mooring sequence plays a central role in the RNA recognition by ACF.


Assuntos
Apolipoproteínas B/genética , Citidina Desaminase/metabolismo , RNA Mensageiro/química , RNA Mensageiro/metabolismo , Proteínas de Ligação a RNA/metabolismo , Desaminase APOBEC-1 , Sequência de Bases , Humanos , Técnicas In Vitro , Modelos Moleculares , Ressonância Magnética Nuclear Biomolecular , Conformação de Ácido Nucleico , Edição de RNA , RNA Mensageiro/genética , Termodinâmica
16.
RNA ; 10(9): 1399-411, 2004 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-15273326

RESUMO

Expression and purification to homogeneity of the apolipoprotein B mRNA editing subunit, APOBEC1, has allowed the demonstration that this apoenzyme has considerable residual enzymatic activity on a minimal apoB mRNA substrate, even in the absence of any auxiliary factors. Assay of this activity as a function of various experimental conditions has led to substantial optimization of assay conditions through the use of incomplete factorial and response surface experiments. Surprisingly, the apoenzyme is thermostable, and has a temperature optimum near 45 degrees C. We have used these optimized conditions, to assess steady-state kinetic parameters for APOBEC1 mRNA editing activity with and without the auxiliary factor, ACF. An important effect of the auxiliary factor is to broaden the temperature range of APOBEC1 activity, lowering the optimal temperature and enabling it to function optimally at lower temperatures. A model consistent with this observation is that at lower temperatures ACF promotes a conformational transition in the RNA substrate that occurs spontaneously at higher temperature. Notably, the substantial RNA editing activity of APOBEC1 alone may be responsible for the "hyperediting" observed upon overexpression of APOBEC1 in transgenic mice.


Assuntos
Apolipoproteínas B/genética , Coenzimas/metabolismo , Citidina Desaminase/metabolismo , Edição de RNA , RNA Mensageiro/metabolismo , Proteínas de Ligação a RNA/fisiologia , Desaminase APOBEC-1 , Animais , Baculoviridae/genética , Pareamento de Bases , Sequência de Bases , Citidina Desaminase/genética , Glutationa Transferase/metabolismo , Dados de Sequência Molecular , Conformação de Ácido Nucleico , RNA Mensageiro/genética , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/isolamento & purificação , Proteínas Recombinantes de Fusão/metabolismo , Temperatura
17.
Nucleic Acids Res ; 32(8): 2421-9, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-15121899

RESUMO

In the absence of the viral vif gene, human immunodeficiency virus (HIV) may be restricted by the APOBEC3G gene on chromosome 22. The role of the HIV Vif protein is to exclude host cell APOBEC3G from the budding virion. As APOBEC3G shows sequence homology to cytidine deaminases, it is presumed that in the absence of Vif, cytidine residues in the cDNA are deaminated yielding uracil. It is not known if additional proteins mediate APOBEC3G function or if deamination occurs in concert with reverse transcription. This report describes an in vitro assay showing that Baculovirus derived APOBEC3G alone extensively deaminates cDNA independently of reverse transcriptase. It reproduces the dinucleotide context typical of G --> A hypermutants derived from a Delta(vif) virus. By using an RNaseH- form of reverse transcriptase, it was shown that the cDNA has to be free of its RNA template to allow deamination. APOBEC3G deamination of dC or dCTP was not detected. In short, APOBEC3G is a single-stranded DNA cytidine deaminase capable of restricting retroviral replication.


Assuntos
Citidina Desaminase/metabolismo , DNA de Cadeia Simples/metabolismo , Transcriptase Reversa do HIV/metabolismo , Proteínas/fisiologia , Desaminase APOBEC-3G , Sequência de Bases , Fosfatos de Dinucleosídeos/metabolismo , Nucleosídeo Desaminases , Proteínas/metabolismo , Proteínas Repressoras , Especificidade por Substrato , Transcrição Gênica
18.
EMBO J ; 22(15): 3971-82, 2003 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-12881431

RESUMO

The C to U editing of apolipoprotein B (apoB) mRNA is mediated by a minimal complex composed of an RNA-binding cytidine deaminase (APOBEC1) and a complementing specificity factor (ACF). This editing generates a premature termination codon and a truncated open reading frame. We demonstrate that the APOBEC1-ACF holoenzyme mediates a multifunctional cycle. The atypical APOBEC1 nuclear localization signal is involved in RNA binding and is used to import ACF into the nucleus as cargo. APOBEC1 alone induces nonsense-mediated decay (NMD). The APOBEC1-ACF complex edits and remains associated with the edited RNA to protect it from NMD. The APOBEC1 nuclear export signal is involved in the export of ACF and the edited apoB mRNA together, to the site of translation.


Assuntos
Citidina Desaminase/fisiologia , RNA Mensageiro/metabolismo , Desaminase APOBEC-1 , Animais , Transporte Biológico , Linhagem Celular , Núcleo Celular/metabolismo , Citidina Desaminase/metabolismo , Citoplasma/metabolismo , Humanos , Sinais de Localização Nuclear , Edição de RNA , RNA Mensageiro/genética , Técnicas do Sistema de Duplo-Híbrido
19.
Genomics ; 79(3): 285-96, 2002 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-11863358

RESUMO

The cytidine (C) to uridine (U) editing of apolipoprotein (apo) B mRNA is mediated by tissue-specific, RNA-binding cytidine deaminase APOBEC1. APOBEC1 is structurally homologous to Escherichia coli cytidine deaminase (ECCDA), but has evolved specific features required for RNA substrate binding and editing. A signature sequence for APOBEC1 has been used to identify other members of this family. One of these genes, designated APOBEC2, is found on chromosome 6. Another gene corresponds to the activation-induced deaminase (AID) gene, which is located adjacent to APOBEC1 on chromosome 12. Seven additional genes, or pseudogenes (designated APOBEC3A to 3G), are arrayed in tandem on chromosome 22. Not present in rodents, this locus is apparently an anthropoid-specific expansion of the APOBEC family. The conclusion that these new genes encode orphan C to U RNA-editing enzymes of the APOBEC family comes from similarity in amino acid sequence with APOBEC1, conserved intron/exon organization, tissue-specific expression, homodimerization, and zinc and RNA binding similar to APOBEC1. Tissue-specific expression of these genes in a variety of cell lines, along with other evidence, suggests a role for these enzymes in growth or cell cycle control.


Assuntos
Cromossomos Humanos Par 22/genética , Citidina Desaminase/genética , Genoma Humano , Família Multigênica/genética , Edição de RNA/genética , Proteínas de Ligação a RNA/genética , Desaminase APOBEC-1 , Sequência de Aminoácidos/genética , Animais , Northern Blotting , Domínio Catalítico/genética , Éxons/genética , Etiquetas de Sequências Expressas , Haplorrinos/genética , Humanos , Íntrons/genética , Dados de Sequência Molecular , Filogenia , Alinhamento de Sequência , Análise de Sequência de DNA , Homologia de Sequência
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA