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











Base de dados
Intervalo de ano de publicação
1.
Cancers (Basel) ; 13(13)2021 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-34282773

RESUMO

Histone H2AX phosphorylated at serine 139 (γ-H2AX) is a hallmark of DNA damage, signaling the presence of DNA double-strand breaks and global replication stress in mammalian cells. While γ-H2AX can be visualized with antibodies in fixed cells, its detection in living cells was so far not possible. Here, we used immune libraries and phage display to isolate nanobodies that specifically bind to γ-H2AX. We solved the crystal structure of the most soluble nanobody in complex with the phosphopeptide corresponding to the C-terminus of γ-H2AX and show the atomic constituents behind its specificity. We engineered a bivalent version of this nanobody and show that bivalency is essential to quantitatively visualize γ-H2AX in fixed drug-treated cells. After labelling with a chemical fluorophore, we were able to detect γ-H2AX in a single-step assay with the same sensitivity as with validated antibodies. Moreover, we produced fluorescent nanobody-dTomato fusion proteins and applied a transduction strategy to visualize with precision γ-H2AX foci present in intact living cells following drug treatment. Together, this novel tool allows performing fast screenings of genotoxic drugs and enables to study the dynamics of this particular chromatin modification in individual cancer cells under a variety of conditions.

2.
Neuron ; 109(11): 1825-1835.e5, 2021 06 02.
Artigo em Inglês | MEDLINE | ID: mdl-33887199

RESUMO

Neuronal intranuclear inclusion disease (NIID) is a neurodegenerative disease characterized by the presence of intranuclear inclusions of unknown origin. NIID is caused by an expansion of GGC repeats in the 5' UTR of the NOTCH2NLC (N2C) gene. We found that these repeats are embedded in a small upstream open reading frame (uORF) (uN2C), resulting in their translation into a polyglycine-containing protein, uN2CpolyG. This protein accumulates in intranuclear inclusions in cell and mouse models and in tissue samples of individuals with NIID. Furthermore, expression of uN2CpolyG in mice leads to locomotor alterations, neuronal cell loss, and premature death of the animals. These results suggest that translation of expanded GGC repeats into a novel and pathogenic polyglycine-containing protein underlies the presence of intranuclear inclusions and neurodegeneration in NIID.


Assuntos
Doenças Neurodegenerativas/genética , Peptídeos/toxicidade , Expansão das Repetições de Trinucleotídeos , Animais , Morte Celular , Núcleo Celular/metabolismo , Núcleo Celular/patologia , Células Cultivadas , Células HEK293 , Humanos , Corpos de Inclusão Intranuclear/genética , Corpos de Inclusão Intranuclear/metabolismo , Corpos de Inclusão Intranuclear/patologia , Locomoção , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Doenças Neurodegenerativas/metabolismo , Doenças Neurodegenerativas/patologia , Fases de Leitura Aberta , Peptídeos/genética , Peptídeos/metabolismo
3.
Cancers (Basel) ; 11(3)2019 Mar 13.
Artigo em Inglês | MEDLINE | ID: mdl-30871194

RESUMO

Phosphorylated histone H2AX (γ-H2AX), a central player in the DNA damage response (DDR), serves as a biomarker of DNA double-strand break repair. Although DNA damage is generally visualized by the formation of γ-H2AX foci in injured nuclei, it is unclear whether the widespread uniform nuclear γ-H2AX (called pan-nuclear) pattern occurring upon intense replication stress (RS) is linked to DDR. Using a novel monoclonal antibody that binds exclusively to the phosphorylated C-terminus of H2AX, we demonstrate that H2AX phosphorylation is systematically pan-nuclear in cancer cells stressed with RS-inducing drugs just before they die. The pan-nuclear γ-H2AX pattern is abolished by inhibition of the DNA-PK kinase. Cell death induction of cancer cells treated with increasing combinations of replication and kinase (ATR and Chk1) inhibitory drugs was proportional to the appearance of pan-nuclear γ-H2AX pattern. Delivery of labeled anti-γ-H2AX Fabs in stressed cells demonstrated at a single cell level that pan-nuclear γ-H2AX formation precedes irreversible cell death. Moreover, we show that H2AX is not required for RS-induced cell death in HeLa cells. Thus, the nuclear-wide formation of γ-H2AX is an incident of RS-induced cell death and, thus, the pan nuclear H2AX pattern should be regarded as an indicator of lethal RS-inducing drug efficacy.

4.
J Cell Biol ; 217(4): 1537-1552, 2018 04 02.
Artigo em Inglês | MEDLINE | ID: mdl-29440513

RESUMO

Fluorescent labeling of endogenous proteins for live-cell imaging without exogenous expression of tagged proteins or genetic manipulations has not been routinely possible. We describe a simple versatile antibody-based imaging approach (VANIMA) for the precise localization and tracking of endogenous nuclear factors. Our protocol can be implemented in every laboratory allowing the efficient and nonharmful delivery of organic dye-conjugated antibodies, or antibody fragments, into different metazoan cell types. Live-cell imaging permits following the labeled probes bound to their endogenous targets. By using conventional and super-resolution imaging we show dynamic changes in the distribution of several nuclear transcription factors (i.e., RNA polymerase II or TAF10), and specific phosphorylated histones (γH2AX), upon distinct biological stimuli at the nanometer scale. Hence, considering the large panel of available antibodies and the simplicity of their implementation, VANIMA can be used to uncover novel biological information based on the dynamic behavior of transcription factors or posttranslational modifications in the nucleus of single live cells.


Assuntos
Núcleo Celular/metabolismo , Técnica Direta de Fluorescência para Anticorpo , Histonas/metabolismo , Microscopia Confocal , Análise de Célula Única/métodos , Fatores de Transcrição/metabolismo , Animais , Apoptose , Neoplasias Ósseas/genética , Neoplasias Ósseas/metabolismo , Neoplasias Ósseas/patologia , Linhagem Celular Tumoral , Núcleo Celular/patologia , Proliferação de Células , Fibroblastos/metabolismo , Humanos , Cinética , Camundongos , Células-Tronco Embrionárias Murinas/metabolismo , Osteossarcoma/genética , Osteossarcoma/metabolismo , Osteossarcoma/patologia , Fosforilação , RNA Polimerase II/genética , RNA Polimerase II/metabolismo , Fatores Associados à Proteína de Ligação a TATA/genética , Fatores Associados à Proteína de Ligação a TATA/metabolismo , Proteína de Ligação a TATA-Box/genética , Proteína de Ligação a TATA-Box/metabolismo , Fator de Transcrição TFIID/genética , Fator de Transcrição TFIID/metabolismo , Fatores de Transcrição/genética
5.
Neuron ; 93(2): 331-347, 2017 Jan 18.
Artigo em Inglês | MEDLINE | ID: mdl-28065649

RESUMO

Fragile X-associated tremor/ataxia syndrome (FXTAS) is a neurodegenerative disorder caused by a limited expansion of CGG repeats in the 5' UTR of FMR1. Two mechanisms are proposed to cause FXTAS: RNA gain-of-function, where CGG RNA sequesters specific proteins, and translation of CGG repeats into a polyglycine-containing protein, FMRpolyG. Here we developed transgenic mice expressing CGG repeat RNA with or without FMRpolyG. Expression of FMRpolyG is pathogenic, while the sole expression of CGG RNA is not. FMRpolyG interacts with the nuclear lamina protein LAP2ß and disorganizes the nuclear lamina architecture in neurons differentiated from FXTAS iPS cells. Finally, expression of LAP2ß rescues neuronal death induced by FMRpolyG. Overall, these results suggest that translation of expanded CGG repeats into FMRpolyG alters nuclear lamina architecture and drives pathogenesis in FXTAS.


Assuntos
Ataxia/genética , Proteínas de Ligação a DNA/metabolismo , Proteína do X Frágil da Deficiência Intelectual/genética , Síndrome do Cromossomo X Frágil/genética , Proteínas de Membrana/metabolismo , Lâmina Nuclear/metabolismo , Peptídeos/genética , Biossíntese de Proteínas , RNA Mensageiro/metabolismo , Tremor/genética , Expansão das Repetições de Trinucleotídeos/genética , Animais , Ataxia/metabolismo , Encéfalo/metabolismo , Encéfalo/patologia , Proteína do X Frágil da Deficiência Intelectual/metabolismo , Síndrome do Cromossomo X Frágil/metabolismo , Humanos , Masculino , Camundongos , Camundongos Transgênicos , Lâmina Nuclear/patologia , Peptídeos/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Tremor/metabolismo
6.
Toxicol In Vitro ; 40: 1-10, 2017 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-27979589

RESUMO

Benzene is extensively used in industry despite its leukemogenic activity, representing a significant occupational hazard. We investigated if long-term treatment with low-doses hydroquinone (HQ), a benzene metabolite, might be sufficient to alter in vitro the epigenetic signature underlining LINE-1 sequences, a poorly explored step in health risks associated with benzene exposure. In HL-60 cell line, exploring the epigenetic events occurring in chromatin, we found the transient instauration of the distinctive signature combining the repressive H3Lys27 tri-methylation mark and the activating H3Lys4 tri-methylation mark (H3K27me3/H3K4me3), indicating a tendency toward a poised chromatin conformation. These alterations are lost in time after short-term treatments, while the long-term setting, performed using a concentration within the levels of total HQ in peripheral blood of benzene-exposed workers, showed a gradual increase in H3K4me3. We observed the absence of statistically significant variations in DNA methylation and expression levels of LINE-1, despite a decrease in protein levels of UHRF1, DNA methyl-transferases and histone methyl-transferases. In conclusion, in vitro treatment with low-dose HQ determined the instauration of a reversible poised state of chromatin in LINE-1 sequences, suggesting that prolonged exposure could cause persistent epigenetic alterations.


Assuntos
Hidroquinonas/toxicidade , Elementos Nucleotídeos Longos e Dispersos/efeitos dos fármacos , Proteínas Estimuladoras de Ligação a CCAAT/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Cromatina/metabolismo , Metilação de DNA/efeitos dos fármacos , Epigênese Genética/efeitos dos fármacos , Células HL-60 , Histonas/metabolismo , Humanos , Metiltransferases/metabolismo , Ubiquitina-Proteína Ligases
7.
EMBO J ; 35(12): 1276-97, 2016 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-27103069

RESUMO

An intronic expansion of GGGGCC repeats within the C9ORF72 gene is the most common genetic cause of amyotrophic lateral sclerosis and frontotemporal dementia (ALS-FTD). Ataxin-2 with intermediate length of polyglutamine expansions (Ataxin-2 Q30x) is a genetic modifier of the disease. Here, we found that C9ORF72 forms a complex with the WDR41 and SMCR8 proteins to act as a GDP/GTP exchange factor for RAB8a and RAB39b and to thereby control autophagic flux. Depletion of C9orf72 in neurons partly impairs autophagy and leads to accumulation of aggregates of TDP-43 and P62 proteins, which are histopathological hallmarks of ALS-FTD SMCR8 is phosphorylated by TBK1 and depletion of TBK1 can be rescued by phosphomimetic mutants of SMCR8 or by constitutively active RAB39b, suggesting that TBK1, SMCR8, C9ORF72, and RAB39b belong to a common pathway regulating autophagy. While depletion of C9ORF72 only has a partial deleterious effect on neuron survival, it synergizes with Ataxin-2 Q30x toxicity to induce motor neuron dysfunction and neuronal cell death. These results indicate that partial loss of function of C9ORF72 is not deleterious by itself but synergizes with Ataxin-2 toxicity, suggesting a double-hit pathological mechanism in ALS-FTD.


Assuntos
Esclerose Lateral Amiotrófica/patologia , Ataxina-2/metabolismo , Autofagia , Demência Frontotemporal/patologia , Neurônios Motores/fisiologia , Peptídeos/metabolismo , Proteínas/metabolismo , Proteína C9orf72 , Morte Celular , Humanos , Neurônios Motores/metabolismo
8.
Exp Cell Res ; 342(2): 145-58, 2016 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-26968636

RESUMO

Although chemical inhibition of the DNA damage response (DDR) in cancer cells triggers cell death, it is not clear if the fork blockade achieved with inhibitors that neutralise proteins of the replisome is sufficient on its own to overcome the DDR. Monoclonal antibodies to PCNA, which block the DNA elongation process in vitro, have been developed. When these antibodies were transduced into cancer cells, they are able to inhibit the incorporation of nucleoside analogues. When co-delivered with anti-PCNA siRNA, the cells were flattened and the size of their nuclei increased by up to 3-fold, prior to cell death. Analysis of these nuclei by super-resolution microscopy revealed the presence of large numbers of phosphorylated histone H2AX foci. A senescence-like phenotype of the transduced cells was also observed upon delivery of the corresponding Fab molecules or following PCNA gene disruption or when the Fab fragment of an antibody that neutralises DNA polymerase alpha was used. Primary melanoma cells and leukaemia cells that are resistant to chemical inhibitors were similarly affected by these antibody treatments. These results demonstrate that transduced antibodies can trigger a lethal DNA replication stress, which kills cancer cells by abolishing the biological activity of several constituents of the replisome.


Assuntos
Anticorpos Monoclonais Murinos/farmacologia , Antineoplásicos/farmacologia , Replicação do DNA/efeitos dos fármacos , DNA de Neoplasias/genética , Animais , Quebras de DNA de Cadeia Dupla , DNA Polimerase III/antagonistas & inibidores , DNA de Neoplasias/metabolismo , Resistencia a Medicamentos Antineoplásicos , Ensaios de Seleção de Medicamentos Antitumorais , Técnicas de Silenciamento de Genes , Células HeLa , Histonas/metabolismo , Humanos , Fragmentos Fab das Imunoglobulinas/farmacologia , Camundongos Endogâmicos BALB C , Antígeno Nuclear de Célula em Proliferação/genética , Antígeno Nuclear de Célula em Proliferação/imunologia , Antígeno Nuclear de Célula em Proliferação/metabolismo , Estresse Fisiológico
9.
Cell Cycle ; 14(16): 2655-66, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26101806

RESUMO

The transcription factor ATF7 undergoes multiple post-translational modifications, each of which has distinct effects upon ATF7 function. Here, we show that ATF7 phosphorylation on residue Thr112 exclusively occurs during mitosis, and that ATF7 is excluded from the condensed chromatin. Both processes are CDK1/cyclin B dependent. Using a transduced neutralizing monoclonal antibody directed against the Thr112 epitope in living cells, we could demonstrate that Thr112 phosphorylation protects endogenous ATF7 protein from degradation, while it has no effect on the displacement of ATF7 from the condensed chromatin. The crucial role of Thr112 phosphorylation in stabilizing ATF7 protein during mitosis was confirmed using phospho-mimetic and phospho-deficient mutants. Finally, silencing ATF7 by CRISPR/Cas9 technology leads to a decrease of cyclin D1 protein expression levels. We propose that mitotic stabilized ATF7 protein re-localizes onto chromatin at the end of telophase and contributes to induce the cyclin D1 gene expression.


Assuntos
Fatores Ativadores da Transcrição/metabolismo , Ciclina D1/genética , Quinases Ciclina-Dependentes/fisiologia , Mitose , Animais , Proteína Quinase CDC2 , Cromatina/metabolismo , Ciclina D1/metabolismo , Células HeLa , Humanos , Camundongos , Fosforilação , Ligação Proteica , Processamento de Proteína Pós-Traducional , Estabilidade Proteica , Ativação Transcricional
10.
MAbs ; 5(4): 518-22, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23765067

RESUMO

Antibodies are valuable tools for functional studies in vitro, but their use in living cells remains challenging because they do not naturally cross the cell membrane. Here, we present a simple and highly efficient method for the intracytoplasmic delivery of any antibody into cultured cells. By following the fate of monoclonal antibodies that bind to nuclear antigens, it was possible to image endogenous targets and to show that inhibitory antibodies are able to induce cell growth suppression or cell death. Our electrotransfer system allowed the cancer cells we studied to be transduced without loss of viability and may have applications for a variety of intracellular immuno-interventions.


Assuntos
Anticorpos Monoclonais Murinos , Antígenos Nucleares , Apoptose , Neoplasias , Anticorpos Monoclonais Murinos/química , Anticorpos Monoclonais Murinos/imunologia , Anticorpos Monoclonais Murinos/farmacologia , Antígenos Nucleares/química , Antígenos Nucleares/imunologia , Antígenos Nucleares/metabolismo , Apoptose/efeitos dos fármacos , Apoptose/imunologia , Morte Celular/efeitos dos fármacos , Células HeLa , Humanos , Neoplasias/química , Neoplasias/imunologia , Neoplasias/metabolismo
11.
J Immunol Methods ; 369(1-2): 42-50, 2011 Jun 30.
Artigo em Inglês | MEDLINE | ID: mdl-21501618

RESUMO

In spite of their many potential applications, recombinant antibody molecules selected by phage display are rarely available commercially, one reason being the absence of robust bacterial expression systems that yield sufficient quantities of reagents for routine applications. We previously described the construction and validation of an intrabody library that allows the selection of single-chain Fv (scFv) fragments solubly expressed in the cytoplasm. Here, we show that it is possible to obtain monomeric scFvs binding specifically to human papillomavirus type 16 E6 and cellular gankyrin oncoproteins in quantities higher than 0.5 g/L of shake-flask culture in E. coli cytoplasm after auto-induction. In addition, stable bivalent scFvs of increased avidity were produced by tagging the scFvs with the dimeric glutathione-S-transferase enzyme (GST). These minibody-like molecules were further engineered by fusion with green fluorescent protein (GFPuv), leading to high yield of functional bivalent fluorescent antibody fragments. Our results demonstrate that scFvs selected from an intrabody library can be engineered into cost-effective bivalent reagents suitable for many biomedical and industrial applications.


Assuntos
Proteínas Oncogênicas Virais/análise , Biblioteca de Peptídeos , Complexo de Endopeptidases do Proteassoma/análise , Proteínas Proto-Oncogênicas/análise , Proteínas Repressoras/análise , Anticorpos de Cadeia Única/imunologia , Células HeLa , Papillomavirus Humano 16/química , Papillomavirus Humano 16/imunologia , Humanos , Modelos Moleculares , Proteínas Oncogênicas Virais/química , Proteínas Oncogênicas Virais/imunologia , Complexo de Endopeptidases do Proteassoma/química , Complexo de Endopeptidases do Proteassoma/imunologia , Estrutura Quaternária de Proteína , Proteínas Proto-Oncogênicas/química , Proteínas Proto-Oncogênicas/imunologia , Proteínas Repressoras/química , Proteínas Repressoras/imunologia , Anticorpos de Cadeia Única/análise , Anticorpos de Cadeia Única/química
12.
Biomaterials ; 31(7): 1699-706, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19954837

RESUMO

In this work, we designed replica particles based on poly (L-lysine) (PLL) polymers crosslinked via a homobifunctional linker to support coadsorption of a plasmid DNA and a peptide hormone for concurrent transfection and induction of a cellular function. PLL replica particles (PLL(RP)) were prepared by infiltrating polymer into mesoporous silica (MS) particles, crosslinking the adsorbed chains by using a homobifunctional crosslinker and finally removing the template particles. Moreover, we verified their cytotoxicity. Furthermore, based on this PLL(RP) gene delivery system, we simultaneously evaluated the melanin stimulation and gene expression in these cells by fluorescence microscopy. To further understand the bi-functionality, we labeled the SPT7pTL and PGA-alpha-MSH with YOYO-1 and Rhodamine, respectively, to follow its intracellular pathway by confocal microscopy. Our data suggests that the PLL(RP) is a promising vector for gene therapy and hormone stimulation.


Assuntos
Técnicas de Transferência de Genes , Nanoestruturas/química , Polilisina/farmacologia , alfa-MSH/farmacologia , Adsorção/efeitos dos fármacos , Animais , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Espaço Intracelular/efeitos dos fármacos , Espaço Intracelular/metabolismo , Melaninas/biossíntese , Redes e Vias Metabólicas/efeitos dos fármacos , Camundongos , Nanoestruturas/ultraestrutura , Plasmídeos/genética , Ácido Poliglutâmico/farmacologia , Porosidade/efeitos dos fármacos , alfa-MSH/metabolismo
13.
J Immunol ; 183(4): 2303-11, 2009 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-19620299

RESUMO

Helios is a member of the Ikaros family of zinc finger transcription factors. It is expressed mainly in T cells, where it associates with Ikaros-containing complexes and has been proposed to act as a rate-limiting factor for Ikaros function. Overexpression of wild-type or dominant-negative Helios isoforms profoundly alters alphabeta T cell differentiation and activation, and endogenous Helios is expressed at strikingly high levels in regulatory T cells. Helios has also been implicated as a tumor suppressor in human T cell acute lymphoblastic leukemias. These studies suggest a central role for Helios in T cell development and homeostasis, but whether this protein is physiologically required in T cells is unclear. We report herein that inactivation of the Helios gene by homologous recombination does not impair the differentiation and effector cell function of alphabeta and gammadelta T cells, NKT cells, and regulatory T cells. These results suggest that Helios is not essential for T cells, and that its function can be compensated for by other members of the Ikaros family.


Assuntos
Diferenciação Celular/genética , Diferenciação Celular/imunologia , Proteínas de Ligação a DNA/deficiência , Proteínas de Ligação a DNA/genética , Subpopulações de Linfócitos T/citologia , Subpopulações de Linfócitos T/imunologia , Fatores de Transcrição/deficiência , Fatores de Transcrição/genética , Animais , Linfócitos T CD4-Positivos/citologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD8-Positivos/citologia , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Proliferação de Células , Células Cultivadas , Células Clonais , Proteínas de Ligação a DNA/fisiologia , Inativação Gênica/imunologia , Fator de Transcrição Ikaros/genética , Fator de Transcrição Ikaros/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Família Multigênica/imunologia , Receptores de Antígenos de Linfócitos T/fisiologia , Subpopulações de Linfócitos T/metabolismo , Fatores de Transcrição/fisiologia
14.
Mol Cancer Ther ; 6(6): 1728-35, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17575104

RESUMO

Cervical cancer is caused by high-risk types of human papillomaviruses (HPV) that encode the E6 and E7 oncogenes. Silencing of E6 gene expression in HPV-positive cell lines by transfection of small interfering RNA (siRNA) with cationic lipids restores the dormant p53 tumor suppressor pathway. Because cationic lipids can also be used for intracytoplasmic delivery of proteins, we tested whether the delivery of monoclonal antibodies that bind to HPV16 E6 and neutralize its biological activity in vitro could restore p53 function in tumor cells. Here, we show that the 4C6 antibody is efficiently delivered into the cell cytoplasm using a lipidic reagent used for siRNA transfection. The delivery of 4C6 resulted in the nuclear accumulation of p53 protein in CaSki and SiHa cells but not in HeLa cells. Furthermore, the antibody-mediated p53 response was dramatically increased when a peptide corresponding to the 4C6 epitope and bearing a COOH-terminal cysteine residue was added to the transduction mixture. We found that a fraction of the added peptides were dimers that allowed the formation of antibody polymers adsorbed onto the lipidic matrix. With this system, the proliferation of CaSki and SiHa cells was strongly diminished, but no apoptosis was detectable. Remarkably, cell growth was almost totally suppressed by the addition of E6-specific siRNA to the transduction complex. The results indicate that the activity of E6 oncoprotein can be down-regulated in vivo by lipid-mediated antibody delivery and that antibodies and siRNA act synergistically when codelivered. This novel targeting strategy is simple to implement and may find therapeutic applications.


Assuntos
Anticorpos Monoclonais/administração & dosagem , Divisão Celular/genética , Divisão Celular/imunologia , Papillomaviridae/imunologia , RNA Interferente Pequeno , Neoplasias do Colo do Útero/patologia , Proteínas Virais/imunologia , Sequência de Bases , Feminino , Humanos , Microscopia de Fluorescência , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Células Tumorais Cultivadas
15.
J Mol Biol ; 371(1): 235-44, 2007 Aug 03.
Artigo em Inglês | MEDLINE | ID: mdl-17560603

RESUMO

Abnormally expanded polyglutamine (polyQ) tracts provide a gain of toxic functions to nine otherwise unrelated human proteins and induce progressive neurodegenerative diseases. Over the past ten years, it was suggested that only polyQ tracts longer than a specific threshold adopt a particular structure, which would be the cause of the apparent polyQ length-dependent toxicity threshold observed in polyQ diseases. We have used a combination of biochemical and biophysical approaches to compare the structural properties of polyQ of pathogenic and non-pathogenic lengths under various conditions. We observe that pathogenic and non-pathogenic polyQ, as soluble species and upon interaction with a partner, during aggregation, or as mature aggregates, display very similar structural properties. PolyQ length only influences the aggregation kinetics and, to a lesser extent, the stability of the aggregates. We thus propose that polyQ toxicity does not depend on a structural transition occurring above a specific threshold, but rather that polyQ tracts are inherently toxic sequences, whose deleterious effect gradually increases with their length. We discuss how polyQ properties and other cellular factors may explain the existence of an apparent polyQ length-dependent toxicity threshold.


Assuntos
Peptídeos/química , Peptídeos/metabolismo , Proteínas , Sequência de Aminoácidos , Humanos , Doenças Neurodegenerativas/metabolismo , Ressonância Magnética Nuclear Biomolecular , Peptídeos/genética , Proteínas/genética , Proteínas/toxicidade
16.
J Biol Chem ; 279(32): 34048-61, 2004 Aug 06.
Artigo em Inglês | MEDLINE | ID: mdl-15175332

RESUMO

Tamoxifen is a selective estrogen receptor modulator widely used for the prophylactic treatment of breast cancer. In addition to the estrogen receptor (ER), tamoxifen binds with high affinity to the microsomal antiestrogen binding site (AEBS), which is involved in ER-independent effects of tamoxifen. In the present study, we investigate the modulation of the biosynthesis of cholesterol in tumor cell lines by AEBS ligands. As a consequence of the treatment with the antitumoral drugs tamoxifen or PBPE, a selective AEBS ligand, we show that tumor cells produced a significant concentration- and time-dependent accumulation of cholesterol precursors. Sterols have been purified by HPLC and gas chromatography, and their chemical structures determined by mass spectrometric analysis. The major metabolites identified were 5alpha-cholest-8-en-3beta-ol for tamoxifen treatment and 5alpha-cholest-8-en-3beta-ol and cholesta-5,7-dien-3beta-ol, for PBPE treatment, suggesting that these AEBS ligands affect at least two enzymatic steps: the 3beta-hydroxysterol-Delta8-Delta7-isomerase and the 3beta-hydroxysterol-Delta7-reductase. Steroidal antiestrogens such as ICI 182,780 and RU 58,668 did not affect these enzymatic steps, because they do not bind to the AEBS. Transient co-expression of human 3beta-hydroxysterol-Delta8-Delta7-isomerase and 3beta-hydroxysterol-Delta7-reductase and immunoprecipitation experiments showed that both enzymes were required to reconstitute the AEBS in mammalian cells. Altogether, these data provide strong evidence that the AEBS is a hetero-oligomeric complex including 3beta-hydroxysterol-Delta8-Delta7-isomerase and the 3beta-hydroxysterol-Delta7-reductase as subunits that are necessary and sufficient for tamoxifen binding in mammary cells. Furthermore, because selective AEBS ligands are antitumoral compounds, these data suggest a link between cholesterol metabolism at a post-lanosterol step and tumor growth control. These data afford both the identification of the AEBS and give new insight into a novel molecular mechanism of action for drugs of clinical value.


Assuntos
Antagonistas de Estrogênios/metabolismo , Microssomos/química , Receptores de Droga/química , Tamoxifeno/metabolismo , Animais , Sítios de Ligação , Neoplasias da Mama , Células COS , Chlorocebus aethiops , Colesterol/biossíntese , Colesterol/metabolismo , Cromatografia Líquida de Alta Pressão , Antagonistas de Estrogênios/farmacologia , Cromatografia Gasosa-Espectrometria de Massas , Expressão Gênica , Humanos , Técnicas de Imunoadsorção , Osteossarcoma , Oxirredutases atuantes sobre Doadores de Grupo CH-CH/genética , Oxirredutases atuantes sobre Doadores de Grupo CH-CH/metabolismo , Éteres Fenílicos/farmacologia , Pirrolidinas/farmacologia , Esteroide Isomerases/genética , Esteroide Isomerases/metabolismo , Tamoxifeno/farmacologia , Transfecção , Células Tumorais Cultivadas
17.
J Biol Chem ; 279(21): 21802-7, 2004 May 21.
Artigo em Inglês | MEDLINE | ID: mdl-14988402

RESUMO

BTAF1 (formerly named TAF(II)170/TAF-172) is an essential, evolutionarily conserved member of the SNF2-like family of ATPase proteins and together with TATA-binding protein (TBP) forms the B-TFIID complex. BTAF1 has been proposed to play a key role in the dynamic regulation of TBP function in RNA polymerase II transcription. We have determined the structure of native B-TFIID purified from human cells by electron microscopy and by image analysis of single particles at a resolution of 28 A. B-TFIID is 15 x 9 nm in size and is organized into a large domain of about 170 kDa, which can be subdivided into two domains. Extending from this domain is a long thumb, which in turn is divided into subdomains of about 25, 15, and 35 kDa, the largest of which is located at the end of the thumb. Immunolabeling experiments localize the extreme carboxyl terminus of BTAF1 within the 170-kDa domain, whereas the amino terminus and TBP co-localize to the end of the protruding thumb. The central portion of BTAF1 localizes to the base of the thumb. Comparison of the native B-TFIID with its recombinant form shows that both share a similar domain organization. Collectively, these data provide the first structural model of the B-TFIID complex and map its key functional domains.


Assuntos
Fatores Associados à Proteína de Ligação a TATA/química , Proteína de Ligação a TATA-Box/química , Fator de Transcrição TFIID/química , Trifosfato de Adenosina/química , Células HeLa , Humanos , Microscopia Eletrônica , Microscopia Imunoeletrônica , Ligação Proteica , Conformação Proteica , Estrutura Terciária de Proteína , RNA Polimerase II/química , Proteínas Recombinantes/química , Fatores Associados à Proteína de Ligação a TATA/genética , Fator de Transcrição TFIID/genética , Transcrição Gênica
18.
J Cell Sci ; 115(Pt 17): 3439-48, 2002 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-12154074

RESUMO

The transcriptional intermediary factor 1 (TIF1) family protein TIF1beta is a corepressor for Krüppel-associated box (KRAB)-domain-containing zinc finger proteins and plays a critical role in early embryogenesis. Here, we examined TIF1beta distribution in the nucleus of mouse embryonic carcinoma F9 cells during retinoic-acid-induced primitive endodermal differentiation. Using confocal immunofluorescence microscopy, we show that, although TIF1beta is diffusely distributed throughout the nucleoplasm of undifferentiated cells, it relocates and concentrates into distinct foci of centromeric heterochromatin in differentiated cells characterized by a low proliferation rate and a well developed cytokeratin network. This relocation was not observed in isoleucine-deprived cells, which are growth arrested, or in compound RXR alpha(-/-)/RAR gamma(-/-) null mutant cells, which are resistant to RA-induced differentiation. Amino-acid substitutions in the PxVxL motif of TIF1beta, which abolish interaction with members of the heterochromatin protein 1 (HP1) family, prevent its centromeric localization in differentiated cells. Collectively, these data provide compelling evidence for a dynamic nuclear compartmentalization of TIF1beta that is regulated during cell differentiation through a mechanism that requires HP1 interaction.


Assuntos
Diferenciação Celular/fisiologia , Centrômero/metabolismo , Proteínas Cromossômicas não Histona/metabolismo , Proteínas de Ligação a DNA/metabolismo , Heterocromatina/metabolismo , Proteínas Nucleares , Proteínas Repressoras/metabolismo , Fatores de Transcrição , Sequência de Aminoácidos , Animais , Ciclo Celular/fisiologia , Núcleo Celular/metabolismo , Homólogo 5 da Proteína Cromobox , Proteínas de Ligação a DNA/genética , Embrião de Mamíferos/fisiologia , Regulação da Expressão Gênica no Desenvolvimento , Isoleucina/metabolismo , Camundongos , Microscopia Confocal , Dados de Sequência Molecular , Proteínas Repressoras/genética , Tretinoína/metabolismo , Proteína 28 com Motivo Tripartido , Células Tumorais Cultivadas
19.
EMBO Rep ; 3(8): 753-9, 2002 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12151334

RESUMO

A 400 bp fragment of the spermatogonia-specific Stra8 locus was sufficient to direct gene expression to the germinal stem cells in transgenic mice. A fractionation procedure was devised, based on immunomagnetic sorting of cells in which the promoter drives the expression of a surface functionally neutral protein tag. The purified cells expressed the known molecular markers of spermatogonia Rbm, cyclin A2 and EP-Cam, and the beta1- and alpha6-integrins characteristic of the stem cell fraction. A 700-fold enrichment in stem cells was determined by the ability of the purified fractions to re-establish spermatogenesis in germ cell-depleted recipient testes.


Assuntos
Separação Celular/métodos , Espermatogônias/citologia , Células-Tronco/metabolismo , Transgenes , Fatores Etários , Animais , Sequência de Bases , Antígenos CD4/biossíntese , Ciclina A/metabolismo , Ciclina A2 , DNA Complementar/metabolismo , Éxons , Biblioteca Gênica , Genes Reporter , Humanos , Imuno-Histoquímica , Integrina alfa6/metabolismo , Integrina beta1/metabolismo , Luciferases/metabolismo , Magnetismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Camundongos Transgênicos , Dados de Sequência Molecular , Proteínas Nucleares , Plasmídeos , Regiões Promotoras Genéticas , Proteínas de Ligação a RNA/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Testículo/metabolismo , Testículo/patologia , Distribuição Tecidual
20.
J Biol Chem ; 277(28): 24859-62, 2002 Jul 12.
Artigo em Inglês | MEDLINE | ID: mdl-12032135

RESUMO

Nuclear retinoic acid (RA) receptors (RARs) are phosphorylated at conserved serine residues located in their N-terminal domain. Phosphorylation of RARgamma2 at these residues is increased in response to RA subsequently to the activation of p38MAPK. We show here that this RA-induced phosphorylation of RARgamma2 resulted from the down-regulation of the phosphatidylinositol 3-kinase (PI3K)/Akt pathway. By overexpressing Akt and by using agents that activated or inhibited the PI3K/Akt pathway, we also demonstrated that the RA-induced down-regulation of the PI3K/Akt pathway targeted not only the phosphorylation of RARgamma2 but also the turnover and transcriptional activity of the receptor. Altogether these data indicate that the PI3K/Akt pathway plays an important role in retinoic acid signaling.


Assuntos
Regulação para Baixo , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Serina-Treonina Quinases , Proteínas Proto-Oncogênicas/metabolismo , Receptores do Ácido Retinoico/fisiologia , Transcrição Gênica/efeitos dos fármacos , Tretinoína/farmacologia , Animais , Linhagem Celular , Ativação Enzimática , Hidrólise , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Fosforilação , Proteínas Proto-Oncogênicas c-akt , Transcrição Gênica/fisiologia , Proteínas Quinases p38 Ativadas por Mitógeno , Receptor gama de Ácido Retinoico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA