Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 85
Filtrar
1.
EBioMedicine ; 41: 333-344, 2019 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-30827928

RESUMO

BACKGROUND: Innate lymphoid cells (ILCs) are a newly discovered family of immune cells that have similar cytokine-secreting profiles as T helper cell subsets. Although ILCs are critical for host defense against infections and tissue homeostasis, their roles in tumor development are not well established. METHODS: We studied the function of ILC3 cells in the liver for the development of hepatocellular carcinoma (HCC) in murine HCC models using flow cytometry, adoptive transfer, and in vitro functional assays. FINDINGS: We found that ILC3 lacking the natural cytotoxicity-triggering receptor (NCR-ILC3) promoted the development of HCC in response to interleukin 23 (IL-23). IL-23 serum level is elevated in HCC patients and its high expression is associated with poor clinical outcomes. We found that IL-23 could promote tumor development in murine HCC tumor models. IL-23 promoted the expansion of NCR-ILC3 and its differentiation from group 1 ILCs (ILC1s). Furthermore, NCR-ILC3 initiated IL-17 production upon IL-23 stimulation and directly inhibited CD8+ T cell immunity by promoting lymphocyte apoptosis and limiting their proliferation. INTERPRETATION: Together, our findings suggest that NCR-ILC3 initiates the IL-17-rich immunosuppressive tumor microenvironment and promotes the development of HCC, thus may serve as a promising target for future cancer immunotherapy. FUND: This work was supported by grants from National Natural Science Foundation of China (81471586, 81571556), the Priority Academic Program Development of Jiangsu Higher Education Institutions, the collaborative Innovation Center of Hematology, start-up grant from National University of Singapore, the Cancer Prevention and Research Institute of Texas CPRIT (RR180017), and the National Cancer Institute's Cancer Center Support (Core) Grant CA016672 (to The University of Texas MD Anderson Cancer Center).


Assuntos
Carcinoma Hepatocelular/patologia , Interleucina-17/metabolismo , Interleucina-23/metabolismo , Neoplasias Hepáticas/patologia , Animais , Apoptose , Carcinoma Hepatocelular/imunologia , Carcinoma Hepatocelular/metabolismo , Diferenciação Celular , Linhagem Celular Tumoral , Técnicas de Cocultura , Modelos Animais de Doenças , Imunidade Inata , Interleucina-12/metabolismo , Interleucina-17/análise , Interleucina-23/análise , Interleucina-23/genética , Neoplasias Hepáticas/imunologia , Neoplasias Hepáticas/metabolismo , Linfócitos/citologia , Linfócitos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Subunidades Proteicas/deficiência , Subunidades Proteicas/genética , Subunidades Proteicas/metabolismo , Transplante Homólogo
2.
DNA Repair (Amst) ; 73: 64-70, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30470508

RESUMO

Human DNA polymerase δ is normally present in unstressed, non-dividing cells as a heterotetramer (Pol δ4). Its smallest subunit, p12, is transiently degraded in response to UV damage, as well as during the entry into S-phase, resulting in the conversion of Pol δ4 to a trimer (Pol δ3). In order to further understand the specific cellular roles of these two forms of Pol δ, the gene (POLD4) encoding p12 was disrupted by CRISPR/Cas9 to produce p12 knockout (p12KO) cells. Thus, Pol δ4 is absent in p12KO cells, leaving Pol δ3 as the sole source of Pol δ activity. GFP reporter assays revealed that the p12KO cells exhibited a defect in homologous recombination (HR) repair, indicating that Pol δ4, but not Pol δ3, is required for HR. Expression of Flag-tagged p12 in p12KO cells to restore Pol δ4 alleviated the HR defect. These results establish a specific requirement for Pol δ4 in HR repair. This leads to the prediction that p12KO cells should be more sensitive to chemotherapeutic agents, and should exhibit synthetic lethal killing by PARP inhibitors. These predictions were confirmed by clonogenic cell survival assays of p12KO cells treated with cisplatin and mitomycin C, and with the PARP inhibitors Olaparib, Talazoparib, Rucaparib, and Niraparib. The sensitivity to PARP inhibitors in H1299-p12KO cells was alleviated by expression of Flag-p12. These findings have clinical significance, as the expression levels of p12 could be a predictive biomarker of tumor response to PARP inhibitors. In addition, small cell lung cancers (SCLC) are known to exhibit a defect in p12 expression. Analysis of several SCLC cell lines showed that they exhibit hypersensitivity to PARP inhibitors, providing evidence that loss of p12 expression could represent a novel molecular basis for HR deficiency.


Assuntos
DNA Polimerase III/antagonistas & inibidores , DNA Polimerase III/genética , Técnicas de Inativação de Genes , Recombinação Homóloga/genética , Inibidores de Poli(ADP-Ribose) Polimerases/farmacologia , Subunidades Proteicas/deficiência , Subunidades Proteicas/genética , Linhagem Celular Tumoral , Cisplatino/farmacologia , Quebras de DNA de Cadeia Dupla/efeitos dos fármacos , DNA Polimerase III/deficiência , Células HeLa , Recombinação Homóloga/efeitos dos fármacos , Humanos , Mitomicina/farmacologia
3.
J Proteome Res ; 17(11): 3959-3975, 2018 11 02.
Artigo em Inglês | MEDLINE | ID: mdl-30285449

RESUMO

The α7-nicotinic acetylcholine receptor (α7-nAChR) is a ligand-gated ion channel that is expressed widely in vertebrates and is the principal high-affinity α-bungarotoxin (α-bgtx) binding protein in the mammalian CNS. α7-nAChRs associate with proteins that can modulate its properties. The α7-nAChR interactome is the summation of proteins interacting or associating with α7-nAChRs in a protein complex. To identify an α7-nAChR interactome in neural tissue, we isolated α-bgtx-affinity protein complexes from wild-type and α7-nAChR knockout (α7 KO) mouse whole brain tissue homogenates using α-bgtx-affinity beads. Affinity precipitated proteins were trypsinized and analyzed with an Orbitrap Fusion mass spectrometer. Proteins isolated with the α7-nAChR specific ligand, α-bgtx, were determined to be α7-nAChR associated proteins. The α7-nAChR subunit and 120 additional proteins were identified. Additionally, 369 proteins were identified as binding to α-bgtx in the absence of α7-nAChR expression, thereby identifying nonspecific proteins for α7-nAChR investigations using α-bgtx enrichment. These results expand on our previous investigations of α7-nAChR interacting proteins using α-bgtx-affinity bead isolation by controlling for differences between α7-nAChR and α-bgtx-specific proteins, developing an improved protein isolation methodology, and incorporating the latest technology in mass spectrometry. The α7-nAChR interactome identified in this study includes proteins associated with the expression, localization, function, or modulation of α7-nAChRs, and it provides a foundation for future studies to elucidate how these interactions contribute to human disease.


Assuntos
Encéfalo/metabolismo , Bungarotoxinas/química , Mapeamento de Interação de Proteínas/métodos , Subunidades Proteicas/química , Proteoma/metabolismo , Receptor Nicotínico de Acetilcolina alfa7/química , Animais , Encéfalo/citologia , Bungarotoxinas/metabolismo , Cromatografia de Afinidade , Expressão Gênica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neurônios/citologia , Neurônios/metabolismo , Peptídeos/análise , Ligação Proteica , Subunidades Proteicas/deficiência , Subunidades Proteicas/genética , Proteólise , Proteoma/genética , Proteômica/métodos , Receptor Nicotínico de Acetilcolina alfa7/deficiência , Receptor Nicotínico de Acetilcolina alfa7/genética
4.
Biochim Biophys Acta Mol Cell Biol Lipids ; 1863(9): 1121-1131, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29902570

RESUMO

Phosphoinositides are bioactive lipids essential in the regulation of cell signaling as well as cytoskeleton and membrane dynamics. Their metabolism is highly active in blood platelets where they play a critical role during activation, at least through two well identified pathways involving phospholipase C and phosphoinositide 3-kinases (PI3K). Here, using a sensitive high-performance liquid chromatography-mass spectrometry method recently developed, we monitored for the first time the profiling of phosphatidylinositol (PI), PIP, PIP2 and PIP3 molecular species (fatty-acyl profiles) in human and mouse platelets during the course of stimulation by thrombin and collagen-related peptide. Furthermore, using class IA PI3K p110α or p110ß deficient mouse platelets and a pharmacological inhibitor, we show the crucial role of p110ß and the more subtle role of p110α in the production of PIP3 molecular species following stimulation. This comprehensive platelet phosphoinositides profiling provides important resources for future studies and reveals new information on phosphoinositides biology, similarities and differences in mouse and human platelets and unexpected dramatic increase in low-abundance molecular species of PIP2 during stimulation, opening new perspectives in phosphoinositide signaling in platelets.


Assuntos
Plaquetas/efeitos dos fármacos , Classe I de Fosfatidilinositol 3-Quinases/genética , Fosfatidilinositol 4,5-Difosfato/metabolismo , Fosfatos de Fosfatidilinositol/metabolismo , Animais , Plaquetas/citologia , Plaquetas/metabolismo , Proteínas de Transporte/farmacologia , Classe I de Fosfatidilinositol 3-Quinases/antagonistas & inibidores , Classe I de Fosfatidilinositol 3-Quinases/deficiência , Inibidores Enzimáticos/farmacologia , Expressão Gênica , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Peptídeos/farmacologia , Ativação Plaquetária/efeitos dos fármacos , Cultura Primária de Células , Subunidades Proteicas/antagonistas & inibidores , Subunidades Proteicas/deficiência , Subunidades Proteicas/genética , Pirimidinonas/farmacologia , Trombina/farmacologia , ortoaminobenzoatos/farmacologia
5.
Sci Rep ; 7(1): 7169, 2017 08 02.
Artigo em Inglês | MEDLINE | ID: mdl-28769029

RESUMO

Succinyl Coenzyme A synthetase (SCS) is a key mitochondrial enzyme. Defected SCS ADP-forming ß subunit (SCS A-ß) is linked to lethal infantile Leigh or leigh-like syndrome. However, the impacts of SCS A-ß deficiency on mitochondria specifically in neurons have not yet been comprehensively investigated. Here, by down-regulating the expression levels of SCS A-ß in cultured mouse neurons, we have found that SCS A-ß deficiency induces severe mitochondrial dysfunction including lowered oxidative phosphorylation (OXPHOS) efficiency, increased mitochondrial superoxide production, and mtDNA depletion as well as aberrations of mitochondrial fusion and fission proteins, which eventually leads to neuronal stress. Our data also suggest that the deregulation of mitochondrial nucleoside diphosphate kinase (NDPK) together with defects in mitochondrial transcription factors including mitochondrial DNA pol γ and Twinkle contribute to SCS A-ß deficiency-mediated mtDNA instability. Furthermore, we have found that SCS A-ß deficiency has detrimental influence on neuronal mitochondrial dynamics. Put together, the results have furnished our knowledge on the pathogenesis of SCS A-ß deficiency-related mitochondrial diseases and revealed the vital role of SCS A-ß in maintaining neuronal mitochondrial quality control and neuronal physiology.


Assuntos
DNA Mitocondrial , Mitocôndrias/genética , Mitocôndrias/metabolismo , Neurônios/metabolismo , Subunidades Proteicas/deficiência , Succinato-CoA Ligases/deficiência , Difosfato de Adenosina/metabolismo , Trifosfato de Adenosina/metabolismo , Animais , Dendritos/metabolismo , Dosagem de Genes , Expressão Gênica , Potencial da Membrana Mitocondrial , Camundongos , Camundongos Knockout , Subunidades Proteicas/genética , Espécies Reativas de Oxigênio/metabolismo , Succinato-CoA Ligases/genética , Sinapses/metabolismo
6.
Alcohol ; 57: 65-70, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27793544

RESUMO

The prevalent co-abuse of nicotine and alcohol suggests a common neural mechanism underlying the actions of the two drugs. Nicotine, the addictive component of tobacco, activates nicotinic acetylcholine receptors (nAChRs) containing the α6 subunit (α6* nAChRs) in dopaminergic (DAergic) neurons of the ventral tegmental area (VTA), a region known to be crucial for drug reward. Recent evidence suggests that ethanol may potentiate ACh activation of these receptors as well, although whether α6* nAChR expression is necessary for behavioral effects of acute ethanol exposure is unknown. We compared binge-like ethanol consumption and ethanol reward sensitivity between knockout (KO) mice that do not express chrna6 (the gene encoding the α6 nAChR subunit, the α6 KO line) and wild-type (WT) littermates using the Drinking-in-the-Dark (DID) and Conditioned Place Preference (CPP) assay, respectively. In the DID assay, α6 KO female and male mice consumed ethanol similarly to WT mice at all concentrations tested. In the CPP assay, 2.0-g/kg and 3.0-g/kg, but not 0.5-mg/kg, ethanol conditioned a place preference in WT female and male mice, whereas only 2.0-g/kg ethanol conditioned a place preference in α6 KO mice. Acute challenge with ethanol reduced locomotor activity, an effect that developed tolerance with repeated injections, similarly between genotypes in both female and male mice. Together, these data indicate that expression of α6* nAChRs is not required for binge-like ethanol consumption and reward, but modulate sensitivity to the rewarding properties of the drug.


Assuntos
Consumo de Bebidas Alcoólicas/metabolismo , Etanol/administração & dosagem , Subunidades Proteicas/deficiência , Receptores Nicotínicos/deficiência , Recompensa , Consumo de Bebidas Alcoólicas/psicologia , Animais , Relação Dose-Resposta a Droga , Feminino , Locomoção/efeitos dos fármacos , Locomoção/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
7.
Nature ; 520(7549): 697-701, 2015 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-25901683

RESUMO

TP53, a well-known tumour suppressor gene that encodes p53, is frequently inactivated by mutation or deletion in most human tumours. A tremendous effort has been made to restore p53 activity in cancer therapies. However, no effective p53-based therapy has been successfully translated into clinical cancer treatment owing to the complexity of p53 signalling. Here we demonstrate that genomic deletion of TP53 frequently encompasses essential neighbouring genes, rendering cancer cells with hemizygous TP53 deletion vulnerable to further suppression of such genes. POLR2A is identified as such a gene that is almost always co-deleted with TP53 in human cancers. It encodes the largest and catalytic subunit of the RNA polymerase II complex, which is specifically inhibited by α-amanitin. Our analysis of The Cancer Genome Atlas (TCGA) and Cancer Cell Line Encyclopedia (CCLE) databases reveals that POLR2A expression levels are tightly correlated with its gene copy numbers in human colorectal cancer. Suppression of POLR2A with α-amanitin or small interfering RNAs selectively inhibits the proliferation, survival and tumorigenic potential of colorectal cancer cells with hemizygous TP53 loss in a p53-independent manner. Previous clinical applications of α-amanitin have been limited owing to its liver toxicity. However, we found that α-amanitin-based antibody-drug conjugates are highly effective therapeutic agents with reduced toxicity. Here we show that low doses of α-amanitin-conjugated anti-epithelial cell adhesion molecule (EpCAM) antibody lead to complete tumour regression in mouse models of human colorectal cancer with hemizygous deletion of POLR2A. We anticipate that inhibiting POLR2A will be a new therapeutic approach for human cancers containing such common genomic alterations.


Assuntos
Neoplasias Colorretais/tratamento farmacológico , Neoplasias Colorretais/genética , Genes p53/genética , Proteína Supressora de Tumor p53/deficiência , Alfa-Amanitina/efeitos adversos , Alfa-Amanitina/química , Alfa-Amanitina/farmacologia , Alfa-Amanitina/uso terapêutico , Animais , Anticorpos/química , Anticorpos/imunologia , Antígenos de Neoplasias/imunologia , Domínio Catalítico , Moléculas de Adesão Celular/imunologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Neoplasias Colorretais/enzimologia , Neoplasias Colorretais/patologia , Bases de Dados Genéticas , Modelos Animais de Doenças , Molécula de Adesão da Célula Epitelial , Feminino , Deleção de Genes , Dosagem de Genes/genética , Humanos , Imunoconjugados/efeitos adversos , Imunoconjugados/química , Imunoconjugados/imunologia , Imunoconjugados/uso terapêutico , Camundongos , Subunidades Proteicas/química , Subunidades Proteicas/deficiência , Subunidades Proteicas/genética , RNA Polimerase II/antagonistas & inibidores , RNA Polimerase II/química , RNA Polimerase II/deficiência , RNA Polimerase II/genética , Proteína Supressora de Tumor p53/biossíntese , Proteína Supressora de Tumor p53/genética , Ensaios Antitumorais Modelo de Xenoenxerto
8.
Nucleic Acids Res ; 43(6): 3056-67, 2015 Mar 31.
Artigo em Inglês | MEDLINE | ID: mdl-25735743

RESUMO

Cohesin, which in somatic vertebrate cells consists of SMC1, SMC3, RAD21 and either SA1 or SA2, mediates higher-order chromatin organization. To determine how cohesin contributes to the establishment of tissue-specific transcriptional programs, we compared genome-wide cohesin distribution, gene expression and chromatin architecture in cerebral cortex and pancreas from adult mice. More than one third of cohesin binding sites differ between the two tissues and these show reduced overlap with CCCTC-binding factor (CTCF) and are enriched at the regulatory regions of tissue-specific genes. Cohesin/CTCF sites at active enhancers and promoters contain, at least, cohesin-SA1. Analyses of chromatin contacts at the Protocadherin (Pcdh) and Regenerating islet-derived (Reg) gene clusters, mostly expressed in brain and pancreas, respectively, revealed remarkable differences that correlate with the presence of cohesin. We could not detect significant changes in the chromatin contacts at the Pcdh locus when comparing brains from wild-type and SA1 null embryos. In contrast, reduced dosage of SA1 altered the architecture of the Reg locus and decreased the expression of Reg genes in the pancreas of SA1 heterozygous mice. Given the role of Reg proteins in inflammation, such reduction may contribute to the increased incidence of pancreatic cancer observed in these animals.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Cromatina/metabolismo , Proteínas Cromossômicas não Histona/metabolismo , Animais , Sítios de Ligação , Fator de Ligação a CCCTC , Caderinas/genética , Proteínas de Ciclo Celular/deficiência , Proteínas de Ciclo Celular/genética , Córtex Cerebral/metabolismo , Cromatina/química , Cromatina/genética , Proteínas Cromossômicas não Histona/deficiência , Proteínas Cromossômicas não Histona/genética , Expressão Gênica , Heterozigoto , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Família Multigênica , Pâncreas/metabolismo , Subunidades Proteicas/deficiência , Subunidades Proteicas/genética , Subunidades Proteicas/metabolismo , RNA/genética , RNA/metabolismo , Proteínas Repressoras/metabolismo , Distribuição Tecidual , Coesinas
9.
Cell Death Differ ; 21(9): 1442-50, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24902901

RESUMO

The phosphatidylinositol 3-kinase (PI3K) regulatory subunits p55α and p50α are coordinately transcriptionally upregulated by signal transducer and activator of transcription 3 (Stat3) at the onset of mammary gland involution, a process that requires Stat3. Deletion of both p55α and p50α subunits in vivo abrogated mammary epithelial cell death during involution. This was associated also with reduced cytosolic levels and activity of the cysteine protease cathepsin L, which is implicated in lysosomal-mediated programmed cell death (LM-PCD) and is upregulated in involution. Furthermore, involution is delayed in cathepsin L-deficient mice suggesting that the p55α/p50α subunits mediate cell death in part by elevating the level of cathepsin L resulting in increased cytosolic activity. Surprisingly, we found that p55α/p50α localize to the nucleus where they bind to chromatin and regulate transcription of a subset of inflammatory/acute phase genes that are also Stat3 targets. Our findings reveal a novel role for these PI3K regulatory subunits as regulators of LM-PCD in vivo.


Assuntos
Fosfatidilinositol 3-Quinases/química , Fosfatidilinositol 3-Quinases/metabolismo , Subunidades Proteicas/química , Subunidades Proteicas/metabolismo , Animais , Morte Celular/genética , Feminino , Masculino , Camundongos , Camundongos Knockout , Fosfatidilinositol 3-Quinases/deficiência , Fosfatidilinositol 3-Quinases/genética , Subunidades Proteicas/deficiência , Subunidades Proteicas/genética
10.
Cell Metab ; 20(1): 73-84, 2014 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-24836559

RESUMO

Bromodomain-containing protein 7 (BRD7) is a member of the bromodomain-containing protein family that is known to play a role as tumor suppressors. Here, we show that BRD7 is a component of the unfolded protein response (UPR) signaling through its ability to regulate X-box binding protein 1 (XBP1) nuclear translocation. BRD7 interacts with the regulatory subunits of phosphatidylinositol 3-kinase (PI3K) and increases the nuclear translocation of both p85α and p85ß and the spliced form of XBP1 (XBP1s). Deficiency of BRD7 blocks the nuclear translocation of XBP1s. Furthermore, our in vivo studies have shown that BRD7 protein levels are reduced in the liver of obese mice, and reinstating BRD7 levels in the liver restores XBP1s nuclear translocation, improves glucose homeostasis, and ultimately reduces the blood glucose levels in the obese and diabetic mouse models.


Assuntos
Proteínas Cromossômicas não Histona/metabolismo , Proteínas de Ligação a DNA/metabolismo , Glucose/metabolismo , Fosfatidilinositol 3-Quinase/metabolismo , Fatores de Transcrição/metabolismo , Animais , Linhagem Celular , Núcleo Celular/metabolismo , Proteínas Cromossômicas não Histona/antagonistas & inibidores , Proteínas Cromossômicas não Histona/genética , Proteínas de Ligação a DNA/química , Estresse do Retículo Endoplasmático , Células HEK293 , Humanos , Fígado/metabolismo , Masculino , Camundongos , Camundongos Obesos , Fosfatidilinositol 3-Quinase/química , Fosfatidilinositol 3-Quinase/deficiência , Subunidades Proteicas/química , Subunidades Proteicas/deficiência , Subunidades Proteicas/metabolismo , Interferência de RNA , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/metabolismo , Fatores de Transcrição de Fator Regulador X , Fatores de Transcrição/química , Proteína 1 de Ligação a X-Box
11.
PLoS One ; 9(1): e86358, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24466051

RESUMO

Ku70 and Ku80 form a heterodimer called Ku that forms a holoenzyme with DNA dependent-protein kinase catalytic subunit (DNA-PKCS) to repair DNA double strand breaks (DSBs) through the nonhomologous end joining (NHEJ) pathway. As expected mutating these genes in mice caused a similar DSB repair-defective phenotype. However, ku70(-/-) cells and ku80(-/-) cells also appeared to have a defect in base excision repair (BER). BER corrects base lesions, apurinic/apyrimidinic (AP) sites and single stand breaks (SSBs) utilizing a variety of proteins including glycosylases, AP endonuclease 1 (APE1) and DNA Polymerase ß (Pol ß). In addition, deleting Ku70 was not equivalent to deleting Ku80 in cells and mice. Therefore, we hypothesized that free Ku70 (not bound to Ku80) and/or free Ku80 (not bound to Ku70) possessed activity that influenced BER. To further test this hypothesis we performed two general sets of experiments. The first set showed that deleting either Ku70 or Ku80 caused an NHEJ-independent defect. We found ku80(-/-) mice had a shorter life span than dna-pkcs(-/-) mice demonstrating a phenotype that was greater than deleting the holoenzyme. We also found Ku70-deletion induced a p53 response that reduced the level of small mutations in the brain suggesting defective BER. We further confirmed that Ku80-deletion impaired BER via a mechanism that was not epistatic to Pol ß. The second set of experiments showed that free Ku70 and free Ku80 could influence BER. We observed that deletion of either Ku70 or Ku80, but not both, increased sensitivity of cells to CRT0044876 (CRT), an agent that interferes with APE1. In addition, free Ku70 and free Ku80 bound to AP sites and in the case of Ku70 inhibited APE1 activity. These observations support a novel role for free Ku70 and free Ku80 in altering BER.


Assuntos
Antígenos Nucleares/genética , Reparo do DNA por Junção de Extremidades , Proteínas de Ligação a DNA/genética , Animais , Ácido Apurínico/genética , DNA Liase (Sítios Apurínicos ou Apirimidínicos)/antagonistas & inibidores , Proteína Quinase Ativada por DNA/deficiência , Proteína Quinase Ativada por DNA/genética , Proteínas de Ligação a DNA/deficiência , Epistasia Genética , Feminino , Deleção de Genes , Indóis/farmacologia , Autoantígeno Ku , Longevidade , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Nucleares/deficiência , Proteínas Nucleares/genética , Fenótipo , Mutação Puntual , Subunidades Proteicas/deficiência , Subunidades Proteicas/genética , Tolerância a Radiação , Proteína Supressora de Tumor p53/metabolismo
12.
PLoS One ; 8(5): e62882, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23667535

RESUMO

The DREAM complex plays an important role in regulation of gene expression during the cell cycle. We have previously shown that the DREAM subunit LIN9 is required for early embryonic development and for the maintenance of the inner cell mass in vitro. In this study we examined the effect of knocking down LIN9 on ESCs. We demonstrate that depletion of LIN9 alters the cell cycle distribution of ESCs and results in an accumulation of cells in G2 and M and in an increase of polyploid cells. Genome-wide expression studies showed that the depletion of LIN9 results in downregulation of mitotic genes and in upregulation of differentiation-specific genes. ChIP-on chip experiments showed that mitotic genes are direct targets of LIN9 while lineage specific markers are regulated indirectly. Importantly, depletion of LIN9 does not alter the expression of pluripotency markers SOX2, OCT4 and Nanog and LIN9 depleted ESCs retain alkaline phosphatase activity. We conclude that LIN9 is essential for proliferation and genome stability of ESCs by activating genes with important functions in mitosis and cytokinesis.


Assuntos
Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Mitose/genética , Subunidades Proteicas/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Fosfatase Alcalina/metabolismo , Animais , Ciclo Celular , Diferenciação Celular , Linhagem da Célula , Proliferação de Células , DNA/metabolismo , Corpos Embrioides/citologia , Corpos Embrioides/metabolismo , Técnicas de Silenciamento de Genes , Instabilidade Genômica , Humanos , Camundongos , Poliploidia , Regiões Promotoras Genéticas/genética , Subunidades Proteicas/deficiência , Subunidades Proteicas/genética , Interferência de RNA , Proteínas Supressoras de Tumor/deficiência , Proteínas Supressoras de Tumor/genética
13.
Cell Biochem Biophys ; 66(3): 599-622, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23329175

RESUMO

Cell invasion through the extracellular matrix (ECM) of connective tissue is an important biomechanical process, which plays a prominent role in tumor progression. The malignancy of tumors depends mainly on the capacity of cancer cells to migrate and metastasize. A prerequisite for metastasis is the invasion of cancer cells through connective tissue to targeted organs. Cellular stiffness and cytoskeletal remodeling dynamics have been proposed to affect the invasiveness of cancer cells. Here, this study investigated whether highly invasive cancer cells are capable of invading into dense 3D-ECMs with an average pore-size of 1.3 or 3.0 µm when phagocytized beads (2.7 and 4.5 µm diameter) increased their cellular stiffness and reduced their cytoskeletal remodeling dynamics compared to weakly invasive cancer cells. The phagocytized beads decreased the invasiveness of the α5ß1(high) cancer cells into 3D-ECMs, whereas the invasiveness of the α5ß1(low) cancer cells was not affected. The effect of phagocytized beads on the highly invasive α5ß1(high) cells was abolished by specific knock-down of the α5 integrin subunit or addition of an anti-α5 integrin blocking antibody. Furthermore, the reduction of contractile forces using MLCK and ROCK inhibitors abolished the effect of phagocytized beads on the invasiveness of α5ß1(high) cells. In addition, the cellular stiffness of α5ß1(high) cells was increased after bead phagocytosis, whereas the bead phagocytosis did not alter the stiffness of α5ß1(low) cells. Taken together, the α5ß1 integrin dependent invasiveness was reduced after bead phagocytosis by altered biomechanical properties, suggesting that the α5ß1(high) cells need an appropriate intermediate cellular stiffness to overcome the steric hindrance of 3D-ECMs, whereas the α5ß1(low) cells were not affected by phagocytized beads.


Assuntos
Integrina alfa5beta1/metabolismo , Fenômenos Mecânicos , Microesferas , Neoplasias/patologia , Fagocitose , Anticorpos Neutralizantes/imunologia , Fenômenos Biomecânicos , Adesão Celular , Linhagem Celular Tumoral , Movimento Celular , Colágeno/metabolismo , Citoesqueleto/metabolismo , Matriz Extracelular/metabolismo , Fibronectinas/imunologia , Fibronectinas/metabolismo , Regulação Neoplásica da Expressão Gênica , Técnicas de Silenciamento de Genes , Humanos , Integrina alfa5beta1/imunologia , Invasividade Neoplásica , Subunidades Proteicas/deficiência , Subunidades Proteicas/genética
14.
Dis Model Mech ; 6(2): 434-42, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23065636

RESUMO

Mutations in the ATP6V0A4 gene lead to autosomal recessive distal renal tubular acidosis in patients, who often show sensorineural hearing impairment. A first Atp6v0a4 knockout mouse model that recapitulates the loss of H(+)-ATPase function seen in humans has been generated and recently reported (Norgett et al., 2012). Here, we present the first detailed analysis of the structure and function of the auditory system in Atp6v0a4(-/-) knockout mice. Measurements of the auditory brainstem response (ABR) showed significantly elevated thresholds in homozygous mutant mice, which indicate severe hearing impairment. Heterozygote thresholds were normal. Analysis of paint-filled inner ears and sections from E16.5 embryos revealed a marked expansion of cochlear and endolymphatic ducts in Atp6v0a4(-/-) mice. A regulatory link between Atp6v0a4, Foxi1 and Pds has been reported and we found that the endolymphatic sac of Atp6v0a4(-/-) mice expresses both Foxi1 and Pds, which suggests a downstream position of Atp6v0a4. These mutants also showed a lack of endocochlear potential, suggesting a functional defect of the stria vascularis on the lateral wall of the cochlear duct. However, the main K(+) channels involved in the generation of endocochlear potential, Kcnj10 and Kcnq1, are strongly expressed in Atp6v0a4(-/-) mice. Our results lead to a better understanding of the role of this proton pump in hearing function.


Assuntos
Orelha Interna/enzimologia , Orelha Interna/patologia , Endolinfa/enzimologia , Perda Auditiva/enzimologia , Perda Auditiva/patologia , Subunidades Proteicas/deficiência , ATPases Translocadoras de Prótons/deficiência , Animais , Animais Recém-Nascidos , Proteínas de Transporte de Ânions/metabolismo , Orelha Interna/fisiopatologia , Saco Endolinfático/patologia , Saco Endolinfático/fisiopatologia , Epitélio/metabolismo , Epitélio/patologia , Potenciais Evocados Auditivos , Fatores de Transcrição Forkhead/metabolismo , Células Ciliadas Auditivas Externas/metabolismo , Células Ciliadas Auditivas Externas/patologia , Células Ciliadas Auditivas Externas/ultraestrutura , Perda Auditiva/fisiopatologia , Humanos , Canal de Potássio KCNQ1/metabolismo , Camundongos , Camundongos Knockout , Mutação/genética , Fenótipo , Canais de Potássio Corretores do Fluxo de Internalização/metabolismo , Subunidades Proteicas/metabolismo , ATPases Translocadoras de Prótons/metabolismo , Estria Vascular/metabolismo , Estria Vascular/patologia , Transportadores de Sulfato , ATPases Vacuolares Próton-Translocadoras
15.
Fundam Clin Pharmacol ; 27(1): 96-103, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23033963

RESUMO

Alzheimer disease (AD) is a neurodegenerative disorder clinically characterized by progressive cognitive and memory dysfunction, which is the most common form of dementia. Although the pathogenesis of neuronal injury in AD is not clear, recent evidences suggest that Na⁺-K⁺-ATPase plays an important role in AD, and may be a potent neuroprotective modulator against AD. This review aims to provide readers with an in-depth understanding of Na⁺-K⁺-ATPase in AD through these modulations of some factors that are as follows, which leads to the change of learning and memory in the process of AD. 1. The deficiency in Na⁺, K⁺-ATPase α1, α2 and α3 isoform genes induced learning and memory deficits, and α isoform was evidently changed in AD, revealing that Na⁺, K⁺-ATPase α isoform genes may play an important role in AD. 2. Some factors, such as ß-amyloid, cholinergic and oxidative stress, can modulate learning and memory in AD through the mondulation of Na⁺-K⁺-ATPase activity. 3. Some substances, such as Zn, s-Ethyl cysteine, s-propyl cysteine, citicoline, rivastigmine, Vit E, memantine, tea polyphenol, curcumin, caffeine, Alpinia galanga (L.) fractions, and Bacopa monnieri could play a role in improving memory performance and exert protective effects against AD by increasing expression or activity of Na⁺, K⁺-ATPase.


Assuntos
Doença de Alzheimer/metabolismo , Encéfalo/enzimologia , Neurônios/enzimologia , ATPase Trocadora de Sódio-Potássio/metabolismo , Doença de Alzheimer/etiologia , Doença de Alzheimer/prevenção & controle , Doença de Alzheimer/terapia , Animais , Encéfalo/metabolismo , Suplementos Nutricionais , Ativação Enzimática/efeitos dos fármacos , Indução Enzimática/efeitos dos fármacos , Humanos , Isoenzimas/química , Isoenzimas/deficiência , Isoenzimas/metabolismo , Neurônios/metabolismo , Fármacos Neuroprotetores/uso terapêutico , Nootrópicos/uso terapêutico , Estresse Oxidativo/efeitos dos fármacos , Subunidades Proteicas/agonistas , Subunidades Proteicas/antagonistas & inibidores , Subunidades Proteicas/deficiência , Subunidades Proteicas/metabolismo , ATPase Trocadora de Sódio-Potássio/antagonistas & inibidores , ATPase Trocadora de Sódio-Potássio/química , ATPase Trocadora de Sódio-Potássio/deficiência
16.
Am J Physiol Endocrinol Metab ; 304(4): E392-404, 2013 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-23233541

RESUMO

The consumption of high-fat/calorie diets in modern societies is likely a major contributor to the obesity epidemic, which can increase the prevalence of cancer, cardiovascular disease, and neurological impairment. Obesity may precipitate decline via inflammatory and oxidative signaling, and one factor linking inflammation to oxidative stress is the proinflammatory, pro-oxidant enzyme NADPH oxidase. To reveal the role of NADPH oxidase in the metabolic and neurological consequences of obesity, the effects of high-fat diet were compared in wild-type C57Bl/6 (WT) mice and in mice deficient in the NAPDH oxidase subunit NOX2 (NOX2KO). While diet-induced weight gains in WT and NOX2KO mice were similar, NOX2KO mice had smaller visceral adipose deposits, attenuated visceral adipocyte hypertrophy, and diminished visceral adipose macrophage infiltration. Moreover, the detrimental effects of HFD on markers of adipocyte function and injury were attenuated in NOX2KO mice; NOX2KO mice had improved glucose regulation, and evaluation of NOX2 expression identified macrophages as the primary population of NOX2-positive cells in visceral adipose. Finally, brain injury was assessed using markers of cerebrovascular integrity, synaptic density, and reactive gliosis, and data show that high-fat diet disrupted marker expression in WT but not NOX2KO mice. Collectively, these data indicate that NOX2 is a significant contributor to the pathogenic effects of high-fat diet and reinforce a key role for visceral adipose inflammation in metabolic and neurological decline. Development of NOX-based therapies could accordingly preserve metabolic and neurological function in the context of metabolic syndrome.


Assuntos
Adiposidade , Encéfalo/metabolismo , Dieta Hiperlipídica/efeitos adversos , Glicoproteínas de Membrana/deficiência , NADPH Oxidases/deficiência , Neurônios/metabolismo , Obesidade/metabolismo , Subunidades Proteicas/deficiência , Animais , Biomarcadores/metabolismo , Encéfalo/imunologia , Encéfalo/patologia , Hipertrofia , Gordura Intra-Abdominal/imunologia , Gordura Intra-Abdominal/patologia , Macrófagos/imunologia , Macrófagos/metabolismo , Macrófagos/patologia , Masculino , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Síndrome Metabólica/etiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , NADPH Oxidase 2 , NADPH Oxidases/genética , NADPH Oxidases/metabolismo , Proteínas do Tecido Nervoso/deficiência , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Neurônios/imunologia , Neurônios/patologia , Obesidade/imunologia , Obesidade/patologia , Obesidade/fisiopatologia , Estresse Oxidativo , Subunidades Proteicas/genética , Subunidades Proteicas/metabolismo , Aumento de Peso
17.
J Immunol ; 189(8): 4182-93, 2012 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-22984077

RESUMO

The immunoproteasome generates peptides presented on MHC class I molecules to cytotoxic T cells. ONX 0914 (formerly called PR-957) is a selective inhibitor of the immunoproteasome subunit low molecular mass polypeptide (LMP) 7 (ß5i) that attenuates disease progression in mouse models of diabetes, colitis, and arthritis. The aim of this study was to investigate the effect of LMP7-specific inhibition on major Th cell differentiation pathways involved in the progression of autoimmune diseases in vitro and in vivo. We used ONX 0914-treated wild-type CD4(+) T cells and also LMP7(-/-) CD4(+) T cells under different Th cell-polarizing conditions, focusing on the effector cytokines and transcription factors involved, and compared them with wild-type CD4(+) T cells. Mouse models of dextran sodium sulfate-induced colitis and a T cell transfer model of colitis were used for in vivo assessment. Deletion or inhibition of LMP7 suppressed generation of Th17 but promoted regulatory T cell (Treg) development. In developing Th17 cells, immunoproteasome inhibition blocked phosphorylation of STAT3, whereas in Tregs, SMAD phosphorylation was enhanced. Additionally, LMP7 inhibition led to reduced STAT1 phosphorylation and Th1 differentiation. These findings were confirmed in vivo as LMP7 inhibition or deficiency resulted in reduced Th1 and Th17 expansion while promoting Treg development in dextran sodium sulfate-induced colitis. Also, in a T cell-dependent transfer model of colitis, LMP7-specific inhibition led to reduced Th1 and Th17 differentiation in vivo. LMP7 governs Th cell lineage determination by affecting the balance of receptor proximal signals during differentiation. These data render LMP7 a promising drug target for the treatment of autoimmune diseases.


Assuntos
Diferenciação Celular/imunologia , Regulação para Baixo/imunologia , Complexo de Endopeptidases do Proteassoma/deficiência , Linfócitos T Reguladores/imunologia , Células Th1/imunologia , Células Th17/imunologia , Regulação para Cima/imunologia , Animais , Diferenciação Celular/genética , Regulação para Baixo/genética , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Complexo de Endopeptidases do Proteassoma/genética , Complexo de Endopeptidases do Proteassoma/metabolismo , Subunidades Proteicas/deficiência , Subunidades Proteicas/genética , Subunidades Proteicas/metabolismo , Linfócitos T Reguladores/citologia , Linfócitos T Reguladores/metabolismo , Células Th1/citologia , Células Th1/metabolismo , Células Th17/citologia , Células Th17/metabolismo , Regulação para Cima/genética
18.
J Immunol ; 189(2): 860-6, 2012 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-22685314

RESUMO

Nucleated cells are equipped with several mechanisms that support their resistance to complement-dependent cytotoxicity (CDC). The role of the NF-κB pathway in cell protection from CDC was examined. Elevated sensitivity to CDC was demonstrated in cells lacking the p65 subunit of NF-κB or the IκB kinases IKKα or IKKß, and in cells treated with p65 small interfering RNA. Pretreatment with the IKK inhibitor PS-1145 also enhanced CDC of wild-type cells (WT) but not of p65(-/-) cells. Furthermore, reconstitution of p65 into p65(-/-) cells and overexpression of p65 in WT cells lowered their sensitivity to CDC. The postulated effect of p65 on the JNK-mediated death-signaling pathway activated by complement was examined. p65 small interfering RNA enhanced CDC in WT cells but not in cells lacking JNK. JNK phosphorylation induced by complement was more pronounced in p65(-/-) cells than in WT cells. The results indicate that the NF-κB pathway mediates cell resistance to CDC, possibly by suppressing JNK-dependent programmed necrotic cell death.


Assuntos
Ativação do Complemento/imunologia , Complexo de Ataque à Membrana do Sistema Complemento/fisiologia , Citotoxicidade Imunológica/imunologia , Transdução de Sinais/imunologia , Fator de Transcrição RelA/fisiologia , Animais , Comunicação Celular/genética , Comunicação Celular/imunologia , Ativação do Complemento/genética , Complexo de Ataque à Membrana do Sistema Complemento/deficiência , Complexo de Ataque à Membrana do Sistema Complemento/metabolismo , Citotoxicidade Imunológica/genética , Células-Tronco Embrionárias/enzimologia , Células-Tronco Embrionárias/imunologia , Células-Tronco Embrionárias/metabolismo , Fibroblastos/enzimologia , Fibroblastos/imunologia , Fibroblastos/metabolismo , Células HEK293 , Células HeLa , Humanos , Quinase I-kappa B/deficiência , MAP Quinase Quinase 4/antagonistas & inibidores , MAP Quinase Quinase 4/metabolismo , MAP Quinase Quinase 4/fisiologia , Camundongos , Camundongos Knockout , Subunidades Proteicas/deficiência , Transdução de Sinais/genética , Fator de Transcrição RelA/deficiência , Fator de Transcrição RelA/metabolismo
19.
EMBO J ; 31(9): 2076-89, 2012 May 02.
Artigo em Inglês | MEDLINE | ID: mdl-22415365

RESUMO

Cohesin is a protein complex originally identified for its role in sister chromatid cohesion, although increasing evidence portrays it also as a major organizer of interphase chromatin. Vertebrate cohesin consists of Smc1, Smc3, Rad21/Scc1 and either stromal antigen 1 (SA1) or SA2. To explore the functional specificity of these two versions of cohesin and their relevance for embryonic development and cancer, we generated a mouse model deficient for SA1. Complete ablation of SA1 results in embryonic lethality, while heterozygous animals have shorter lifespan and earlier onset of tumourigenesis. SA1-null mouse embryonic fibroblasts show decreased proliferation and increased aneuploidy as a result of chromosome segregation defects. These defects are not caused by impaired centromeric cohesion, which depends on cohesin-SA2. Instead, they arise from defective telomere replication, which requires cohesion mediated specifically by cohesin-SA1. We propose a novel mechanism for aneuploidy generation that involves impaired telomere replication upon loss of cohesin-SA1, with clear implications in tumourigenesis.


Assuntos
Aneuploidia , Proteínas de Ciclo Celular/deficiência , Proteínas Cromossômicas não Histona/deficiência , Subunidades Proteicas/deficiência , Telômero/metabolismo , Animais , Carcinógenos , Proteínas de Ciclo Celular/genética , Linhagem Celular , Cromátides/metabolismo , Proteínas Cromossômicas não Histona/genética , Segregação de Cromossomos , Dietilnitrosamina , Fibrossarcoma/induzido quimicamente , Fibrossarcoma/genética , Fibrossarcoma/patologia , Neoplasias Hepáticas/induzido quimicamente , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patologia , Masculino , Metilcolantreno , Camundongos , Camundongos Knockout , Neoplasias Experimentais/induzido quimicamente , Neoplasias Experimentais/genética , Neoplasias Experimentais/patologia , Subunidades Proteicas/genética , Troca de Cromátide Irmã , Coesinas
20.
EMBO J ; 31(9): 2090-102, 2012 May 02.
Artigo em Inglês | MEDLINE | ID: mdl-22415368

RESUMO

Vertebrates have two cohesin complexes that consist of Smc1, Smc3, Rad21/Scc1 and either SA1 or SA2, but their functional specificity is unclear. Mouse embryos lacking SA1 show developmental delay and die before birth. Comparison of the genome-wide distribution of cohesin in wild-type and SA1-null cells reveals that SA1 is largely responsible for cohesin accumulation at promoters and at sites bound by the insulator protein CTCF. As a consequence, ablation of SA1 alters transcription of genes involved in biological processes related to Cornelia de Lange syndrome (CdLS), a genetic disorder linked to dysfunction of cohesin. We show that the presence of cohesin-SA1 at the promoter of myc and of protocadherin genes positively regulates their expression, a task that cannot be assumed by cohesin-SA2. Lack of SA1 also alters cohesin-binding pattern along some gene clusters and leads to dysregulation of genes within. We hypothesize that impaired cohesin-SA1 function in gene expression underlies the molecular aetiology of CdLS.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Proteínas Cromossômicas não Histona/metabolismo , Desenvolvimento Embrionário , Regulação da Expressão Gênica , Subunidades Proteicas/metabolismo , Animais , Proteínas de Ciclo Celular/deficiência , Proteínas de Ciclo Celular/genética , Proteínas Cromossômicas não Histona/deficiência , Proteínas Cromossômicas não Histona/genética , Síndrome de Cornélia de Lange/genética , Embrião de Mamíferos , Fibroblastos , Camundongos , Camundongos Knockout , Subunidades Proteicas/deficiência , Subunidades Proteicas/genética , Proteínas Proto-Oncogênicas c-myc/genética , Coesinas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA