Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Forensic Sci Int Genet ; 68: 102942, 2024 01.
Artigo em Inglês | MEDLINE | ID: mdl-37862769

RESUMO

If a DNA sample collected in the field is old or degraded, short tandem repeat analysis is difficult to perform, a representative analysis method currently used for individual identification. Given that microorganisms exist everywhere and within the human body, in similar amounts to human cells, microbial analysis could be used to identify individuals even in cases in which human DNA-based identification is difficult. Research has demonstrated that the types of microorganisms within the human body differ depending on various internal or external factors, such as body part or bodily fluid type, lifestyle, geographical area of residence, sex, and age. In this study, we aimed to examine the relationship between lifestyle factors and the composition and diversity of the oral microbiome in individuals living in Korea. We collected 43 saliva samples from Korean individuals and analyzed the oral microbiome and its variations due to external factors, such as coffee consumption, drinking, and smoking. Linear discriminant analysis effect size revealed that Oribacterium, Campylobacter, and Megasphaera were abundant in coffee consumers, whereas Saccharimonadales, Clostridia, and Catonella were abundant in alcohol non-drinkers. We found increased levels of Stomatobaculum in the saliva of smokers, compared with that of non-smokers. Thus, our analysis revealed characteristic microorganisms for each parameter that was evaluated (coffee consumption, smoking, drinking). Consequently, our study provides insight into the oral microbiome in the Korean population and lays the foundation for developing the Korean Forensic Microbiome Database.


Assuntos
Café , Microbiota , Humanos , Fumar/epidemiologia , Estilo de Vida , DNA , República da Coreia
2.
Mucosal Immunol ; 13(2): 216-229, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31772323

RESUMO

Mononuclear phagocytes are a heterogeneous population of leukocytes essential for immune homeostasis that develop tissue-specific functions due to unique transcriptional programs driven by local microenvironmental cues. Single cell RNA sequencing (scRNA-seq) of colonic myeloid cells from specific pathogen free (SPF) and germ-free (GF) C57BL/6 mice revealed extensive heterogeneity of both colon macrophages (MPs) and dendritic cells (DCs). Modeling of developmental pathways combined with inference of gene regulatory networks indicate two major trajectories from common CCR2+ precursors resulting in colon MP populations with unique transcription factors and downstream target genes. Compared to SPF mice, GF mice had decreased numbers of total colon MPs, as well as selective proportional decreases of two major CD11c+CD206intCD121b+ and CD11c-CD206hiCD121b- colon MP populations, whereas DC numbers and proportions were not different. Importantly, these two major colon MP populations were clearly distinct from other colon MP populations regarding their gene expression profile, localization within the lamina propria (LP) and ability to phagocytose macromolecules from the blood. These data uncover the diversity of intestinal myeloid cell populations at the molecular level and highlight the importance of microbiota on the unique developmental as well as anatomical and functional fates of colon MPs.


Assuntos
Colo/imunologia , Células Dendríticas/imunologia , Macrófagos/imunologia , Células Mieloides/fisiologia , Animais , Antígeno CD11c/metabolismo , Diferenciação Celular , Células Cultivadas , Ontologia Genética , Redes Reguladoras de Genes , Homeostase , Lectinas Tipo C/genética , Lectinas Tipo C/metabolismo , Receptor de Manose , Lectinas de Ligação a Manose/genética , Lectinas de Ligação a Manose/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fagocitose/genética , Fenótipo , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/metabolismo , Análise de Sequência de RNA , Análise de Célula Única , Transcriptoma
3.
Cell Death Dis ; 10(12): 938, 2019 12 09.
Artigo em Inglês | MEDLINE | ID: mdl-31819032

RESUMO

Infection of hepatitis B virus (HBV) increase the incidence of chronic liver disease and hepatocellular carcinoma (HCC). The hepatitis B viral x (HBx) protein encoded by the HBV genome contributes to the pathogenesis of HCC and thus, negative regulation of HBx is beneficial for the alleviation of the disease pathogenesis. MARCH5 is a mitochondrial E3 ubiquitin ligase and here, we show that high MARCH5 expression levels are correlated with improved survival in HCC patients. MARCH5 interacts with HBx protein mainly accumulated in mitochondria and targets it for degradation. The N-terminal RING domain of MARCH5 was required for the interaction with HBx, and MARCH5H43W lacking E3 ligase activity failed to reduce HBx protein levels. High expression of HBx results in the formation of protein aggregates in semi-denaturing detergent agarose gels and MARCH5 mediates the elimination of protein aggregates through the proteasome pathway. HBx-induced ROS production, mitophagy, and cyclooxygenase-2 gene expression were suppressed in the presence of high MARCH5 expression. These results suggest MARCH5 as a target for alleviating HBV-mediated liver disease.


Assuntos
Carcinoma Hepatocelular/metabolismo , Vírus da Hepatite B/química , Hepatite B/metabolismo , Neoplasias Hepáticas/metabolismo , Proteínas de Membrana/metabolismo , Agregados Proteicos , Proteólise , Transativadores/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Proteínas Virais Reguladoras e Acessórias/metabolismo , Carcinoma Hepatocelular/etiologia , Carcinoma Hepatocelular/mortalidade , Carcinoma Hepatocelular/patologia , Células HEK293 , Células HeLa , Hepatite B/complicações , Hepatite B/virologia , Humanos , Neoplasias Hepáticas/etiologia , Neoplasias Hepáticas/mortalidade , Neoplasias Hepáticas/patologia , Proteínas de Membrana/genética , Mitocôndrias/metabolismo , Agregação Patológica de Proteínas/metabolismo , Taxa de Sobrevida , Transfecção , Ubiquitina-Proteína Ligases/genética
4.
J Biol Chem ; 289(15): 10738-10747, 2014 Apr 11.
Artigo em Inglês | MEDLINE | ID: mdl-24573680

RESUMO

Activator of G-protein signaling 3 (AGS3, gene name G-protein signaling modulator-1, Gpsm1), an accessory protein for G-protein signaling, has functional roles in the kidney and CNS. Here we show that AGS3 is expressed in spleen, thymus, and bone marrow-derived dendritic cells, and is up-regulated upon leukocyte activation. We explored the role of AGS3 in immune cell function by characterizing chemokine receptor signaling in leukocytes from mice lacking AGS3. No obvious differences in lymphocyte subsets were observed. Interestingly, however, AGS3-null B and T lymphocytes and bone marrow-derived dendritic cells exhibited significant chemotactic defects as well as reductions in chemokine-stimulated calcium mobilization and altered ERK and Akt activation. These studies indicate a role for AGS3 in the regulation of G-protein signaling in the immune system, providing unexpected venues for the potential development of therapeutic agents that modulate immune function by targeting these regulatory mechanisms.


Assuntos
Proteínas de Transporte/metabolismo , Quimiocinas/metabolismo , Leucócitos/metabolismo , Transdução de Sinais , Motivos de Aminoácidos , Animais , Linfócitos B/citologia , Células da Medula Óssea/citologia , Cálcio/metabolismo , Quimiotaxia , Células Dendríticas/citologia , Feminino , Proteínas de Ligação ao GTP/metabolismo , Inibidores de Dissociação do Nucleotídeo Guanina , Sistema Imunitário , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Baço/citologia , Linfócitos T/citologia , Timócitos/citologia
5.
PLoS One ; 9(1): e86680, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24466196

RESUMO

Resistance to inhibitors of cholinesterase (Ric)-8A is a guanine nucleotide exchange factor for Gαi, Gαq, and Gα12/13, which is implicated in cell signaling and as a molecular chaperone required for the initial association of nascent Gα subunits with cellular membranes. Ric-8A, Gαi subunits, and their regulators are localized at the midbody prior to abscission and linked to the final stages of cell division. Here, we identify a molecular mechanism by which Ric-8A affects cytokinesis and abscission by controlling Vps34 activity. We showed that Ric-8A protein expression is post-transcriptionally controlled during the cell cycle reaching its maximum levels at mitosis. A FRET biosensor created to measure conformational changes in Ric-8A by FLIM (Fluorescence Lifetime Imaging Microscopy) revealed that Ric-8A was in a close-state during mitosis and particularly so at cytokinesis. Lowering Ric-8A expression delayed the abscission time of dividing cells, which correlated with increased intercellular bridge length and multinucleation. During cytokinesis, Ric-8A co-localized with Vps34 at the midbody along with Gαi and LGN, where these proteins functioned to regulate Vps34 phosphatidylinositol 3-kinase activity.


Assuntos
Classe III de Fosfatidilinositol 3-Quinases/metabolismo , Citocinese/fisiologia , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Linhagem Celular Tumoral , Células HeLa , Humanos , Mitose/fisiologia , Proteínas Nucleares/metabolismo , Processamento Pós-Transcricional do RNA/fisiologia
6.
Mol Cell Biol ; 32(22): 4561-71, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22966200

RESUMO

Neutrophils are first responders rapidly mobilized to inflammatory sites by a tightly regulated, nonredundant hierarchy of chemoattractants. These chemoattractants engage neutrophil cell surface receptors triggering heterotrimeric G-protein Gα(i) subunits to exchange GDP for GTP. By limiting the duration that Gα(i) subunits remain GTP bound, RGS proteins modulate chemoattractant receptor signaling. Here, we show that neutrophils with a genomic knock in of a mutation that disables regulator of G-protein signaling (RGS)-Gα(i2) interactions accumulate in the bone marrow and mobilize poorly to inflammatory sites. These defects are attributable to enhanced sensitivity to background signals, prolonged chemoattractant receptor signaling, and inappropriate CXCR2 downregulation. Intravital imaging revealed a failure of the mutant neutrophils to accumulate at and stabilize sites of sterile inflammation. Furthermore, these mice could not control a nonlethal Staphylococcus aureus infection. Neutrophil RGS proteins establish a threshold for Gα(i) activation, helping to coordinate desensitization mechanisms. Their loss renders neutrophils functionally incompetent.


Assuntos
Quimiotaxia/genética , Subunidade alfa Gi2 de Proteína de Ligação ao GTP/metabolismo , Inflamação/enzimologia , Neutrófilos/metabolismo , Proteínas RGS/metabolismo , Infecções Estafilocócicas/enzimologia , Animais , Medula Óssea/imunologia , Medula Óssea/patologia , Quimiotaxia/imunologia , Regulação para Baixo , Ativação Enzimática , Subunidade alfa Gi2 de Proteína de Ligação ao GTP/genética , Subunidade alfa Gi2 de Proteína de Ligação ao GTP/imunologia , Técnicas de Introdução de Genes , Guanosina Difosfato/metabolismo , Guanosina Trifosfato/metabolismo , Inflamação/complicações , Inflamação/imunologia , Inflamação/microbiologia , Camundongos , Camundongos Transgênicos , Mutação , Neutrófilos/imunologia , Neutrófilos/patologia , Proteínas RGS/genética , Proteínas RGS/imunologia , Receptores de Interleucina-8B/genética , Receptores de Interleucina-8B/imunologia , Transdução de Sinais/genética , Transdução de Sinais/imunologia , Infecções Estafilocócicas/complicações , Infecções Estafilocócicas/imunologia , Infecções Estafilocócicas/microbiologia , Staphylococcus aureus/fisiologia , Fatores de Tempo
7.
J Biol Chem ; 286(35): 30615-30623, 2011 Sep 02.
Artigo em Inglês | MEDLINE | ID: mdl-21768102

RESUMO

The E3 ubiquitin-protein ligase Chfr is a mitotic stress checkpoint protein that delays mitotic entry in response to microtubule damage; however, the molecular mechanism by which Chfr accomplishes this remains elusive. Here, we show that Chfr levels are elevated in response to microtubule-damaging stress. Moreover, G(2)/M transition is associated with cell cycle-dependent turnover of Chfr accompanied by high autoubiquitylation activity, suggesting that regulation of Chfr levels and auto-ubiquitylation activity are functionally significant. To test this, we generated Chfr mutants Chfr-K2A and Chfr-K5A in which putative lysine target sites of auto-ubiquitylation were replaced with alanine. Chfr-K2A did not undergo cell cycle-dependent degradation, and its levels remained high during G(2)/M phase. The elevated levels of Chfr-K2A caused a significant reduction in phosphohistone H3 levels and cyclinB1/Cdk1 kinase activities, leading to mitotic entry delay. Notably, polo-like kinase 1 levels at G(2) phase, but not at S phase, were ∼2-3-fold lower in cells expressing Chfr-K2A than in wild-type Chfr-expressing cells. Consistent with this, ubiquitylation of Plk1 at G(2) phase was accelerated in Chfr-K2A-expressing cells. In contrast, Aurora A levels remained constant, indicating that Plk1 is a major target of Chfr in controlling the timing of mitotic entry. Indeed, overexpression of Plk1 in Chfr-K2A-expressing cells restored cyclin B1/Cdk1 kinase activity and promoted mitotic entry. Collectively, these data indicate that Chfr auto-ubiquitylation is required to allow Plk1 to accumulate to levels necessary for activation of cyclin B1/Cdk1 kinase and mitotic entry. Our results provide the first evidence that Chfr auto-ubiquitylation and degradation are important for the G(2)/M transition.


Assuntos
Proteínas de Ciclo Celular/química , Proteínas de Ciclo Celular/metabolismo , Mitose , Proteínas de Neoplasias/química , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Ubiquitina-Proteína Ligases/química , Ubiquitina/química , Alanina/química , Animais , Sítios de Ligação , Proteína Quinase CDC2/metabolismo , Ciclina B1/metabolismo , Citometria de Fluxo/métodos , Fase G2 , Células HeLa , Humanos , Lisina/química , Modelos Biológicos , Proteínas de Ligação a Poli-ADP-Ribose , Conformação Proteica , Quinase 1 Polo-Like
8.
Mol Cell Biol ; 30(14): 3519-30, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20479129

RESUMO

In model organisms, resistance to inhibitors of cholinesterase 8 (Ric-8), a G protein alpha (G alpha) subunit guanine nucleotide exchange factor (GEF), functions to orient mitotic spindles during asymmetric cell divisions; however, whether Ric-8A has any role in mammalian cell division is unknown. We show here that Ric-8A and G alpha(i) function to orient the metaphase mitotic spindle of mammalian adherent cells. During mitosis, Ric-8A localized at the cell cortex, spindle poles, centromeres, central spindle, and midbody. Pertussis toxin proved to be a useful tool in these studies since it blocked the binding of Ric-8A to G alpha(i), thus preventing its GEF activity for G alpha(i). Linking Ric-8A signaling to mammalian cell division, treatment of cells with pertussis toxin, reduction of Ric-8A expression, or decreased G alpha(i) expression similarly affected metaphase cells. Each treatment impaired the localization of LGN (GSPM2), NuMA (microtubule binding nuclear mitotic apparatus protein), and dynein at the metaphase cell cortex and disturbed integrin-dependent mitotic spindle orientation. Live cell imaging of HeLa cells expressing green fluorescent protein-tubulin also revealed that reduced Ric-8A expression prolonged mitosis, caused occasional mitotic arrest, and decreased mitotic spindle movements. These data indicate that Ric-8A signaling leads to assembly of a cortical signaling complex that functions to orient the mitotic spindle.


Assuntos
Antígenos Nucleares/metabolismo , Dineínas/metabolismo , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/metabolismo , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas Associadas à Matriz Nuclear/metabolismo , Fuso Acromático/metabolismo , Animais , Sequência de Bases , Proteínas de Ciclo Celular , Divisão Celular/fisiologia , Linhagem Celular , Cães , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/antagonistas & inibidores , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/genética , Fatores de Troca do Nucleotídeo Guanina/antagonistas & inibidores , Fatores de Troca do Nucleotídeo Guanina/genética , Células HeLa , Humanos , Interfase/fisiologia , Metáfase/fisiologia , Toxina Pertussis/farmacologia , Ligação Proteica/efeitos dos fármacos , RNA Interferente Pequeno/genética , Transdução de Sinais , Fuso Acromático/efeitos dos fármacos
9.
J Mol Med (Berl) ; 88(4): 359-69, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19940973

RESUMO

Chronic inflammation can be a major risk factor for cancer development and may contribute to the high worldwide incidence of hepatocellular carcinoma (HCC). Cyclooxygenase-2 (COX-2) is known to be an important mediator of inflammatory responses; however, its link to hepatitis B virus (HBV)-mediated inflammatory responses has not been established. Here, we demonstrate that the expression of COX-2 mRNA and protein was significantly elevated in cells transfected by HBV replicon but not in cells transfected by HBV genome lacking the HBx gene. Notably, COX-2 induction was correlated with HBx's ability to increase reactive oxygen species (ROS) levels. Consistently with this, antioxidant treatment and ectopic expression of manganese superoxide dismutase or catalase completely abolished COX-2 induction. Interestingly, a mitochondria localization-defective mutant of HBx (HBx(Delta 68-117)) neither increased intracellular ROS levels nor induced COX-2 expression. HBx(68-117), which encodes only amino acids 68-117 and is sufficient for mitochondria localization, increased ROS levels but did not induce COX-2 expression. Similarly, HBx targeting to the outer membrane of mitochondria (Mito-HBx) increased ROS but also failed to increase COX-2 expression, suggesting that other cytoplasmic signaling pathways are involved in HBx-mediated COX-2 induction. Indeed, inhibition of cytoplasmic calcium signaling by cyclosporine A, blocking mitochondrial permeability transition pore, and herbimycin, and inhibition of calcium-dependent tyrosine kinase suppressed HBV-mediated COX-2 induction. Thus, the data indicate that both mitochondrial ROS and cytoplasmic calcium signaling are necessary for the COX-2 induction. Our studies revealed a pathophysiological link between HBV infection and hepatic inflammation, and this chain of events might contribute to early steps in HBV-associated liver carcinogenesis.


Assuntos
Ciclo-Oxigenase 2/biossíntese , Vírus da Hepatite B/metabolismo , Mitocôndrias/metabolismo , Transativadores/metabolismo , Linhagem Celular , Linhagem Celular Tumoral , Citoplasma/metabolismo , Citometria de Fluxo/métodos , Humanos , Inflamação , Plasmídeos/metabolismo , Espécies Reativas de Oxigênio , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais , Frações Subcelulares , Proteínas Virais Reguladoras e Acessórias
10.
Cell Cycle ; 8(11): 1754-64, 2009 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-19411850

RESUMO

The role of BubR1 has been established mainly in mitosis as an essential mitotic checkpoint protein although it is expressed throughout the cell cycle. To explore a possible role of BubR1 in regulating the G(2) phase of cell cycle, we have employed siRNA-mediated hBubR1 knockdown in HeLa cells. Here, we demonstrate that reducing BubR1 levels during the G(2) phase causes accelerated mitotic entry. As expected, BubR1 depletion leads to degradation of cyclin B(1) in the G(2) phase. Intriguingly, cyclin B(1) is prematurely targeted to centrosomes appearing at early G(2) phase in BubR1-depleted cells despite its low levels. This is in contrast to control cells where cyclin B(1) appears at the centrosomes in early prophase based on cell cycle-specific localization of CENP-F. Furthermore, cyclin B/Cdk1 kinase activity in early G(2) is aberrantly high in BubR1-depleted cells. Together, our results indicate that hBubR1 depletion triggers premature centrosomal localization of cyclin B(1) probably leading to premature mitotic entry. This study is the first to suggest a role of hBubR1 in controlling centrosome targeting of cyclin B(1) and timing of mitotic entry.


Assuntos
Centrossomo/fisiologia , Ciclina B/metabolismo , Mitose , Proteínas Serina-Treonina Quinases/metabolismo , Proteína Quinase CDC2/metabolismo , Linhagem Celular Tumoral , Proteínas Cromossômicas não Histona/metabolismo , Ciclina B1 , Fase G2 , Técnicas de Silenciamento de Genes , Células HeLa , Humanos , Proteínas dos Microfilamentos/metabolismo , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/fisiologia , RNA Interferente Pequeno/metabolismo
11.
Immunity ; 22(3): 343-54, 2005 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15780991

RESUMO

Signaling by G protein-coupled receptors coupled to Galpha(i) assists in triggering lymphocyte movement into and out of lymph nodes. Here, we show that modulating the signaling output from these receptors dramatically alters B cell trafficking. Intravital microscopy of adoptively transferred B cells from wild-type and Rgs1-/- mice revealed that Rgs1-/- B cells stick better to lymph node high endothelial venules, home better to lymph nodes, and move more rapidly within lymph node follicles than do wild-type B cells. In contrast, B cells from Gnai2-/- mice enter lymph nodes poorly and move more slowly than do wild-type B cells. The Gnai2-/- mice often lack multiple peripheral lymph nodes, and their B cells respond poorly to chemokines, indicating that Galpha(i1) and Galpha(i3) poorly compensate for the loss of Galpha(i2). These results demonstrate opposing roles for Rgs1 and Gnai2 in B cell trafficking into and within lymph nodes.


Assuntos
Linfócitos B/imunologia , Quimiotaxia de Leucócito/imunologia , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/imunologia , Linfonodos/imunologia , Proteínas Proto-Oncogênicas/imunologia , Proteínas RGS/imunologia , Animais , Feminino , Citometria de Fluxo , Subunidade alfa Gi2 de Proteína de Ligação ao GTP , Processamento de Imagem Assistida por Computador , Immunoblotting , Camundongos
12.
J Biol Chem ; 280(1): 805-14, 2005 Jan 07.
Artigo em Inglês | MEDLINE | ID: mdl-15520006

RESUMO

RGS14, a member of the regulator of G-protein signaling (RGS) protein family, possesses an N-terminal RGS domain, two Raf-like Ras-binding domains, and a GoLoco motif, which has GDP dissociation inhibitor activity. In this study we show that unique among the known mammalian RGS proteins, RGS14 localizes in centrosomes. Its first Ras-binding domain is sufficient to target RGS14 to centrosomes. RGS14 also shuttles between the cytoplasm and nucleus, and its nuclear export depends on the CRM-1 nuclear export receptor. Mutation of a nuclear export signal or treatment with leptomycin B causes nuclear accumulation of RGS14 and its association with promyelocytic leukemia protein nuclear bodies. Furthermore, a point mutant defective in nuclear export fails to target to centrosomes, suggesting that nuclear cytoplasmic shuttling is necessary for its proper localization. Mild heat stress, but not proteotoxic or transcription-linked stresses, re-localizes the RGS14 from the cytoplasm to promyelocytic leukemia nuclear bodies. Expression of RGS14, but not point mutants that disrupt the functional activity of its RGS domain or GoLoco motif, enhances the reporter gene activity. The multifunctional domains and the dynamic subcellular localization of RGS14 implicate it in a diverse set of cellular processes including centrosome and nuclear functions and stress-induced signaling pathways.


Assuntos
Centrossomo/metabolismo , Proteínas de Neoplasias/metabolismo , Proteínas Nucleares/metabolismo , Proteínas RGS/metabolismo , Fatores de Transcrição/metabolismo , Sequência de Aminoácidos , Animais , Núcleo Celular/metabolismo , Citoplasma/metabolismo , Células HeLa , Temperatura Alta , Humanos , Corpos de Inclusão Intranuclear/metabolismo , Camundongos , Dados de Sequência Molecular , Células NIH 3T3 , Matriz Nuclear/metabolismo , Proteína da Leucemia Promielocítica , Transporte Proteico , Proteínas RGS/genética , Proteínas Supressoras de Tumor
13.
Mol Cancer Res ; 2(3): 159-69, 2004 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15037655

RESUMO

Multinucleated cells have been noted in pathophysiological states of the liver including infection with hepatitis B virus (HBV), the status of which is also closely associated with genomic instability in liver cancer. Here, we showed that hepatitis B virus X oncoprotein (HBx) expression in Chang cells results in a multinuclear phenotype and an abnormal number of centrosomes (n >or=3). Regulation of centrosome duplication in HBx-expressing ChangX-34 cells was defective and uncoupled from the cell cycle. HBx induced amplification of centrosomes, multipolar spindle formation, and chromosomal missegregation during mitosis and subsequently increased the generation of multinucleated cells and micronuclei formation. Treatment with PD98059, a mitogen-activated protein/extracellular signal-regulated kinase (MEK) 1/2 inhibitor, significantly reduced the number of cells with hyperamplified centrosomes and decreased the multinucleated cells and micronuclei formation. Consistently, the phospho-ERK level during cell progression was substantially higher in ChangX-34 cells than that of Chang cells. In contrast, neither wortmannin, an inhibitor of phosphoinositide-3 kinase, nor SB203589, an inhibitor of p38 mitogen-activated protein kinase (MAPK), showed any effects. Introduction of Ras dominant-negative (D/N) and MEK2 D/N genes into ChangX-34 cells significantly alleviated centrosome amplification, whereas introduction of the PKC D/N and PKB D/N genes did not. Thus, our results demonstrate that the HBx induced centrosome hyperamplification and mitotic aberration by activation of the Ras-MEK-MAPK. Intervention of this signaling pathway could suppress the centrosome amplification as well as mitotic aberration. These findings may provide a possible mechanism by which HBx promotes phenotypic progression by predisposing chromosomal alteration in HBV-infected liver.


Assuntos
Centrossomo/metabolismo , Sistema de Sinalização das MAP Quinases , Mitose/fisiologia , Transativadores/metabolismo , Proteínas ras/metabolismo , Linhagem Celular , Centrossomo/efeitos dos fármacos , Flavonoides/farmacologia , Humanos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Micronúcleos com Defeito Cromossômico/efeitos dos fármacos , Micronúcleos com Defeito Cromossômico/metabolismo , Proteínas Quinases Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Mutação/genética , Fuso Acromático/efeitos dos fármacos , Fuso Acromático/metabolismo , Transfecção , Proteínas Virais Reguladoras e Acessórias , Proteínas ras/genética
14.
Am J Pathol ; 162(3): 721-9, 2003 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-12598306

RESUMO

All blood capillaries consist of endothelial tubes surrounded by mural cells referred to as pericytes. The origin, recruitment, and function of the pericytes is poorly understood, but the importance of these cells is underscored by the severe cardiovascular defects in mice genetically devoid of factors regulating pericyte recruitment to embryonic vessels, and by the association between pericyte loss and microangiopathy in diabetes mellitus. A general problem in the study of pericytes is the shortage of markers for these cells. To identify new markers for pericytes, we have taken advantage of the platelet-derived growth factor (PDGF)-B knockout mouse model, in which developing blood vessels in the central nervous system are almost completely devoid of pericytes. Using cDNA microarrays, we analyzed the gene expression in PDGF-B null embryos in comparison with corresponding wild-type embryos and searched for down-regulated genes. The most down-regulated gene present on our microarray was RGS5, a member of the RGS family of GTPase-activating proteins for G proteins. In situ hybridization identified RGS5 expression in brain pericytes, and in pericytes and vascular smooth muscle cells in certain other, but not all, locations. Absence of RGS5 expression in PDGF-B and PDGFR beta-null embryos correlated with pericyte loss in these mice. Residual RGS5 expression in rare pericytes suggested that RGS5 is a pericyte marker expressed independently of PDGF-B/R beta signaling. With RGS5 as a proof-of-principle, our data demonstrate the usefulness of microarray analysis of mouse models for abnormal pericyte development in the identification of new pericyte-specific markers.


Assuntos
Músculo Liso Vascular/embriologia , Pericitos/citologia , Fator de Crescimento Derivado de Plaquetas/genética , Proteínas RGS/análise , Receptor beta de Fator de Crescimento Derivado de Plaquetas/genética , Transcrição Gênica , Animais , Becaplermina , Biomarcadores , Impressões Digitais de DNA , Embrião de Mamíferos , Feminino , Proteínas de Ligação ao GTP/genética , Regulação da Expressão Gênica no Desenvolvimento , Imuno-Histoquímica , Hibridização In Situ , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Músculo Liso Vascular/citologia , Análise de Sequência com Séries de Oligonucleotídeos , Fator de Crescimento Derivado de Plaquetas/deficiência , Fator de Crescimento Derivado de Plaquetas/fisiologia , Gravidez , Proteínas Proto-Oncogênicas c-sis , Proteínas RGS/genética , Receptor beta de Fator de Crescimento Derivado de Plaquetas/deficiência , Receptor beta de Fator de Crescimento Derivado de Plaquetas/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA