Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
1.
Biochem Biophys Res Commun ; 508(1): 230-236, 2019 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-30482390

RESUMO

We previously reported that the suppression of SIRT2, an NAD + -dependent protein deacetylases, induces p53 accumulation via degradation of p300 and the subsequent MDM2 degradation, eventually leading to apoptosis in HeLa cells. The present study identified a novel pathway of p53 accumulation by SIRT2 suppression in HCT116(p53+/+) cells in which SIRT2 suppression led to escape from mitotic cell death caused by spindle assembly checkpoint activation induced by microtubule inhibitors such as nocodazole but not apoptosis or G1 or G2 arrest. We found that SIRT2 interacts with P/CAF, a histone acetyltransferase, which also acts as a ubiquitin ligase against MDM2. SIRT2 suppression led to an increase of P/CAF acetylation and its stabilization followed by a decrease in MDM2 and activation of the p53-p21 pathway. Depression of mitotic cell death in HCT116(p53+/+) cells with SIRT2 suppression was released by suppression of P/CAF or p21. Thus, the P/CAF-MDM2-p53-p21 axis enables the escape from mitotic cell death and confers resistance to nocodazole in HCT116(p53+/+) cells with SIRT2 suppression. As SIRT2 has attracted attention as a potential target for cancer therapeutics for p53 regulation, the present study provides a molecular basis for the efficacy of SIRT2 for future cancer therapy based on p53 regulation. These findings also suggest an undesirable function of the SIRT2 suppression associated with activation of the p53-p21 pathway in the suppression of mitotic cell death caused by spindle assembly checkpoint activation.


Assuntos
Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Mitose , Proteínas Proto-Oncogênicas c-mdm2/metabolismo , Sirtuína 2/antagonistas & inibidores , Proteína Supressora de Tumor p53/metabolismo , Fatores de Transcrição de p300-CBP/metabolismo , Morte Celular/efeitos dos fármacos , Células HCT116 , Humanos , Mitose/efeitos dos fármacos , Sirtuína 2/metabolismo
2.
Chromosome Res ; 23(1): 111-33, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25657031

RESUMO

Microcell-mediated chromosome transfer (MMCT) is a technique to transfer a chromosome from defined donor cells into recipient cells and to manipulate chromosomes as gene delivery vectors and open a new avenue in somatic cell genetics. However, it is difficult to uncover the function of a single specific gene via the transfer of an entire chromosome or fragment, because each chromosome or fragment contains a set of numerous genes. Thus, alternative tools are human artificial chromosome (HAC) and mouse artificial chromosome (MAC) vectors, which can carry a gene or genes of interest. HACs/MACs have been generated mainly by either a "top-down approach" (engineered creation) or a "bottom-up approach" (de novo creation). HACs/MACs with one or more acceptor sites exhibit several characteristics required by an ideal gene delivery vector, including stable episomal maintenance and the capacity to carry large genomic loci plus their regulatory elements, thus allowing the physiological regulation of the introduced gene in a manner similar to that of native chromosomes. The MMCT technique is also applied for manipulating HACs and MACs in donor cells and delivering them to recipient cells. This review describes the lessons learned and prospects identified from studies on the construction of HACs and MACs, and their ability to drive exogenous gene expression in cultured cells and transgenic animals via MMCT. New avenues for a variety of applications to bio-medical challenges are also proposed.


Assuntos
Tecnologia Biomédica/métodos , Cromossomos Artificiais/genética , Epigênese Genética , Técnicas de Transferência de Genes , Engenharia Genética/métodos , Modelos Genéticos , Animais , Vetores Genéticos/genética , Humanos , Camundongos
3.
Molecules ; 21(5)2016 May 19.
Artigo em Inglês | MEDLINE | ID: mdl-27213315

RESUMO

Microtubule poisons inhibit spindle function, leading to activation of spindle assembly checkpoint (SAC) and mitotic arrest. Cell death occurring in prolonged mitosis is the first target of microtubule poisons in cancer therapies. However, even in the presence of microtubule poisons, SAC and mitotic arrest are not permanent, and the surviving cells exit the mitosis without cytokinesis (mitotic slippage), becoming tetraploid. Another target of microtubule poisons-based cancer therapy is antiproliferative fate after mitotic slippage. The ultimate goal of both the microtubule poisons-based cancer therapies involves the induction of a mechanism defined as mitotic catastrophe, which is a bona fide intrinsic oncosuppressive mechanism that senses mitotic failure and responds by driving a cell to an irreversible antiproliferative fate of death or senescence. This mechanism of antiproliferative fate after mitotic slippage is not as well understood. We provide an overview of mitotic catastrophe, and explain new insights underscoring a causal association between basal autophagy levels and antiproliferative fate after mitotic slippage, and propose possible improved strategies. Additionally, we discuss nuclear alterations characterizing the mitotic catastrophe (micronuclei, multinuclei) after mitotic slippage, and a possible new type of nuclear alteration (clustered micronuclei).


Assuntos
Proliferação de Células/efeitos dos fármacos , Microtúbulos/efeitos dos fármacos , Neoplasias/patologia , Tetraploidia , Humanos
4.
Cell Struct Funct ; 40(1): 51-9, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25736016

RESUMO

Although most cell lines undergo mitotic arrest after prolonged exposure to microtubule inhibitors, some cells subsequently exit this state and become tetraploid. Among these cells, limited numbers of rodent cells are known to undergo multinucleation to generate multiple small independent nuclei, or micronuclei by prolonged colcemid treatment. Micronuclei are thought to be formed when cells shift to a pseudo G1 phase, during which the onset of chromosomal decondensation allows individual chromosomes distributed throughout the cell to serve as sites for the reassembly of nuclear membranes. To better define this process, we used long-term live cell imaging to observe micronucleation induced in mouse A9 cells by treating with the microtubule inhibitor colcemid. Our observations confirm that nuclear envelope formation occurs when mitotic-arrested cells shift to a pseudo G1 phase and adopt a tetraploid state, accompanied by chromosome decondensation. Unexpectedly, only a small number of cells containing large micronuclei were formed. We found that tetraploid micronucleated cells proceeded through an additional cell cycle, shifting to a pseudo G1 phase and forming octoploid micronucleated cells that were smaller and more numerous compared with the tetraploid micronucleated cells. Our data suggest that micronucleation occur when cells shift from mitotic arrest to a pseudo G1 phase, and demonstrate that, rather than being a single event, micronucleation is an inducible recurrent process that leads to the formation of progressively smaller and more numerous micronuclei.


Assuntos
Ciclo Celular/efeitos dos fármacos , Núcleo Celular/efeitos dos fármacos , Núcleo Celular/metabolismo , Microtúbulos/efeitos dos fármacos , Animais , Células CHO , Cromossomos/efeitos dos fármacos , Cricetinae , Cricetulus , Demecolcina/farmacologia , Fase G1/efeitos dos fármacos , Humanos , Camundongos , Mitose/efeitos dos fármacos , Imagem Molecular , Membrana Nuclear/efeitos dos fármacos , Membrana Nuclear/metabolismo , Ploidias
5.
Genes Cells ; 19(10): 766-77, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25195573

RESUMO

Substantially high rate of glycolysis, known as the Warburg effect, is a well-known feature of cancers, and emerging evidence suggests that it supports cancerous proliferation/tumor growth. Phosphoglycerate mutase (PGAM), a glycolytic enzyme, is commonly up-regulated in several cancers, and recent reports show its involvement in the Warburg effect. Here, a comprehensive analysis shows that PGAM is acetylated at lysines 100/106/113/138 in its central region, and a member of the Sirtuin family (class III deacetylase), SIRT2, is responsible for its deacetylation. Over-expression of SIRT2 or mutations at the acetylatable lysines of PGAM attenuates cancer cell proliferation with a concomitant decrease in PGAM activity. We also report that the acetyltransferase PCAF (p300/CBP-associated factor) interacts with PGAM and acetylates its C-terminus, but not the central region. As prior evidence suggests that SIRT2 functions as a tumor suppressor, our results would provide support for the mechanistic basis of this activity.


Assuntos
Fosfoglicerato Mutase/metabolismo , Sirtuína 2/metabolismo , Fatores de Transcrição de p300-CBP/metabolismo , Acetilação , Animais , Arginina/metabolismo , Linhagem Celular Tumoral , Proliferação de Células , Regulação para Baixo , Humanos , Lisina/metabolismo , Camundongos , Mutação , Estrutura Terciária de Proteína , Sirtuína 2/genética , Sirtuínas/metabolismo
6.
Biochem Biophys Res Commun ; 453(3): 588-94, 2014 Oct 24.
Artigo em Inglês | MEDLINE | ID: mdl-25285631

RESUMO

Mitotic catastrophe, a form of cell death that occurs during mitosis and after mitotic slippage to a tetraploid state, plays an important role in the efficacy of cancer cell killing by microtubule inhibitors. Prolonged mitotic arrest at the spindle assembly checkpoint (SAC) is a well-known requirement for mitotic catastrophe and, thus, for conferring sensitivity to microtubule inhibitors. We previously reported that downregulation of SIRT2, a member of the sirtuin family of NAD+-dependent deacetylases, confers resistance to microtubule inhibitors by abnormally prolonging mitotic arrest and thus compromising the cell death pathway after mitotic slippage. Thus, turning off SAC activation after a defined period is an additional requirement for efficient post-slippage death. Here, we investigated whether SIRT2 deacetylates BubR1, which is a core component of the SAC; acetylation of BubR1 at lysine 250 (K250) during prometaphase inhibits its APC/C-dependent proteolysis and thus regulates timing in anaphase entry. We showed that SIRT2 deacetylates BubR1 K250 both in vitro and in vivo. We also found that SIRT2 knockdown leads to increased levels of BubR1 acetylation at prometaphase; however, this increase is not substantial to elevate the levels of total BubR1 or delay the transition from prometaphase to anaphase. The present study shows that SIRT2 is a deacetylase for BubR1 K250, although the abnormally prolonged SAC activation observed in SIRT2 knockdown cells is not accompanied by a change in BubR1 levels or by delayed progression from prometaphase to anaphase.


Assuntos
Anáfase , Prometáfase , Proteínas Serina-Treonina Quinases/metabolismo , Sirtuína 2/metabolismo , Acetilação , Linhagem Celular , Técnicas de Silenciamento de Genes , Humanos , Proteínas Serina-Treonina Quinases/química , Proteólise , Sirtuína 2/química , Sirtuína 2/genética
7.
Genes Cells ; 16(1): 34-45, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21059157

RESUMO

We previously reported that sirtuin 2 (SIRT2), a mammalian member of the NAD+-dependent protein deacetylases, participates in mitotic regulation, specifically, in efficient mitotic cell death caused by the spindle checkpoint. Here, we describe a novel function of SIRT2 that is different from mitotic regulation. SIRT2 down-regulation using siRNA caused apoptosis in cancer cell lines such as HeLa cells, but not in normal cells. The apoptosis was caused by p53 accumulation, which is mediated by p38 MAPK activation-dependent degradation of p300 and the subsequent MDM2 degradation. Sirtuin inhibitors are emerging as antitumor drugs, and this function has been ascribed to the inhibition of SIRT1, the most well-characterized sirtuin that deacetylases p53 to promote cell survival and also binds to other proteins in response to genotoxic stress. This study suggests that SIRT2 can be a novel molecular target for cancer therapy and provides a molecular basis for the efficacy of SIRT2 for future cancer therapy.


Assuntos
Apoptose/genética , Regulação para Baixo , Sirtuína 2/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Antineoplásicos/farmacologia , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/genética , Proteína p300 Associada a E1A/metabolismo , Células HeLa , Humanos , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Sirtuínas/genética , Sirtuínas/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/genética
8.
Mol Ther ; 19(6): 1123-30, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21427707

RESUMO

Pulmonary metastases are the main cause of death in patients with osteosarcoma, however, the molecular mechanisms of metastasis are not well understood. To detect lung metastasis-related microRNA (miRNA) in human osteosarcoma, we compared parental (HOS) and its subclone (143B) human osteosarcoma cell lines showing lung metastasis in a mouse model. miR-143 was the most downregulated miRNA (P < 0.01), and transfection of miR-143 into 143B significantly decreased its invasiveness, but not cell proliferation. Noninvasive optical imaging technologies revealed that intravenous injection of miR-143, but not negative control miRNA, significantly suppressed lung metastasis of 143B (P < 0.01). To search for miR-143 target mRNA in 143B, microarray analyses were performed using an independent RNA pool extracted by two different comprehensive miR-143-target mRNA collecting systems. Western blot analyses revealed that MMP-13 was mostly protein downregulated by miR-143. Immunohistochemistry using clinical samples clearly revealed MMP-13-positive cells in lung metastasis-positive cases, but not in at least three cases showing higher miR-143 expression in the no metastasis group. Taken together, these data indicated that the downregulation of miR-143 correlates with the lung metastasis of human osteosarcoma cells by promoting cellular invasion, probably via MMP-13 upregulation, suggesting that miRNA could be used to develop new molecular targets for osteosarcoma metastasis.


Assuntos
Neoplasias Pulmonares/enzimologia , Neoplasias Pulmonares/secundário , Metaloproteinase 13 da Matriz/metabolismo , MicroRNAs/genética , MicroRNAs/fisiologia , Osteossarcoma/complicações , Animais , Western Blotting , Linhagem Celular Tumoral , Colágeno/química , Humanos , Imuno-Histoquímica , Técnicas In Vitro , Masculino , Metaloproteinase 13 da Matriz/genética , Camundongos , Camundongos Nus , MicroRNAs/química , Reação em Cadeia da Polimerase
9.
Heliyon ; 6(10): e05398, 2020 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-33163667

RESUMO

Mesenchymal stem cells (MSCs) are expected to be useful in bone regeneration treatment for various diseases and conditions, including cleft lip and palate, fracture, and bone absorption. However, to date, MSCs have failed to produce satisfactory results in clinical settings. This is primarily due to the low rate of induced osteogenic differentiation. To realize MSC potential, it is necessary to establish methods for the isolation of MSC-derived living osteoblasts. However, no osteoblast markers have been reported to date. In an attempt to develop a method for the assessment of osteoblast differentiation, we established reporter human immortalized MSC (hiMSC) lines for in vitro monitoring of bone gamma-carboxyglutamate protein (BGLAP, osteocalcin) expression. To this end, we successfully knocked-in an enhanced green fluorescent protein (EGFP) gene cassette immediately downstream of the first ATG of BGLAP via CRISPR-Cas9, and established hiMSC lines expressing EGFP to monitor osteogenic differentiation. On differentiation day 7, EGFP-positive cells were collected by flow cytometric cell sorting, and the expression of EGFP and endogenous BGLAP was analyzed. During osteogenic differentiation, EGFP upregulation was found to correlate with expression of endogenous BGLAP. Moreover, mineralization was confirmed using Alizarin red-S staining after two weeks of osteogenic differentiation of the modified hiMSC lines. The modified hiMSC lines, as well as the derived differentiated osteoblasts obtained herein, are valuable tools for the monitoring osteoblast gene and protein expression, and can be used to develop novel methods for isolating living osteoblasts.

10.
Biochim Biophys Acta ; 1770(2): 206-12, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17184921

RESUMO

Human artificial chromosome (HAC) vectors possess several characteristics sufficient for the requirements of gene therapy vectors, including stable episomal maintenance and mediation of long-term transgene expression. In this study, we adopted an antigen-mediated genetically modified cell amplification (AMEGA) system employing an antibody/cytokine receptor chimera that triggers a growth signal in response to a cognate non-toxic antigen, and applied it to growth control of HAC-transferred cells by adding an antigen that differed from cytokines that may manifest pleiotropic effects. We previously constructed a novel HAC vector, 21 Delta qHAC, derived from human chromosome 21, housed in CHO cells. Here, we constructed an HAC vector harboring an ScFv-gp130 chimera responsive to fluorescein-conjugated BSA (BSA-FL) as well as a model transgene, enhanced green fluorescent protein (EGFP), in CHO cells. The modified HAC was transferred into interleukin (IL)-6-dependent hybridoma 7TD1 cells by microcell-mediated chromosome transfer, and the cells were subsequently found to show BSA-FL-dependent cell growth and sustained expression of EGFP in the absence of IL-6. The AMEGA system in combination with HAC technology will be useful for increasing the efficacy of gene therapy by conferring a growth advantage on the genetically modified cells.


Assuntos
Divisão Celular/fisiologia , Cromossomos Artificiais Humanos/fisiologia , Hibridomas/citologia , Animais , Células CHO , Linhagem Celular , Cricetinae , Cricetulus , Técnicas de Transferência de Genes , Genes Reporter , Proteínas de Fluorescência Verde/genética , Humanos , Interleucina-6/farmacologia , Camundongos , Proteínas Recombinantes de Fusão , Transfecção
11.
Cancer Sci ; 99(1): 23-30, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17956589

RESUMO

The human runt-related transcription factor 3 gene (RUNX3) is considered to be a candidate tumor suppressor gene in gastric carcinoma. However, the role of RUNX3 in the regulation of cell proliferation remains unclear. In the present study, we constructed an adenoviral vector encoding human RUNX3 cDNA under the control of a Tet-responsive promoter (Ad-Tet-FLAG-RUNX3), which regulates the expression of RUNX3 in the presence or absence of doxycycline. A recombinant adenoviral expression vector encoding LacZ (Ad-Tet-LacZ) was used as a negative control. The effect of the transduction of RUNX3 on cell growth was examined using the Tet-On system in a human gastric carcinoma cell line, MKN-1. Exogenous RUNX3 expression was induced successfully by Ad-Tet-FLAG-RUNX3, but not Ad-Tet-LacZ, in the presence of doxycycline in the MKN-1 cells. At 72 h after infection, the proliferative activity in RUNX3-expressing cells was 55% or less of that of the control cells. Flow cytometry revealed that the sub-G(1) peak was increased in cells expressing RUNX3 (34.11%), indicating that the inhibition of cell growth was due to apoptosis, which was confirmed based on Hoechst 33258 staining, the release of cytochrome c from mitochondria into the cytosol, and detection of cleaved caspase-3 by western blotting in MKN-1 cells. Comprehensive analysis using a cDNA microarray showed that RUNX3 upregulated 17 apoptosis-related genes (including FADD, TRAF6, caspase-2, ING1, ING4, Calpain 10, and DNase1) and downregulated 135 apoptosis-related genes (including FLIP, PEA15, TXN2, HSPD1, IKK, and TIAL1) in MKN-1 cells. Pathway analyses to generate functional networks of the genes suggested that promotion of the formation of the death-inducing signaling complex and activation of the mitochondria-mediated pathway were associated with RUNX3-induced apoptosis. In conclusion, our findings suggest that exogenous RUNX3 expression suppressed cell proliferation by inducing apoptosis via the death-receptor mitochondria-mediated pathway in MKN-1 cells.


Assuntos
Apoptose/genética , Subunidade alfa 3 de Fator de Ligação ao Core/genética , Neoplasias Gástricas/genética , Adenoviridae/genética , Processos de Crescimento Celular/genética , Linhagem Celular Tumoral , Subunidade alfa 3 de Fator de Ligação ao Core/biossíntese , DNA Complementar/genética , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Vetores Genéticos/genética , Humanos , Análise de Sequência com Séries de Oligonucleotídeos , Neoplasias Gástricas/patologia , Tetraciclina/farmacologia , Transdução Genética
12.
Biochem Biophys Res Commun ; 372(3): 469-74, 2008 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-18502196

RESUMO

The aim of this study was to elucidate the mechanisms for regulations of cardiac Kv1.5 channel expression. We particularly focused on the role of heat shock proteins (Hsps). We tested the effects of Hsps on the stability of Kv1.5 channels using biochemical and electrophysiological techniques: co-expression of Kv1.5 and Hsp family proteins in mammalian cell lines, followed by Western blotting, immunoprecipitation, pulse-chase analysis, immunofluorescence and whole-cell patch clamp. Hsp70 and heat shock factor 1 increased the expression of Kv1.5 protein in HeLa and COS7 cells, whereas either Hsp40, 27 or 90 did not. Hsp70 prolonged the half-life of Kv1.5 protein. Hsp70 was co-immunoprecipitated and co-localized with Kv1.5-FLAG. Hsp70 significantly increased the immunoreactivity of Kv1.5 in the endoplasmic reticulum, Golgi apparatus and on the cell membrane. Hsp70 enhanced Kv1.5 current of transfected cells, which was abolished by pretreatment with brefeldin A or colchicine. Thus, Hsp70, but not other Hsps, stabilizes functional Kv1.5 protein.


Assuntos
Proteínas de Choque Térmico HSP70/metabolismo , Canal de Potássio Kv1.5/metabolismo , Animais , Western Blotting , Células COS , Membrana Celular/metabolismo , Chlorocebus aethiops , Retículo Endoplasmático/metabolismo , Complexo de Golgi/metabolismo , Proteínas de Choque Térmico HSP70/genética , Células HeLa , Humanos , Imunoprecipitação , Canal de Potássio Kv1.5/genética , Células Musculares/metabolismo , Ratos
13.
Methods Mol Biol ; 1772: 289-296, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29754235

RESUMO

It is a major challenge in biology to know whether chromosome functions of replication, segregation, gene expression, inheritance, etc. are conserved among evolutionary distant organisms where common structural features are maintained. Establishment of hybrid cell lines between evolutionary distant organisms, such as humans and plants, would be one of the promising synthetic approaches to study the evolutionary conservation of chromosome functions. In this chapter, we describe the protocol for successful establishment of human cell lines with a functional plant chromosome. Systematic analyses of hybrid cells will facilitate the evolutionary study of organisms with respect to chromosome functions. It will also provide a basic platform for genome writing and construction of chromosomal shuttle vectors .


Assuntos
Cromossomos de Plantas/genética , Expressão Gênica/genética , Linhagem Celular , Segregação de Cromossomos/genética , Evolução Molecular , Humanos , Células Híbridas/metabolismo
14.
Virchows Arch ; 451(4): 771-9, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17674037

RESUMO

Mitotic arrest deficiency 1 (MAD1) is a component of the spindle checkpoint factors that monitor fidelity of chromosomal segregation. We previously confirmed that the level of MAD1 protein was decreased in gastric carcinoma compared with non-tumoral mucosa by conducting proteome-based analyses (Nishigaki R, Osaki M, Hiratsuka M, Toda T, Murakami K, Jeang KT, Ito H, Inoue T, Oshimura M, Proteomics 5:3205-3213, 29). In this study, an immunohistochemical analysis was performed to examine MAD1 expression histologically in gastric mucosa and tumor. MAD1 was detected in the supranuclear portion of normal epithelial, intestinal metaplasia, and adenoma cells, but its expression was not restricted to any specific area in carcinoma cells. Lower levels of expression were noted in 16 (47.1%) of 34 adenomas and in 52 (60.5%) of 86 carcinomas, whereas all normal mucosae and intestinal metaplasias were grouped into cases with higher level of expression. Moreover, the expression of MAD1 was significantly lower in advanced carcinomas than early carcinomas and in intestinal than diffuse type, respectively (P < 0.05). Exogenous expression of wild-type MAD1, but not the mutant MAD1, inhibited cell proliferation and resulted in G2/M accumulation in MKN-1, a gastric carcinoma cell line. Taken together, our findings suggest that the MAD1 gene could be a candidate tumor suppressor gene and that down-regulation of MAD1 expression contribute to tumorigenesis in human stomach.


Assuntos
Adenocarcinoma/genética , Adenoma/genética , Proteínas de Ciclo Celular/genética , Genes Supressores de Tumor , Proteínas Nucleares/genética , Neoplasias Gástricas/genética , Adenocarcinoma/metabolismo , Adenocarcinoma/patologia , Adenoma/metabolismo , Adenoma/patologia , Ciclo Celular , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular Tumoral , Proliferação de Células , Regulação para Baixo/genética , Epitélio/metabolismo , Epitélio/patologia , Mucosa Gástrica/metabolismo , Mucosa Gástrica/patologia , Regulação Neoplásica da Expressão Gênica , Humanos , Proteínas Nucleares/metabolismo , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patologia
15.
Nucleic Acids Res ; 33(8): 2650-60, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-15888726

RESUMO

Loss of genomic imprinting is involved in a number of developmental abnormalities and cancers. ZAC is an imprinted gene expressed from the paternal allele of chromosome 6q24 within a region known to harbor a tumor suppressor gene for several types of neoplasia. p57(KIP2) (CDKN1C) is a maternally expressed gene located on chromosome 11p15.5 which encodes a cyclin-dependent kinase inhibitor that may also act as a tumor suppressor gene. Mutations in ZAC and p57KIP2 have been implicated in transient neonatal diabetes mellitus (TNDB) and Beckwith-Wiedemann syndrome, respectively. Patients with these diseases share many characteristics. Here we show that mouse Zac1 and p57Kip2 have a strikingly similar expression pattern. ZAC, a sequence-specific DNA-binding protein, binds within the CpG island of LIT1 (KCNQ1OT1), a paternally expressed, anti-sense RNA thought to negatively regulate p57(KIP2) in cis. ZAC induces LIT1 transcription in a methylation-dependent manner. Our data suggest that ZAC may regulate p57(KIP2) through LIT1, forming part of a novel signaling pathway regulating cell growth. Mutations in ZAC may, therefore, contribute to Beckwith-Wiedemann syndrome. Furthermore, we find changes in DNA methylation at the LIT1 putative imprinting control region in two patients with TNDB.


Assuntos
Síndrome de Beckwith-Wiedemann/genética , Proteínas de Ciclo Celular/genética , Impressão Genômica , Proteínas de Membrana/genética , Proteínas Nucleares/genética , Canais de Potássio de Abertura Dependente da Tensão da Membrana/genética , Fatores de Transcrição/genética , Animais , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular Tumoral , Ilhas de CpG , Inibidor de Quinase Dependente de Ciclina p57 , Metilação de DNA , Diabetes Mellitus/genética , Epigênese Genética , Expressão Gênica , Inativação Gênica , Genes Supressores de Tumor , Predisposição Genética para Doença , Humanos , Camundongos , Proteínas Nucleares/metabolismo , Fatores de Transcrição/metabolismo , Ativação Transcricional , Transfecção , Proteínas Supressoras de Tumor
16.
ACS Synth Biol ; 6(2): 301-310, 2017 02 17.
Artigo em Inglês | MEDLINE | ID: mdl-27696824

RESUMO

Replication, segregation, gene expression, and inheritance are essential features of all eukaryotic chromosomes. To delineate the extent of conservation of chromosome functions between humans and plants during evolutionary history, we have generated the first human cell line containing an Arabidopsis chromosome. The Arabidopsis chromosome was mitotically stable in hybrid cells following cell division, and initially existed as a translocated chromosome. During culture, the translocated chromosomes then converted to two types of independent plant chromosomes without human DNA sequences, with reproducibility. One pair of localization signals of CENP-A, a marker of functional centromeres was detected in the Arabidopsis genomic region in independent plant chromosomes. These results suggest that the chromosome maintenance system was conserved between human and plants. Furthermore, the expression of plant endogenous genes was observed in the hybrid cells, implicating that the plant chromosomal region existed as euchromatin in a human cell background and the gene expression system is conserved between two organisms. The present study suggests that the essential chromosome functions are conserved between evolutionarily distinct organisms such as humans and plants. Systematic analyses of hybrid cells may lead to the production of a shuttle vector between animal and plant, and a platform for the genome writing.


Assuntos
Cromossomos de Plantas/genética , Sequência de Bases/genética , Fusão Celular/métodos , Linhagem Celular Tumoral , Centrômero/genética , DNA/genética , Expressão Gênica/genética , Humanos , Células Híbridas/metabolismo , Reprodutibilidade dos Testes
17.
Nucleic Acids Res ; 31(16): 4717-28, 2003 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-12907712

RESUMO

DNA binding by the ternary complex factor (TCF) subfamily of ETS-domain transcription factors is tightly regulated by intramolecular and intermolecular interactions. The helix-loop-helix (HLH)-containing Id proteins are trans-acting negative regulators of DNA binding by the TCFs. In the TCF, SAP-2/Net/ERP, intramolecular inhibition of DNA binding is promoted by the cis-acting NID region that also contains an HLH-like motif. The NID also acts as a transcriptional repression domain. Here, we have studied the role of HLH motifs in regulating DNA binding and transcription by the TCF protein SAP-1 and how Cdk-mediated phosphorylation affects the inhibitory activity of the Id proteins towards the TCFs. We demonstrate that the NID region of SAP-1 is an autoinhibitory motif that acts to inhibit DNA binding and also functions as a transcription repression domain. This region can be functionally replaced by fusion of Id proteins to SAP-1, whereby the Id moiety then acts to repress DNA binding in cis. Phosphorylation of the Ids by cyclin-Cdk complexes results in reduction in protein-protein interactions between the Ids and TCFs and relief of their DNA-binding inhibitory activity. In revealing distinct mechanisms through which HLH motifs modulate the activity of TCFs, our results therefore provide further insight into the role of HLH motifs in regulating TCF function and how the inhibitory properties of the trans-acting Id HLH proteins are themselves regulated by phosphorylation.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Sequências Hélice-Alça-Hélice/genética , Fatores de Transcrição/metabolismo , Sequência de Aminoácidos , Sítios de Ligação/genética , Linhagem Celular , Proteínas de Ligação a DNA/genética , Genes Reporter/genética , Humanos , Proteína 1 Inibidora de Diferenciação , Proteína 2 Inibidora de Diferenciação , Dados de Sequência Molecular , Mutação , Fosforilação , Ligação Proteica , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Sequências Reguladoras de Ácido Nucleico/genética , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Homologia de Sequência de Aminoácidos , Fatores de Transcrição/genética , Transcrição Gênica/genética , Proteínas Elk-1 do Domínio ets , Proteínas Elk-4 do Domínio ets
18.
Stem Cell Rev ; 2(1): 43-50, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-17142886

RESUMO

The recent emergence of stem cell-based tissue engineering has now opened up new venues for gene therapy. The task now is to develop safe and effective vectors that can deliver therapeutic genes into specific stem cell lines and maintain long-term regulated expression of these genes. Human artificial chromosomes (HACs) possess several characteristics that require gene therapy vectors, including a stable episomal maintenance, and the capacity for large gene inserts. HACs can also carry genomic loci with regulatory elements, thus allowing for the expression of transgenes in a genetic environment similar to the chromosome. Currently, HACs are constructed by a two prone approaches. Using a top-down strategy, HACs can be generated from fragmenting endogenous chromosomes. By a bottom-up strategy, HACs can be created de novo from cloned chromosomal components using chromosome engineering. This review describes the current advances in developing HACs, with the main focus on their applications and potential value in gene delivery, such as HAC-mediated gene expression in embryonic, adult stem cells, and transgenic animals.


Assuntos
Cromossomos Artificiais Humanos , Técnicas de Transferência de Genes , Células-Tronco/fisiologia , Animais , Engenharia Genética/métodos , Humanos , Camundongos
19.
Yonago Acta Med ; 58(1): 23-9, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26190894

RESUMO

BACKGROUND: Mesenchymal stem cells (MSCs) hold promise for application in adult stem cell-mediated regenerative medicine in bone remodeling and fracture repair. MSCs in vitro can be directed to osteogenic lineage by dexamethasone (DEX); however, the use of DEX is not practical in clinical settings because of adverse side effects such as glucocorticoid-induced osteoporosis. For identifying substances that facilitate osteogenesis, a monitoring system, which detects the osteogenic differentiation stage of MSCs accurately and easily, is required. METHODS: By focusing on the human osteocalcin (OC) gene whose expression profile is described along with osteogenic differentiation, we constructed the luciferase (Luc) reporter gene driven by the enhancer/promoter sequence of the human OC gene (OC-Luc) utilizing a mammalian artificial chromosome. Mammalian artificial chromosome is a suitable platform for loading reporter constructs, because of its stable episomal maintenance in host cells, transferability into any cell and assurance of long-term physiological transgene expression. We loaded the OC-Luc on a mammalian artificial chromosome vector engineered from mouse chromosome (designated as mouse artificial chromosome, MAC) in Chinese hamster ovary cells (OC-Luc/MAC) and transferred this into human MSC cells via chromosome transfer. RESULTS: OC-Luc/MAC in human MSC cells are responsive to positive and negative stimulation by 1 alpha,25-dihydroxyvitamin D3 and DEX in differentiation stage of MSCs to osteoblasts, reflecting the manner of physiological expression. CONCLUSION: The OC-Luc/MAC reporter system may contribute not only to monitoring the osteogenic differentiation stage from MSC but also to identify novel osteogenic drugs.

20.
J Biochem ; 133(4): 405-13, 2003 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-12761286

RESUMO

We previously cloned a gene for a novel myosin (called MysPDZ) containing a PDZ-domain from bone marrow stromal cells. This new myosin is found in humans and classified as one of the class XVIII myosins (Myo18A). Here, we report the hematopoietic cell-specific splicing isoform (MysPDZbeta) in addition to the previously reported isoform (MysPDZalpha). Combined with mouse genome sequence data, the overall genome structure and generation of the two spliced isoforms are deduced. The MysPDZbeta protein lacks a PDZ-domain in the N-terminal region. Studies of the subcellular localization of the two spliced isoforms indicated that MysPDZalpha containing the PDZ domain co-localizes with the ER-Golgi complex, while MysPDZbeta, which lacks the PDZ domain, localizes diffusely in the cytoplasm. These results suggest that the isoforms differ in their subcellular localization and may have different functions in membrane ruffling and membrane traffic pathways. The PDZ-containing spliced isoform (MysPDZalpha) is not expressed in bone marrow hematopoietic cells, whereas MysPDZbeta lacking the PDZ is specifically expressed in most hematopoietic cells. It is noted that neither isoform is expressed in red blood cells. Interestingly, MysPDZalpha was detected in mature but not in immature macrophages, and its level increased after the induction of differentiation of M1 cells, suggesting a functional role of PDZ-containing myosin in macrophages.


Assuntos
Miosinas/biossíntese , Animais , Sequência de Bases , Células Cultivadas , DNA Complementar/biossíntese , DNA Complementar/isolamento & purificação , Imunofluorescência , Regulação da Expressão Gênica , Genoma , Complexo de Golgi/metabolismo , Células-Tronco Hematopoéticas/metabolismo , Humanos , Macrófagos/metabolismo , Camundongos , Dados de Sequência Molecular , Miosinas/genética , Miosinas/isolamento & purificação , Isoformas de Proteínas/biossíntese , Isoformas de Proteínas/genética , RNA Mensageiro/análise , RNA Mensageiro/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA