Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Mol Carcinog ; 63(4): 714-727, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38251858

RESUMO

The histone variant, macroH2A (mH2A) influences gene expression through epigenetic regulation. Tumor suppressive function of mH2A isoforms has been reported in various cancer types, but few studies have investigated the functional role of mH2A2 in breast cancer pathophysiology. This study aimed to determine the significance of mH2A2 in breast cancer development and progression by exploring its downstream regulatory mechanisms. Knockdown of mH2A2 facilitated the migration and invasion of breast cancer cells, whereas its overexpression exhibited the opposite effect. In vivo experiments revealed that augmenting mH2A2 expression reduced tumor growth and lung metastasis. Microarray analysis showed that TM4SF1 emerged as a likely target linked to mH2A2 owing to its significant suppression in breast cancer cell lines where mH2A2 was overexpressed among the genes that exhibited over twofold upregulation upon mH2A2 knockdown. Suppressing TM4SF1 reduced the migration, invasion, tumor growth, and metastasis of breast cancer cells in vitro and in vivo. TM4SF1 depletion reversed the increased aggressiveness triggered by mH2A2 knockdown, suggesting a close interplay between mH2A2 and TM4SF1. Our findings also highlight the role of the mH2A2/TM4SF1 axis in activating the AKT/NF-κB pathway. Consequently, activated NF-κB signaling leads to increased expression and secretion of MMP13, a potent promoter of metastasis. In summary, we propose that the orchestrated regulation of the mH2A2/TM4SF1 axis in conjunction with the AKT/NF-κB pathway and the subsequent elevation in MMP13 expression constitute pivotal factors governing the malignancy of breast cancer.


Assuntos
Neoplasias da Mama , NF-kappa B , Humanos , Feminino , NF-kappa B/genética , NF-kappa B/metabolismo , Histonas/metabolismo , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Neoplasias da Mama/metabolismo , Epigênese Genética , Metaloproteinase 13 da Matriz/genética , Metaloproteinase 13 da Matriz/metabolismo , Proteínas de Neoplasias/genética , Linhagem Celular Tumoral , Movimento Celular/genética , Regulação Neoplásica da Expressão Gênica , Proliferação de Células/fisiologia , Antígenos de Superfície/genética , Antígenos de Superfície/metabolismo
2.
EMBO Rep ; 24(8): e56335, 2023 08 03.
Artigo em Inglês | MEDLINE | ID: mdl-37341560

RESUMO

While there is growing evidence that many epigenetically silenced genes in cancer are tumour suppressor candidates, their significance in cancer biology remains unclear. Here, we identify human Neuralized (NEURL), which acts as a novel tumour suppressor targeting oncogenic Wnt/ß-catenin signalling in human cancers. The expression of NEURL is epigenetically regulated and markedly suppressed in human colorectal cancer. We, therefore, considered NEURL to be a bona fide tumour suppressor in colorectal cancer and demonstrate that this tumour suppressive function depends on NEURL-mediated oncogenic ß-catenin degradation. We find that NEURL acts as an E3 ubiquitin ligase, interacting directly with oncogenic ß-catenin, and reducing its cytoplasmic levels in a GSK3ß- and ß-TrCP-independent manner, indicating that NEURL-ß-catenin interactions can lead to a disruption of the canonical Wnt/ß-catenin pathway. This study suggests that NEURL is a therapeutic target against human cancers and that it acts by regulating oncogenic Wnt/ß-catenin signalling.


Assuntos
Neoplasias do Colo , beta Catenina , Humanos , beta Catenina/genética , beta Catenina/metabolismo , Via de Sinalização Wnt , Neoplasias do Colo/genética , Ubiquitina-Proteína Ligases/metabolismo , Proteínas Contendo Repetições de beta-Transducina/genética , Proteínas Contendo Repetições de beta-Transducina/metabolismo , Linhagem Celular Tumoral
3.
Mech Ageing Dev ; 173: 50-60, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29625135

RESUMO

Chromatin change is one of the crucial causes of aging. Specifically, maintenance of heterochromatin stability is critical for cellular integrity, and its loss induces genomic instability and cellular aging. However, the causes and effects of heterochromatin instability in multicellular tissue aging still remain unclear. Here, in the adult Drosophila midgut, we report age-related loss of heterochromatin stability in enterocytes (ECs) due to the loss and dispersion of tri-methylated histone H3 Lys9 (H3K9me3) and heterochromatin protein 1 (HP1). Our study further shows that EC-specific knockdown of Su(var)3-9, histone lysine methyltransferase for H3K9me3 formation, or HP1a leads to intestinal stem cell (ISC) aging through genomic stress, JNK signaling, and apoptotic death in ECs. Our findings revealed the plausible causes of age-related loss of heterochromatin stability in ECs, including oxidative stress and nutrient-sensing AKT/TOR signaling. Taken together, the loss of heterochromatin stability may be the crucial niche aging mechanism for ISC aging which is the prime determinant of intestinal tissue aging. Furthermore, our study provides new clues on the link between heterochromatin and aging.


Assuntos
Envelhecimento/metabolismo , Heterocromatina/metabolismo , Mucosa Intestinal/metabolismo , Transdução de Sinais , Células-Tronco/metabolismo , Envelhecimento/genética , Envelhecimento/patologia , Animais , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster , Técnicas de Silenciamento de Genes , Heterocromatina/genética , Heterocromatina/patologia , Histona-Lisina N-Metiltransferase/genética , Histona-Lisina N-Metiltransferase/metabolismo , Intestinos/patologia , Células-Tronco/patologia
4.
Oncol Lett ; 11(3): 1895-1902, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26998096

RESUMO

Breast cancer is one of the most common cancer types among women, acting as a distinct cause of mortality, and has a high incidence of recurrence. External stimuli, including 17ß-estradiol (E2), transforming growth factor (TGF)-ß1 and hypoxia, may be important in breast cancer growth and metastasis. However, the effects of these stimuli on breast cancer stem cell (CSC) regulation have not been fully investigated. In the present study, the proportion of cluster of differentiation (CD)44+/CD24-/low cells increased following treatment with E2, TGF-ß1 and hypoxia in MCF-7 cells. The expression of CSC markers, including SOX2, KLF4 and ABCG2, was upregulated continually by E2, TGF-ß1 and hypoxia. In addition, the expression levels of epithelial-mesenchymal transition-associated factors increased following treatment with E2, TGF-ß1 and hypoxia. Therefore, the migration ability of E2-, TGF-ß1- and hypoxia-treated MCF-7 cells was enhanced compared with control cells. In addition, the enhancement of apoptosis by 5-flurouracil or radiation was abolished following treatment with E2, TGF-ß1 and hypoxia. These results indicate that E2, TGF-ß1 and hypoxia are important for regulating breast CSCs, and that the modulation of the microenvironment in tumors may improve the efficiency of breast cancer therapy.

5.
BMC Genomics ; 16: 56, 2015 Feb 06.
Artigo em Inglês | MEDLINE | ID: mdl-25887185

RESUMO

BACKGROUND: Exposure to ionizing radiation (IR) results in the simultaneous activation or downregulation of multiple signaling pathways that play critical roles in cell type-specific control of survival or death. IR is a well-known genotoxic agent and human carcinogen that induces cellular damage through direct and indirect mechanisms. However, its impact on epigenetic mechanisms has not been elucidated, and more specifically, little information is available regarding genome-wide DNA methylation changes in cancer cells after IR exposure. Recently, genome-wide DNA methylation profiling technology using the Illumina HumanMethylation450K platform has emerged that allows us to query >450,000 loci within the genome. This improved technology is capable of identifying genome-wide DNA methylation changes in CpG islands and other CpG island-associated regions. RESULTS: In this study, we employed this technology to test the hypothesis that exposure to IR not only induces differential DNA methylation patterns at a genome-wide level, but also results in locus- and gene-specific DNA methylation changes. We screened for differential DNA methylation changes in colorectal cancer cells after IR exposure with 2 and 5 Gy. Twenty-nine genes showed radiation-induced hypomethylation in colon cancer cells, and of those, seven genes showed a corresponding increase in gene expression by reverse transcriptase polymerase chain reaction (RT-PCR). In addition, we performed chromatin immunoprecipitation (ChIP) to confirm that the DNA-methyltransferase 1 (DNMT1) level associated with the promoter regions of these genes correlated with their methylation level and gene expression changes. Finally, we used a gene ontology (GO) database to show that a handful of hypomethylated genes induced by IR are associated with a variety of biological pathways related to cancer. CONCLUSION: We identified alterations in global DNA methylation patterns and hypomethylation at specific cancer-related genes following IR exposure, which suggests that radiation exposure plays a critical role in conferring epigenetic alterations in cancer.


Assuntos
Neoplasias do Colo/genética , Metilação de DNA/genética , Epigênese Genética , Neoplasias Induzidas por Radiação/genética , Neoplasias do Colo/patologia , Ilhas de CpG , DNA (Citosina-5-)-Metiltransferases/genética , Metilação de DNA/efeitos da radiação , Regulação Neoplásica da Expressão Gênica/genética , Regulação Neoplásica da Expressão Gênica/efeitos da radiação , Genoma Humano/efeitos da radiação , Células HCT116 , Humanos , Neoplasias Induzidas por Radiação/patologia , Regiões Promotoras Genéticas , Radiação Ionizante
6.
PLoS One ; 9(8): e105405, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25136811

RESUMO

Exposure of cells to ionizing radiation (IR) induces, not only, activation of multiple signaling pathways that play critical roles in cell fate determination, but also alteration of molecular pathways involved in cell death or survival. Recently, DNA methylation has been established as a critical epigenetic process involved in the regulation of gene expression in cancer cells, suggesting that DNA methylation inhibition may be an effective cancer treatment strategy. Because alterations of gene expression by DNA methylation have been considered to influence radioresponsiveness, we investigated the effect of a DNA methyltransferase inhibitor, 5-aza-2'-deoxycytidine (5-aza-dC), on radiosensitivity. In addition, we investigated the underlying cellular mechanisms of combination treatments of ionizing irradiation (IR) and 5-aza-dC in human colon cancer cells. Colon cancer cell lines were initially tested for radiation sensitivity by IR in vitro and were treated with two different doses of 5-aza-dC. Survival of these cell lines was measured using MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide) and clonogenic assays. The effects of 5-aza-dC along with irradiation on cell growth, cell cycle distribution, apoptosis, and apoptosis-related gene expression were examined. Combination irradiation treatment with 5-aza-dC significantly decreased growth activity compared with irradiation treatment alone or with 5-aza-dC treatment alone. The percentage of HCT116 cells in the sub-G1 phase and their apoptotic rate was increased when cells were treated with irradiation in combination with 5-aza-dC compared with either treatment alone. These observations were strongly supported by increased caspase activity, increased comet tails using comet assays, and increased protein levels of apoptosis-associated molecules (caspase 3/9, cleaved PARP). Our data demonstrated that 5-aza-dC enhanced radiosensitivity in colon cancer cells, and the combination effects of 5-aza-dC with radiation showed greater cellular effects than that of single treatment, suggesting that the combination of 5-aza-dC and radiation has the potential to become a clinical strategy for the treatment of cancer.


Assuntos
Azacitidina/análogos & derivados , Neoplasias Colorretais/tratamento farmacológico , Neoplasias Colorretais/radioterapia , Epigênese Genética/efeitos dos fármacos , Epigênese Genética/efeitos da radiação , Animais , Apoptose/efeitos dos fármacos , Apoptose/genética , Apoptose/efeitos da radiação , Proteínas Reguladoras de Apoptose/metabolismo , Azacitidina/farmacologia , Linhagem Celular Tumoral , Neoplasias Colorretais/genética , Neoplasias Colorretais/metabolismo , Terapia Combinada/métodos , Metilação de DNA/efeitos dos fármacos , Metilação de DNA/genética , Metilação de DNA/efeitos da radiação , Decitabina , Epigênese Genética/genética , Feminino , Fase G1/efeitos dos fármacos , Fase G1/genética , Fase G1/efeitos da radiação , Células HCT116 , Humanos , Camundongos , Camundongos SCID , Tolerância a Radiação/efeitos dos fármacos , Tolerância a Radiação/genética , Radiação Ionizante
7.
Epigenetics ; 9(7): 1018-30, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24785261

RESUMO

Altered expression of microRNA (miRNA) can significantly contribute to cancer development and recent studies have shown that a number of miRNAs may be regulated by DNA methylation. Through a candidate gene approach, we identified MIR941 and MIR1247 to be transcriptionally silenced by DNA hypermethylation in several gastric cancer cell lines. We confirmed that these miRNAs are also densely methylated in primary gastric cancers but not in normal gastric tissues. In addition, we demonstrated that ectopic expression of these two miRNAs in AGS gastric cancer cells resulted in suppression of growth and migration. Furthermore, we tested genes predicted to be the targets of MIR941 and MIR1247 and identified 7 and 6 genes, whose expressions were significantly downregulated by transfection of MIR941 and MIR1247 mimics, respectively, in gastric cancer cell lines. Some of these genes are known to promote proliferation and invasion, phenotypes we observed upon ectopic expression of the two miRNAs. Thus, we examined these candidates more closely and found that downregulation of mRNA corresponded to a decrease in protein levels (observed by western blot). Our study provides unequivocal evidence that MIR941 and MIR1247 are transcriptionally regulated by DNA methylation in gastric cancer and that they have tumor suppressor properties through their inhibition of key cancer promoting genes in this context.


Assuntos
Adenocarcinoma/patologia , Adenoma/patologia , Movimento Celular , Proliferação de Células , Epigênese Genética , MicroRNAs/genética , RNA Mensageiro/metabolismo , Neoplasias Gástricas/patologia , Adenocarcinoma/metabolismo , Adenoma/metabolismo , Adulto , Idoso , Idoso de 80 Anos ou mais , Linhagem Celular Tumoral , Metilação de DNA , Feminino , Inativação Gênica , Humanos , Masculino , MicroRNAs/metabolismo , Pessoa de Meia-Idade , Neoplasias Gástricas/metabolismo
8.
Radiat Res ; 181(4): 376-86, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24701963

RESUMO

Although the diverse effects of ionizing radiation on biological and pathological processes at various levels ranging from molecular to whole body are well studied, the effects on adult stem cells by ionizing radiation remain largely unknown. In this study, we characterized the functional modifications of adult Drosophila midgut intestinal stem cells after ionizing radiation treatment. A dose of 10 Gy of radiation decreased the proliferative capacity of intestinal stem cells. Interestingly, after irradiation at 2 Gy, the intestinal stem cells exhibited increased proliferative activity, misdifferentiation and γH2AvD and 8-oxo-dG levels. In addition, the guts irradiated with 2 Gy showed increased JNK and AKT activities. Furthermore, we showed that 2 Gy of ionizing radiation induced centrosome amplification in intestinal stem cells of adult midguts. Our data gives molecular insights into the effects of ionizing radiation on functional modifications of stem cells. The adult Drosophila midgut intestinal stem cells offer a potentially rich new system for the exploration of the biological effects of ionizing radiation.


Assuntos
Intestinos/efeitos da radiação , Radiação Ionizante , Células-Tronco/efeitos da radiação , Animais , Proliferação de Células/efeitos da radiação , Centrossomo , Dano ao DNA , Drosophila , Intestinos/citologia , Células-Tronco/citologia
9.
Int J Mol Sci ; 14(7): 15059-73, 2013 Jul 18.
Artigo em Inglês | MEDLINE | ID: mdl-23873297

RESUMO

Cancer is a disease that results from both genetic and epigenetic changes. In recent decades, a number of people have investigated the disparities in gene expression resulting from variable DNA methylation alteration and chromatin structure modification in response to the environment. Especially, colon cancer is a great model system for investigating the epigenetic mechanism for aberrant gene expression alteration. Ionizing radiation (IR) could affect a variety of processes within exposed cells and, in particular, cause changes in gene expression, disruption of cell cycle arrest, and apoptotic cell death. Even though there is growing evidence on the importance of epigenetics and biological processes induced by radiation exposure in various cancer types including colon cancer, specific epigenetic alterations induced by radiation at the molecular level are incompletely defined. This review focuses on discussing possible IR-mediated changes of DNA methylation and histone modification in cancer.


Assuntos
Epigenômica , Neoplasias/radioterapia , Metilação de DNA/efeitos da radiação , Histonas/metabolismo , Humanos , MicroRNAs/metabolismo , Neoplasias/genética , Neoplasias/metabolismo , Radiação Ionizante
10.
Biochem Biophys Res Commun ; 434(4): 722-7, 2013 May 17.
Artigo em Inglês | MEDLINE | ID: mdl-23583388

RESUMO

We studied the roles of JMJD1A and its target gene ADM in the growth of hepatocellular carcinomas (HCCs) and breast cancer cells under hypoxic conditions. Hypoxia stimulated HepG2 and Hep3B cell proliferation but had no effect on MDA-MB-231 cell proliferation. Interestingly, the JMJD1A and ADM expressions were enhanced by hypoxia only in HepG2 and Hep3B cells. Our ChIP results showed that hypoxia-induced HepG2 and Hep3B cell proliferation is mediated by JMJD1A upregulation and subsequent decrease in methylation in the ADM promoter region. Furthermore, JMJD1A gene silencing abrogated the hypoxia-induced ADM expression and inhibited HepG2 and Hep3B cell growth. These data suggest that JMJD1A might function as a proliferation regulator in some cancer cell types.


Assuntos
Adrenomedulina/genética , Proliferação de Células , Regulação Neoplásica da Expressão Gênica , Histona Desmetilases com o Domínio Jumonji/genética , Adrenomedulina/metabolismo , Animais , Western Blotting , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patologia , Hipóxia Celular , Linhagem Celular Tumoral , Metilação de DNA , Feminino , Células Hep G2 , Humanos , Histona Desmetilases com o Domínio Jumonji/metabolismo , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Regiões Promotoras Genéticas/genética , Interferência de RNA , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Ensaios Antitumorais Modelo de Xenoenxerto
11.
Biochim Biophys Acta ; 1819(11-12): 1200-7, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23046878

RESUMO

Histone modifying factors are functional components of chromatin and play a role in gene regulation. The expression level of JMJD2B, a histone demethylase, is notably up-regulated in cancer tissues. Upregulation of JMJD2B promotes cancer cell proliferation under hypoxic conditions through target gene expression. Here, we describe the patterns of histone methylation and JMJD2B expression under various stressed conditions, such as hypoxia and radiation, in a gastric cancer cell line. JMJD2B expression in AGS cells was actively regulated by hypoxia and radiation. Chromatin immunoprecipitation experiments demonstrated that binding of JMJD2B on the cyclin A1 (CCNA1) promoter resulted in CCNA1 upregulation under hypoxic conditions. Furthermore, we confirmed that AGS cell proliferation was directly affected by JMJD2B and CCNA1 expression by performing experiments with JMJD2B depleted cells. Interestingly, the effects of JMJD2B on cell growth under hypoxia were remarkably repressed after gamma-ray irradiation. These results suggest that JMJD2B may play a central role in gastric cancer cell growth and might constitute a novel therapeutic target to overcome hypoxia-induced radio-resistance, thereby improving the efficiency of radiation therapy.


Assuntos
Proliferação de Células/efeitos da radiação , Raios gama , Regulação Enzimológica da Expressão Gênica/efeitos da radiação , Regulação Neoplásica da Expressão Gênica/efeitos da radiação , Histona Desmetilases com o Domínio Jumonji/biossíntese , Proteínas de Neoplasias/metabolismo , Tolerância a Radiação/efeitos da radiação , Neoplasias Gástricas/enzimologia , Hipóxia Celular/efeitos da radiação , Linhagem Celular Tumoral , Ciclina A1/genética , Ciclina A1/metabolismo , Regulação Enzimológica da Expressão Gênica/genética , Regulação Neoplásica da Expressão Gênica/genética , Humanos , Histona Desmetilases com o Domínio Jumonji/genética , Proteínas de Neoplasias/genética , Tolerância a Radiação/genética , Neoplasias Gástricas/genética , Neoplasias Gástricas/patologia , Neoplasias Gástricas/terapia
12.
Food Chem Toxicol ; 50(9): 3174-80, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22699087

RESUMO

Gastrointestinal injury is a major cause of death following exposure to high levels of radiation, and no effective treatments are currently available. In this study, we examined the capacity of granulocyte colony-stimulating factor (G-CSF) to mitigate intestinal injury in, and improve survival of, C3H/HeN mice given a lethal dose (12 Gy) of radiation to the abdomen. G-CSF (100 µg/kg body weight) was injected subcutaneously daily for 3 days after irradiation and shown to improve survival and intestinal morphology at 3.5 days compared with saline-injected controls. The morphological features improved by G-CSF included crypt number and depth, villous length, and the length of basal lamina of 10 enterocytes. G-CSF also normalized the levels of circulating tumor necrosis factor alpha and attenuated the loss of peripheral neutrophils, caused by radiation-induced myelosuppression. In conclusion, our results suggest that G-CSF enhanced the survival of irradiated mice and minimized the effects of radiation on gastrointestinal injury.


Assuntos
Fator Estimulador de Colônias de Granulócitos/farmacologia , Jejuno/efeitos da radiação , Lesões Experimentais por Radiação/prevenção & controle , Animais , Citocinas/sangue , Fator Estimulador de Colônias de Granulócitos/administração & dosagem , Injeções Subcutâneas , Jejuno/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C3H , Taxa de Sobrevida
13.
Cell Struct Funct ; 36(2): 197-208, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21979236

RESUMO

Recent studies have suggested the involvement of epigenetic factors such as methyl-CpG-binding protein-2 (MeCP2) in tumorigenesis. In addition, cancer may represent a stem cell-based disease, suggesting that understanding of stem cell regulation could provide valuable insights into the mechanisms of tumorigenesis. However, the function of epigenetic factors in stem cell regulation in adult tissues remains poorly understood. In the present study, we investigated the role of human MeCP2 (hMeCP2), a bridge factor linked to DNA modification and histone modification, in stem cell proliferation using adult Drosophila midgut, which appears to be an excellent model system to study stem cell biology. Results show that enterocyte (EC)-specific expression of hMeCP2 in adult midgut using an exogenous GAL4/UAS expression system induced intestinal stem cell (ISC) proliferation marked by staining with anti-phospho-histone H3 antibody and BrdU incorporation assays. In addition, hMeCP2 expression in ECs activated extracellular stress-response kinase signals in ISCs. Furthermore, expression of hMeCP2 modulated the distribution of heterochromatin protein-1 in ECs. Our data suggests the hypothesis that the expression of hMeCP2 in differentiated ECs stimulates ISC proliferation, implying a role of MeCP2 as a stem cell regulator.


Assuntos
Intestinos/citologia , Proteína 2 de Ligação a Metil-CpG/metabolismo , Células-Tronco/citologia , Animais , Bromodesoxiuridina/metabolismo , Proliferação de Células , Células Cultivadas , Cromatina/metabolismo , Homólogo 5 da Proteína Cromobox , Proteínas Cromossômicas não Histona/metabolismo , Drosophila , Proteínas de Drosophila/metabolismo , Histonas/metabolismo , Humanos , Proteína 2 de Ligação a Metil-CpG/genética , Células-Tronco/metabolismo , Transfecção
14.
Biochim Biophys Acta ; 1799(7): 510-9, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20346429

RESUMO

The Drosophila midgut is an excellent model for evaluation of gene networks that regulate adult stem cell proliferation and differentiation. The Drosophila p38b (D-p38b) gene has been shown to be involved in intestinal stem cell (ISC) proliferation and differentiation in the adult midgut. Here, we report that D-p38b gene expression is regulated by DREF (DNA replication-related element binding factor) in the adult midgut. We have identified a DRE in the 5'-flanking region of the D-p38b gene and showed that DREF could bind to this DRE via a gel mobility shift assay and a ChIP assay. Base-substitution mutations of the D-p38b promoter DRE and analyses of transformants carrying D-p38b-lacZ or D-p38b-DREmut-lacZ indicated that this DRE is required for the activity of the D-p38b gene promoter. Furthermore, by using the GAL4-UAS system, we showed that DREF regulates the activity of the D-p38b gene promoter in adult ISCs and progenitors. In addition, the D-p38b knockdown phenotypes in the midgut were rescued by DREF overexpression, suggesting a functional link between these two factors. Our results suggest that the D-p38b gene is regulated by the DREF pathway and that DREF is involved in the regulation of proliferation and differentiation of Drosophila ISCs and progenitors.


Assuntos
Proteínas de Drosophila/genética , Drosophila melanogaster/metabolismo , Regulação da Expressão Gênica , Mucosa Intestinal/metabolismo , Proteína Quinase 11 Ativada por Mitógeno/genética , Proteína Quinase 11 Ativada por Mitógeno/metabolismo , Fatores de Transcrição/genética , Animais , Animais Geneticamente Modificados , Linhagem da Célula , Proteínas de Drosophila/metabolismo , Proteínas de Fluorescência Verde/metabolismo , Modelos Biológicos , Mutação , Fenótipo , Regiões Promotoras Genéticas , Células-Tronco/citologia , Fatores de Transcrição/metabolismo
15.
Aging Cell ; 7(3): 318-34, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18284659

RESUMO

Age-associated changes in stem cell populations have been implicated in age-related diseases, including cancer. However, little is known about the underlying molecular mechanisms that link aging to the modulation of adult stem cell populations. Drosophila midgut is an excellent model system for the study of stem cell renewal and aging. Here we describe an age-related increase in the number and activity of intestinal stem cells (ISCs) and progenitor cells in Drosophila midgut. We determined that oxidative stress, induced by paraquat treatment or loss of catalase function, mimicked the changes associated with aging in the midgut. Furthermore, we discovered an age-related increase in the expression of PVF2, a Drosophila homologue of human PDGF/VEGF, which was associated with and required for the age-related changes in midgut ISCs and progenitor cell populations. Taken together, our findings suggest that PDGF/VEGF may play a central role in age-related changes in ISCs and progenitor cell populations, which may contribute to aging and the development of cancer stem cells.


Assuntos
Senescência Celular/fisiologia , Drosophila melanogaster/fisiologia , Fatores de Crescimento do Endotélio Vascular/fisiologia , Animais , Catalase/fisiologia , Diferenciação Celular/fisiologia , Drosophila melanogaster/citologia , Imunoquímica , Intestinos/citologia , Intestinos/fisiologia , Estresse Oxidativo/fisiologia , RNA/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Células-Tronco/citologia , Células-Tronco/fisiologia , Fatores de Crescimento do Endotélio Vascular/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...