Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
1.
Pathol Res Pract ; 252: 154948, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37977034

RESUMO

Lung cancer, known for its high mortality rates and poor prognosis, remains one of the most prevalent cancer types. Early detection and effective treatment methods are crucial for improving survival rates. Non-small cell lung cancer (NSCLC) accounts for approximately 85 % of all lung cancer cases. Long non-coding RNAs (lncRNAs), which play vital roles in various biological processes, have been implicated in the development of cancer and can impact key therapeutic targets in different cancer types. In NSCLC, the dysregulation of specific lncRNAs, such as MALAT1 and NORAD, has been associated with neoplastic initiation, progression, metastasis, tumor angiogenesis, chemoresistance, and genomic instability. Both MALAT1 and NORAD directly regulate the expression of the transcription factor E2F1, thereby influencing cell cycle progression. Additionally, MALAT1 has been reported to affect the expression of p53 target genes, leading to cell cycle progression through the repression of p53 promoter activity. NORAD, on the other hand, is indirectly regulated by p53. The AT-rich interaction domain (ARID) family of DNA-binding proteins, particularly ARID3A and ARID3B, are involved in various biological processes such as cell proliferation, differentiation, and development. They also play significant roles in E2F-dependent transcription and are transcriptional targets of p53. The intricate balance between promoting cellular proliferation through the pRB-E2F pathway and inducing growth arrest through the p53 pathway underscores the crucial regulatory role of ARID3A, ARID3B, and their interaction with lncRNAs MALAT1 and NORAD. In this study, we aimed to investigate the potential interactive and functional connections among ARID3A, ARID3B, MALAT1, and NORAD in NSCLC, considering their involvement in the pRB-E2F and p53 pathways. Our findings strongly suggest that ARID3A and ARID3B play a regulatory role in controlling MALAT1 and NORAD in NSCLC. Specifically, our study demonstrates that the activities of MALAT1 and NORAD were markedly increased upon the overexpression of ARID3A and ARID3B. Therefore, we can conclude that ARID3A and ARID3B likely contribute significantly to the oncogenic functions of MALAT1 and NORAD in NSCLC. Consequently, targeting ARID3A and ARID3B could hold promise as a therapeutic approach in NSCLC, given their direct control over the expression of MALAT1 and NORAD.


Assuntos
Carcinoma Pulmonar de Células não Pequenas , Neoplasias Pulmonares , MicroRNAs , RNA Longo não Codificante , Humanos , Carcinoma Pulmonar de Células não Pequenas/genética , Linhagem Celular Tumoral , Proliferação de Células , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Regulação Neoplásica da Expressão Gênica/genética , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , MicroRNAs/genética , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , Fatores de Transcrição/metabolismo , Proteína Supressora de Tumor p53/metabolismo
2.
J Vis Exp ; (197)2023 Jul 14.
Artigo em Inglês | MEDLINE | ID: mdl-37522726

RESUMO

Conventional bone regeneration therapy using mesenchymal stem cells (MSCs) is difficult to apply to bone defects larger than the critical size because it does not have a mechanism to induce angiogenesis. Implanting artificial cartilage tissue fabricated from MSCs induces angiogenesis and bone formation in vivo via endochondral ossification (ECO). Therefore, this ECO-mediated approach may be a promising bone regeneration therapy in the future. An important aspect of the clinical application of this ECO-mediated approach is establishing a protocol for preparing enough cartilage to be implanted to repair the bone defect. It is especially not practical to design a single mass of grafted cartilage of a size that conforms to the shape of the actual bone defect. Therefore, the cartilage to be transplanted must have the property of forming bone integrally when multiple pieces are implanted. Hydrogels may be an attractive tool for scaling up tissue-engineered grafts for endochondral ossification to meet clinical requirements. Although many naturally derived hydrogels support MSC cartilage formation in vitro and ECO in vivo, the optimal scaffold material to meet the needs of clinical applications has yet to be determined. Hyaluronic acid (HA) is a crucial component of the cartilage extracellular matrix and is a biodegradable and biocompatible polysaccharide. Here, we show that HA hydrogels have excellent properties to support in vitro differentiation of MSC-based cartilage tissue and promote endochondral bone formation in vivo.

3.
PLoS One ; 18(2): e0281345, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36730328

RESUMO

Engineered cartilage tissue from differentiated mesenchymal stem cells (MSCs) can generate bone in vivo through endochondral ossification (ECO). This ECO-mediated approach has the potential to circumvent the severe problems associated with conventional MSC-based bone tissue engineering techniques that lack mechanisms to induce angiogenesis. Hyaluronic acid (HA) is a key component in the cartilage extracellular matrix. However, the ECO-supporting properties of HA remain largely unclear. This study aimed to compare the ability of HA and collagen hydrogels to support in vitro differentiation of MSC-based hypertrophic cartilage tissues and to promote endochondral bone formation in vivo. Following the chondrogenic and hypertrophic differentiation in vitro, both HA and collagen constructs accumulated sulfated glycosaminoglycan (sGAG) and type 1, type II, and type X collagen. However, HA hydrogels exhibited a more uniform distribution of sGAG, type 1 collagen, type X collagen, and osteocalcin proteins; in addition, the cells embedded in the hydrogels had more rounded cell morphologies than those in the collagen constructs. At week 5 of in vitro culture, two to three constructs were implanted into a subcutaneous pocket in nude mice and harvested after 4 and 8 weeks. Both HA and collagen constructs promoted endochondral bone formation with vascularization and bone marrow development; however, the HA constructs fused to form integrated bone tissues and the bone marrow developed along the space between the two adhered grafts in all implanted pockets (n = 5). In the collagen constructs, the integration was observed in 40% of the pockets (n = 5). Microcomputer CT analysis revealed that the bone volume of HA constructs was larger than that of collagen constructs. In conclusion, compared to collagen hydrogels, HA hydrogels had superior potential to generate integrated bone with vascularization and bone marrow development. This study provides valuable insights for applying ECO-mediated bone tissue engineering approaches for the repair of critical-sized bone defects.


Assuntos
Células-Tronco Mesenquimais , Osteogênese , Camundongos , Animais , Ácido Hialurônico/metabolismo , Hidrogéis/metabolismo , Camundongos Nus , Engenharia Tecidual/métodos , Colágeno/metabolismo , Condrogênese
4.
Biochem Biophys Res Commun ; 585: 89-95, 2021 12 31.
Artigo em Inglês | MEDLINE | ID: mdl-34801937

RESUMO

Osteosarcoma (OS) is the most common primary malignant bone tumor which has unclear pathobiology. Hence, enlightening the exact molecular mechanism underlying osteosarcoma progression is crucial for developing new treatment strategies. One member of the ARID family of DNA binding proteins is ARID3A that is implicated in osteosarcoma pathogenesis. ARID3A could bind E2F1 and regulate the transcription of E2F1 targets. At the same time, BECN1 is a well-characterized autophagy regulator gene that is a direct target of E2F1. The present study aimed to investigate the effect of ARID3A on the expression of BECN1 in osteosarcoma cells. First, we determined gene expression levels of ARID3A, BECN1, and E2F1 in U-2 OS by qPCR and confirmed with online datasets from GEO database. In addition, the prognostic value of these genes was also evaluated from Kaplan-Meier plotter database. Next, ARID3A was overexpressed and silenced in order to investigate the effect of ARID3A on BECN1 expression and proliferation of U-2 OS cells. Our results demonstrated that BECN1 was negatively correlated with E2F1 and positively correlated with ARID3A based on initial expression and prognostic effect in OS. Overexpression of ARID3A upregulated BECN1 while silenced ARID3A downregulated BECN1 expression in U-2 OS cells. Additionally, silencing of ARID3A promoted colony formation and proliferation, whereas overexpression of ARID3A suppressed colony formation and proliferation of U-2 OS cells. Taken together, these results indicate that ARID3A could function as tumor suppressor and affect the expression level of BECN1 in U-2 OS cells.


Assuntos
Autofagia/genética , Proteína Beclina-1/genética , Neoplasias Ósseas/genética , Proliferação de Células/genética , Proteínas de Ligação a DNA/genética , Regulação Neoplásica da Expressão Gênica , Osteossarcoma/genética , Fatores de Transcrição/genética , Sequência de Bases , Sítios de Ligação/genética , Neoplasias Ósseas/patologia , Linhagem Celular , Linhagem Celular Tumoral , Fator de Transcrição E2F1/genética , Fator de Transcrição E2F1/metabolismo , Perfilação da Expressão Gênica/métodos , Humanos , Osteossarcoma/patologia , Interferência de RNA , Reação em Cadeia da Polimerase Via Transcriptase Reversa
5.
Int J Oncol ; 58(4)2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33649863

RESUMO

The AT­rich interacting domain (ARID) family of DNA­binding proteins is involved in various biological processes, including the regulation of gene expression during cell proliferation, differentiation and development. ARID3A and ARID3B are involved in chromatin remodeling and can bind to E2F1 and retinoblastoma tumor suppressor protein (RB), respectively. However, their role in regulating E2F target gene expression remains poorly understood. E2F transcription factors are critical regulators of cell cycle progression and are modulated by RB. Herein, putative ARID3­binding sites (BSs) in E2F target genes were identified, including Cdc2, cyclin E1 and p107, and it was found that ARID3A and ARID3B bound to these BSs in living cells. The mutation of ARID3 BSs reduced Cdc2 promoter activity, while ARID3A and ARID3B overexpression increased the promoter activity, depending on both ARID3 and E2F BSs. ARID3B knockdown blocked the transcription of Cdc2, cyclin E1 and p107 in normal human dermal fibroblasts (NHDFs), whereas the effects of ARID3A knockdown varied depending on the target genes. ARID3B overexpression, but not that of ARID3A, upregulated the transcription of E2F target genes, and activated cyclin E1 transcription and induced cell death with E2F1 assistance. Finally, ARID3A and ARID3B knockdown attenuated the cell cycle progression of NHDFs and T98G cells, and suppressed tumor cell growth. On the whole, these results indicate that ARID3A and ARID3B play distinct and overlapping roles in E2F­dependent transcription by directly binding to the E2F target genes. The present study provides novel insight into the mechanisms underlying the E2F dysregulation caused by ARID3A and ARID3B overexpression, which may have a significant influence on the progression of tumorigenesis.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Fatores de Transcrição E2F/metabolismo , Expressão Gênica , Neoplasias/patologia , Regiões Promotoras Genéticas , Fatores de Transcrição/metabolismo , Sítios de Ligação , Linhagem Celular , Linhagem Celular Tumoral , Proteínas de Ligação a DNA/genética , Fatores de Transcrição E2F/genética , Humanos , Neoplasias/genética , Neoplasias/metabolismo , Fatores de Transcrição/genética
6.
Cell Biol Int ; 44(11): 2263-2274, 2020 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-32749762

RESUMO

Long noncoding RNA (lncRNA) dysregulation is known to be taking part in majority of cancers, including osteosarcoma. In one of our previous studies, we showed that lncRNA MEG3 is being regulated by microRNA-664a (miR-664a) suppresses the migratory potential of osteosarcoma cells (U-2OS). We now report a novel lncRNA, namely, ERICD, which is linked to the transcription factor AT-rich interaction domain 3A (ARID3A) in U-2OS cells. We show that ARID3A binds to ERICD and indirectly interacts with each other via the E2F transcription factor 1 (E2F1). Furthermore, small interfering RNA (siRNA)-mediated knockdown of ERICD inhibited cell migration, formation of colonies, and proliferation in U-2OS cells. Overexpression of ARID3A inhibited cell migration, colony formation, and proliferation, whereas siRNA-mediated knockdown of ARID3A promoted cell migration, colony formation, and proliferation. Our findings indicate that ARID3A and lncRNA ERICD have plausible tumor suppressive and oncogenic functions, respectively, in osteosarcoma. Our data demonstrate the converse interaction between ARID3A and lncRNA ERICD that target DNA-binding proteins and dysregulation of their expression through E2F1 augments osteosarcoma progression. The cell rescue experiment also indicated E2F1 to be involved in the regulation of ARID3A and ERICD.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Osteossarcoma/genética , RNA Longo não Codificante/genética , Fatores de Transcrição/metabolismo , Apoptose/genética , Neoplasias Ósseas/patologia , Linhagem Celular Tumoral , Movimento Celular/genética , Proliferação de Células/genética , Proteínas de Ligação a DNA/genética , Fator de Transcrição E2F1/genética , Fator de Transcrição E2F1/metabolismo , Regulação Neoplásica da Expressão Gênica/genética , Humanos , Osteossarcoma/metabolismo , Osteossarcoma/patologia , RNA Longo não Codificante/metabolismo , Fatores de Transcrição/genética
7.
Int J Oncol ; 50(6): 2043-2048, 2017 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-28440416

RESUMO

Fucose residues of cell surface glycans, which play important roles in growth, invasion and metastasis, are added by fucosyltransferases (FUTs) and removed by α-L-fucosidases (FUCAs). By the differential display method, we isolated a 3' non-coding region of α-L-fucosidase-1 (FUCA1) (a gene coding for the lysosomal fucosidase-1 enzyme) as a wild-type p53-inducible gene: 18S and 20S FUCA1 mRNA species were induced in Saos-2 cells transfected with a temperature-sensitive p53 mutant at the permissive temperature. By microarray analyses of thyroid cancer biopsy samples, FUCA1 RNA expression levels were found to be lower in anaplastic thyroid cancer samples (ATCs), while they were higher in papillary thyroid cancer samples (PTCs) and in normal thyroid tissues. Since most ATCs were reported to carry the mutated form of p53, while PTCs carry mostly the wild-type form of p53, it is likely that FUCA1 expression levels are regulated, at least in part, by the p53 status in thyroid cancers. In order to better understand the role played by FUCA genes in thyroid tumorigenesis, we examined the clonogenic potential in vitro of thyroid cell lines transfected with either FUCA1 or FUCA2 (the latter gene coding for a secreted, non-lysosomal enzyme). We found that α-L-fucosidases did not suppress grossly cell growth. Contrary to what we observed with the expression of FUCA1, the FUT8 expression levels were found high in ATCs but lower in PTCs and normal thyroid tissues. Taken together, these results suggest the possibility that the higher fucose levels on cell surface glycans of aggressive ATCs, compared to those of less aggressive PTCs, may be at least in part responsible for the more aggressive and metastatic phenotype of ATCs compared to PTCs, as the expression levels of FUCA1 and FUT8 were inversely related in these two types of cancers.


Assuntos
Carcinoma Papilar/genética , Fucosiltransferases/genética , Carcinoma Anaplásico da Tireoide/genética , Neoplasias da Glândula Tireoide/genética , Proteína Supressora de Tumor p53/genética , alfa-L-Fucosidase/genética , Carcinoma Papilar/patologia , Linhagem Celular Tumoral , Feminino , Fucose/metabolismo , Regulação Neoplásica da Expressão Gênica , Humanos , Masculino , Mutação , Metástase Neoplásica , Câncer Papilífero da Tireoide , Carcinoma Anaplásico da Tireoide/patologia , Glândula Tireoide/metabolismo , Glândula Tireoide/patologia , Neoplasias da Glândula Tireoide/patologia , Análise Serial de Tecidos
8.
Kokubyo Gakkai Zasshi ; 83(1): 13-24, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-27443069

RESUMO

Mesenchymal stem cells (MSCs) offer a promising source of cells for musculoskeletal regeneration because of their potential to differentiate into bone, cartilage and fat. However, their proliferation and multilineage differentiation potential decreases with aging or increased time in in vitro culture. To determine culture conditions capable of enabling maintenance of MSCs for extended periods of time, human bone marrow-derived MSCs (BM-MSCs) were cultured in growth medium containing various combinations of growth factors and small chemical compounds. Upon reaching confluence, MSCs were subcultured continuously and then tested for differentiation capacity. After screening various growth factors and small chemical compounds, we found a combination capable of maintaining the proliferation potential of BM-MSCs obtained from a 19-year-old donor (young MSCs) up to passage 13 (P13). In contrast, unsupplemented MSCs reached senescence at P10. Total population doublings of control (P10) and supplemented MSCs (P12) were estimated at 20.4 and 42, respectively. Young MSCs cultured with supplements maintained osteogenic, adipogenic and chondrogenic differentiation capacities at P12 as confirmed by expression of lineage-specific differentiation markers. Furthermore, the supplementation of to BM-MSCs obtained from 65- and 79-year-old donors (aged MSCs) also continued to proliferate until P12, and maintained osteogenic and adipogenic differentiation capacity until P7 and P8, respectively, whereas, unsupplemented aged MSCs stopped proliferating at P8. These results indicate that our extended culture conditions maintained the proliferative capacity of young MSCs while retaining their multipotent differentiation potential, and improved both proliferation and differentiation of aged MSCs.


Assuntos
Técnicas de Cultura de Células/métodos , Diferenciação Celular , Células-Tronco Mesenquimais/citologia , Adipogenia/efeitos dos fármacos , Adipogenia/genética , Idoso , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/genética , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/genética , Células Cultivadas , Condrogênese/efeitos dos fármacos , Condrogênese/genética , Meios de Cultura/farmacologia , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/farmacologia , Osteogênese/efeitos dos fármacos , Osteogênese/genética , Adulto Jovem
9.
Biochem Biophys Res Commun ; 468(1-2): 248-54, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26519881

RESUMO

ARID3A and ARID3B are transcriptional targets of p53. Recently, it has been reported that ARID3A plays a critical role in the transcriptional activation of pro-arrest p21 in response to DNA damage. However, the role of ARID3B in the p53 regulatory pathway remains poorly understood. Here we show that ARID3A and ARID3B specifically bind to putative ARID3-binding sites in p53 target genes in vitro and in vivo. ARID3B and, to a lesser extent, ARID3A silencing blocked transcriptional activation of pro-apoptotic p53 target genes, such as PUMA, PIG3, and p53. Furthermore, ectopic ARID3B, to a lesser extent, ARID3A expression activated the pro-apoptotic gene expression, and only ARID3B induced apoptosis. Finally, ARID3B but not ARID3A silencing blocked apoptosis induction following DNA damage. These results indicated that, although ARID3B and ARID3A share overlapping functions, ARID3B play a key role in the expression of pro-apoptotic p53-target genes and apoptosis.


Assuntos
Apoptose , Proteínas de Ligação a DNA/genética , Regulação da Expressão Gênica , Proteína Supressora de Tumor p53/genética , Linhagem Celular Tumoral , Inibidor de Quinase Dependente de Ciclina p21/genética , Dano ao DNA , Proteínas de Ligação a DNA/metabolismo , Inativação Gênica , Humanos , Regiões Promotoras Genéticas , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Ativação Transcricional , Proteína Supressora de Tumor p53/metabolismo
10.
Neurosci Lett ; 607: 90-96, 2015 Oct 21.
Artigo em Inglês | MEDLINE | ID: mdl-26391744

RESUMO

Cyclin E is a key regulator of progression through the G1-phase of the cell cycle. Recently, a cell cycle-independent role for cyclin E in the adult mouse central nervous system has been suggested. In the present study, we examined expression of cyclin E in the mouse hippocampal dentate gyrus (DG), a region of neurogenesis in adulthood, using immunofluorescence. In the adult DG, cyclin E-immunoreactive (cyclin E+) cells was limited to postmitotic cells. In the subgranular zone, cyclin E was detected in the vertical process of radial glia-like cells, which were marked by the neural stem cell markers nestin and GFAP. Cyclin E was also detected in the nucleus of cells, which were labeled with stage-specific neuronal cell markers, including Pax6, Sox2, NeuroD, doublecortin, and NeuN. The densities of cyclin E+ cells in the DG reduced and increased with age and running, respectively. Furthermore, the majority of cyclin E+ cells co-expressed active caspase-3, a marker of apoptosis. Together, the results indicate that cyclin E is expressed in the process of quiescent neural stem cells and in the nucleus of active caspase-3+ cells during neuronal cell differentiation, suggesting that cyclin E has a Cdk-independent function, which might be important for the mechanisms regulating adult hippocampal neurogenesis.


Assuntos
Caspase 3/metabolismo , Ciclina E/metabolismo , Hipocampo/citologia , Células-Tronco Neurais/citologia , Neurônios/citologia , Envelhecimento/metabolismo , Animais , Animais Recém-Nascidos , Apoptose , Biomarcadores/metabolismo , Diferenciação Celular , Linhagem da Célula , Núcleo Celular/metabolismo , Proliferação de Células , Embrião de Mamíferos , Hipocampo/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Células-Tronco Neurais/metabolismo , Neurogênese , Neuroglia/metabolismo , Neurônios/metabolismo , Corrida
11.
PLoS One ; 10(6): e0128090, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26086724

RESUMO

Using an asynchronously growing cell population, we investigated how X-irradiation at different stages of the cell cycle influences individual cell-based kinetics. To visualize the cell-cycle phase, we employed the fluorescent ubiquitination-based cell cycle indicator (Fucci). After 5 Gy irradiation, HeLa cells no longer entered M phase in an order determined by their previous stage of the cell cycle, primarily because green phase (S and G2) was less prolonged in cells irradiated during the red phase (G1) than in those irradiated during the green phase. Furthermore, prolongation of the green phase in cells irradiated during the red phase gradually increased as the irradiation timing approached late G1 phase. The results revealed that endoreduplication rarely occurs in this cell line under the conditions we studied. We next established a method for classifying the green phase into early S, mid S, late S, and G2 phases at the time of irradiation, and then attempted to estimate the duration of G2 arrest based on certain assumptions. The value was the largest when cells were irradiated in mid or late S phase and the smallest when they were irradiated in G1 phase. In this study, by closely following individual cells irradiated at different cell-cycle phases, we revealed for the first time the unique cell-cycle kinetics in HeLa cells that follow irradiation.


Assuntos
Ciclo Celular/efeitos da radiação , Raios X/efeitos adversos , Divisão Celular/efeitos da radiação , Relação Dose-Resposta à Radiação , Citometria de Fluxo , Fase G1/efeitos da radiação , Fase G2/efeitos da radiação , Células HeLa/efeitos da radiação , Humanos , Cinética , Microscopia de Fluorescência , Fase S/efeitos da radiação
12.
J Oral Sci ; 56(2): 119-26, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24930748

RESUMO

Chitosan, a multipurpose biomaterial, has been shown to exert effects against several types of cancer including oral cancer. However, the mechanisms underlying the anticancer activities of chitosan on oral squamous cell carcinoma (SCC) cells remain largely unknown. The present study aimed to compare the effects of low-molecular-weight chitosan (LMWC) and cisplatin on oral SCC Ca9-22 and non-cancer keratinocyte HaCaT cell lines. Cell viability and cell cycle profiles were measured by MTT assay and laser scanning cytometry, respectively. Apoptosis was examined by TUNEL assay and electron microscopy, followed by analysis of caspase activity. LMWC exhibited cytotoxic effects on Ca9-22, but not HaCaT cells, whereas cisplatin induced apoptosis in both types of cells. Exposure of Ca9-22 cells to LMWC led to G1/S cell cycle arrest and an increase of TUNEL-positive cells accompanied by an early apoptotic cell morphology and subtle increases of caspase activity. Short-term LMWC exposure was less cytotoxic to HaCaT cells than to Ca9-22 cells, and anticancer activity was exerted through induction of apoptosis and cell cycle arrest, suggesting that LMWC could be a promising natural anticancer agent with fewer side effects on normal cells.


Assuntos
Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Ciclo Celular/efeitos dos fármacos , Quitosana/farmacologia , Neoplasias Bucais/patologia , Linhagem Celular Tumoral , Humanos
13.
Nat Commun ; 3: 1000, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22893124

RESUMO

DNA damage as a result of environmental stress is recognized by sensor proteins that trigger repair mechanisms, or, if repair is unsuccessful, initiate apoptosis. Defects in DNA damage-induced apoptosis promote genomic instability and tumourigenesis. The protein ataxia-telangiectasia mutated (ATM) is activated by DNA double-strand breaks and regulates apoptosis via p53. Here we show that FOXO3 interacts with the ATM-Chk2-p53 complex, augments phosphorylation of the complex and induces the formation of nuclear foci in cells on DNA damage. FOXO3 is essential for DNA damage-induced apoptosis and conversely FOXO3 requires ATM, Chk2 and phosphorylated p53 isoforms to trigger apoptosis as a result of DNA damage. Under these conditions FOXO3 may also have a role in regulating chromatin retention of phosphorylated p53. These results suggest an essential link between FOXO3 and the ATM-Chk2-p53-mediated apoptotic programme following DNA damage.


Assuntos
Apoptose , Proteínas de Ciclo Celular/metabolismo , Dano ao DNA , Proteínas de Ligação a DNA/metabolismo , Fatores de Transcrição Forkhead/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Transdução de Sinais , Proteína Supressora de Tumor p53/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Proteínas Mutadas de Ataxia Telangiectasia , Proteínas de Ciclo Celular/genética , Linhagem Celular Tumoral , Quinase do Ponto de Checagem 2 , Cromatina/genética , Cromatina/metabolismo , Proteínas de Ligação a DNA/genética , Proteína Forkhead Box O3 , Fatores de Transcrição Forkhead/genética , Humanos , Fosforilação , Ligação Proteica , Proteínas Serina-Treonina Quinases/genética , Proteína Supressora de Tumor p53/genética , Proteínas Supressoras de Tumor/genética
14.
Biochem Biophys Res Commun ; 417(3): 931-7, 2012 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-22166212

RESUMO

The p53 tumor suppressor protein plays key roles in protecting cells from tumorigenesis. Phosphorylation of p53 at Ser46 (p53Ser46) is considered to be a crucial modification regulating p53-mediated apoptosis. Because the activity of p53 is impaired in most human cancers, restoration of wild-type p53 (wt-p53) function by its gene transfer or by p53-reactivating small molecules has been extensively investigated. The p53-reactivating compounds Nutlin-3 and RITA activate p53 in the absence of genotoxic stress by antagonizing the action of its negative regulator Mdm2. Although controversial, Nutlin-3 was shown to induce p53-mediated apoptosis in a manner independent of p53 phosphorylation. Recently, RITA was shown to induce apoptosis by promoting p53Ser46 phosphorylation. Here we examined whether Nutlin-3 or RITA can overcome resistance to p53-mediated apoptosis in p53-resistant tumor cell lines lacking the ability to phosphorylate p53Ser46. We show that Nutlin-3 did not rescue the apoptotic defect of a Ser46 phosphorylation-defective p53 mutant in p53-sensitive tumor cells, and that RITA neither restored p53Ser46 phosphorylation nor induced apoptosis in p53Ser46 phosphorylation-deficient cells retaining wt-p53. Furthermore, treatment with Nutlin-3 or RITA together with adenoviral p53 gene transfer also failed to induce apoptosis in p53Ser46 phosphorylation-deficient cells either expressing or lacking wt-p53. These results indicate that neither Nutlin-3 nor RITA in able to induce p53-mediated apoptosis in the absence of p53Ser46 phosphorylation. Thus, the dysregulation of this phosphorylation in tumor cells may be a critical factor that limits the efficacy of these p53-based cancer therapies.


Assuntos
Apoptose/efeitos dos fármacos , Furanos/farmacologia , Imidazóis/farmacologia , Piperazinas/farmacologia , Serina/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Apoptose/genética , Caspase 3 , Linhagem Celular Tumoral , Genes p53 , Humanos , Fosforilação/efeitos dos fármacos , Fosforilação/genética , Poli(ADP-Ribose) Polimerases/metabolismo , Serina/genética , Transdução Genética , Proteína Supressora de Tumor p53/genética
15.
Biochem Biophys Res Commun ; 417(2): 710-6, 2012 Jan 13.
Artigo em Inglês | MEDLINE | ID: mdl-22172947

RESUMO

ARID3A/DRIL1/Bright is a family member of the AT rich interaction domain (ARID) DNA-binding proteins that are involved in diverse biological processes. We have reported that p53 activates ARID3A transcription, and ARID3A overexpression induces G1 arrest. However, the role of ARID3A in the p53 pathway remains unclear. Here, we show that ARID3A cooperates with p53 to transcriptionally activate p21(WAF1), a p53-target gene important for cell-cycle arrest. ARID3A bound to its binding sites in the p21(WAF1) promoter in vivo and in vitro, and induced p21(WAF1) transcription in U2OS cells expressing wild-type p53 but not Saos-2 cells lacking p53. The co-expression of ARID3A with p53 cooperates to activate p21(WAF1) transcription and the stably transfected p21(WAF1) promoter. Mutation of the ARID3A binding sites reduced the p21(WAF1) promoter activity, and siRNA-based ARID3A knockdown suppressed the transcription of p21(WAF1), but not the proapoptotic NOXA and PUMA in response to DNA damage. Furthermore, p53 knockdown decreased ARID3A transcription, and, conversely, ARID3A overexpression and knockdown resulted in an increase or decrease in p53 stability, respectively. These results indicate both cooperative and interdependent roles for ARID3A and p53 in the transcriptional activation of p21(WAF1) in response to DNA damage.


Assuntos
Inibidor de Quinase Dependente de Ciclina p21/genética , Dano ao DNA , Proteínas de Ligação a DNA/metabolismo , Fatores de Transcrição/metabolismo , Ativação Transcricional , Proteína Supressora de Tumor p53/metabolismo , Linhagem Celular Tumoral , Proteínas de Ligação a DNA/genética , Técnicas de Silenciamento de Genes , Humanos , Regiões Promotoras Genéticas , Estabilidade Proteica , Fatores de Transcrição/genética , Proteína Supressora de Tumor p53/genética
16.
Mol Cancer ; 10: 95, 2011 Jul 31.
Artigo em Inglês | MEDLINE | ID: mdl-21801448

RESUMO

BACKGROUND: Caspase-8 is a key upstream mediator in death receptor-mediated apoptosis and also participates in mitochondria-mediated apoptosis via cleavage of proapoptotic Bid. However, the role of caspase-8 in p53- and p73-dependent apoptosis induced by genotoxic drugs remains unclear. We recently reported that the reconstitution of procaspase-8 is sufficient for sensitizing cisplatin- but not etoposide-induced apoptosis, in chemoresistant and caspase-8 deficient HOC313 head and neck squamous cell carcinoma (HNSCC) cells. RESULTS: We show that p53/p73-dependent caspase-8 activation is required for sensitizing etoposide-induced apoptosis by utilizing HOC313 cells carrying a temperature-sensitive p53G285K mutant. Restoration of wild-type p53 function under the permissive conditions, together with etoposide treatment, led to substantial transcriptional activation of proapoptotic Noxa and PUMA, but failed to induce apoptosis. In addition to p53 restoration, caspase-8 reconstitution was needed for sensitization to etoposide-induced apoptosis, mitochondria depolarization, and cleavage of the procaspases-3, and -9. In etoposide-sensitive Ca9-22 cells carrying a temperature-insensitive mutant p53, siRNA-based p73 knockdown blocked etoposide-induced apoptosis and procaspase-8 cleavage. However, induction of p73 protein and up-regulation of Noxa and PUMA, although observed in Ca9-22 cells, were hardly detected in etoposide-treated HOC313 cells under non-permissive conditions, suggesting a contribution of p73 reduction to etoposide resistance in HOC313 cells. Finally, the caspase-9 inhibitor Ac-LEHD-CHO or caspase-9 siRNA blocked etoposide-induced caspase-8 activation, Bid cleavage, and apoptosis in both cell lines, indicating that p53/p73-dependent caspase-8 activation lies downstream of mitochondria. CONCLUSIONS: we conclude that p53 and p73 can act as upstream regulators of caspase-8, and that caspase-8 is an essential mediator of the p53/p73-dependent apoptosis induced by etoposide in HNSCC cells. Our data suggest the importance of caspase-8-mediated positive feedback amplification in the p53/p73-dependent apoptosis induced by etoposide in HNSCC cells.


Assuntos
Apoptose/efeitos dos fármacos , Carcinoma de Células Escamosas/patologia , Caspase 8/fisiologia , Proteínas de Ligação a DNA/fisiologia , Etoposídeo/farmacologia , Neoplasias de Cabeça e Pescoço/patologia , Proteínas Nucleares/fisiologia , Proteína Supressora de Tumor p53/fisiologia , Proteínas Supressoras de Tumor/fisiologia , Antineoplásicos Fitogênicos/farmacologia , Apoptose/genética , Carcinoma de Células Escamosas/tratamento farmacológico , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/metabolismo , Caspase 8/genética , Caspase 8/metabolismo , Linhagem Celular Tumoral , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Avaliação Pré-Clínica de Medicamentos , Ativação Enzimática/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Neoplasias de Cabeça e Pescoço/tratamento farmacológico , Neoplasias de Cabeça e Pescoço/genética , Neoplasias de Cabeça e Pescoço/metabolismo , Humanos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Mitocôndrias/patologia , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , RNA Interferente Pequeno/farmacologia , Carcinoma de Células Escamosas de Cabeça e Pescoço , Transfecção , Proteína Tumoral p73 , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/metabolismo
17.
Gene Expr Patterns ; 11(1-2): 64-71, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-20863901

RESUMO

Cyclin E, a member of the G1 cyclins, is essential for the G1/S transition of the cell cycle in cultured cells, but its roles in vivo are not fully defined. The present study characterized the spatiotemporal expression profile of cyclin E in two representative brain regions in the mouse, the cerebral and cerebellar cortices. Western blotting showed that the levels of cyclin E increased towards adulthood. In situ hybridization and immunohistochemistry showed the distributions of cyclin E mRNA and protein were comparable in the cerebral cortex and the cerebellum. Immunohistochemistry for the proliferating cell marker, proliferating cell nuclear antigen (PCNA) revealed that cyclin E was expressed by both proliferating and non-proliferating cells in the cerebral cortex at embryonic day 12.5 (E12.5) and in the cerebellum at postnatal day 1 (P1). Subcellular localization in neurons was examined using immunofluorescence and western blotting. Cyclin E expression was nuclear in proliferating neuronal precursor cells but cytoplasmic in postmitotic neurons during embryonic development. Nuclear cyclin E expression in neurons remained faint in newborns, increased during postnatal development and was markedly decreased in adults. In various adult brain regions, cyclin E staining was more intense in the cytoplasm than in the nucleus in most neurons. These data suggest a role for cyclin E in the development and function of the mammalian central nervous system and that its subcellular localization in neurons is important. Our report presents the first detailed analysis of cyclin E expression in postmitotic neurons during development and in the adult mouse brain.


Assuntos
Cerebelo/crescimento & desenvolvimento , Cérebro/crescimento & desenvolvimento , Ciclina E/análise , Ciclina E/genética , Animais , Ciclo Celular , Proliferação de Células , Cerebelo/química , Cerebelo/citologia , Cerebelo/metabolismo , Cérebro/química , Cérebro/citologia , Cérebro/metabolismo , Imuno-Histoquímica , Hibridização In Situ , Camundongos , Neurônios/citologia , Neurônios/metabolismo , Células de Purkinje/citologia , Células de Purkinje/metabolismo
18.
Biochem Biophys Res Commun ; 390(3): 989-94, 2009 Dec 18.
Artigo em Inglês | MEDLINE | ID: mdl-19852930

RESUMO

Caspase-8 is a critical upstream mediator of apoptosis in the death receptor pathway. However, the relationship between caspase-8 mutation and chemosensitivity remain unclear in head and neck squamous cell carcinoma (HNSCC) carrying p53 mutation. In this study, we identified a caspase-8 nonsense mutation, accompanied by the loss of the second allele, in a drug-resistant HOC313 HNSCC cell line. The nonsense mutation (R68X) leads to truncation of all defined functional domains. Reconstitution of caspase-8 by stable transfection of wild-type caspase-8 sensitized the cells to cisplatin-, but not etoposide-induced apoptosis. Consistent with this, cisplatin, but not etoposide, induced TNF-alpha and TRAIL mRNA in caspase-8 reconstituted HOC313 cells, accompanied by activation of the reconstituted caspase-8 and its downstream caspase-3. These results indicate that the loss of caspase-8 plays an important role in acquisition of chemoresistance to cisplatin in HOC313 cells.


Assuntos
Antineoplásicos/farmacologia , Carcinoma de Células Escamosas/enzimologia , Caspase 8/genética , Cisplatino/farmacologia , Resistencia a Medicamentos Antineoplásicos/genética , Neoplasias de Cabeça e Pescoço/enzimologia , Linhagem Celular Tumoral , Códon sem Sentido , Humanos , Receptores de Morte Celular/agonistas , Receptores de Morte Celular/metabolismo , Proteína Supressora de Tumor p53/genética
19.
Oral Oncol ; 45(10): e161-6, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19628422

RESUMO

Oral squamous cell carcinoma is the sixth most common cancer in the world and the seventh most common cancer in Vietnam. The RAS and PI3K-AKT signaling pathways play an important role in oral carcinogenesis. Our previous study on PI3K signaling pathway showed the absence of PIK3CA and PTEN gene mutations in Vietnamese oral cancer. We thus hypothesized that the RAS could be more likely activated as an upstream effector. However, the status of RAS mutations in Vietnamese oral cancer had not been studied. In the present study, Fifty six primary tumor DNA samples were screened for mutations of hot spots in exons 1 and 2 of H-RAS and a part of the samples for exon 7 of ERK2 gene in which we previously reported a mutation in an OSCC cell line. The H-RAS mutations were detected in 10 of 56 tumors (18%). Two novel mutations were found, one was an insertion of three nucleotides (GGC) between codons 10 and 11 resulting in in-frame insertion of glycine (10(Gly)11) and the other was a missense mutation in codon 62 (GAG>GGG). We also found T81C single nucleotide polymorphism in 12 of 56 tumors (22%) and there was no mutation in exon 7 of ERK2 gene. The H-RAS mutation incidence showed significant association with advanced stages of the tumor and also with well-differentiated tumor grade. Our study is the first to report H-RAS mutation from Vietnamese ethnicity, with two novel mutations and relatively high incidence of H-RAS mutations. The results suggest that RAS is an important member in the PI3K-AKT signaling and could play an important role in the tumorigenesis of oral carcinoma.


Assuntos
Carcinoma de Células Escamosas/genética , Genes ras/genética , Neoplasias Bucais/genética , Mutação/genética , Adulto , Idoso , Idoso de 80 Anos ou mais , Códon , Éxons , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Proteína Quinase 1 Ativada por Mitógeno/genética , Mutação de Sentido Incorreto/genética , Polimorfismo de Nucleotídeo Único , Fatores de Risco , Vietnã
20.
Genes Cells ; 14(1): 89-99, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19077036

RESUMO

The transcription factor E2F, the main target of the RB tumor suppressor pathway, plays crucial roles not only in cell proliferation but also in tumor suppression. The cyclin-dependent kinase inhibitor p27(Kip1) gene, an upstream negative regulator of E2F, is induced by ectopically expressed E2F1 but not by normal growth stimulation that physiologically activates endogenous E2F. This suggests that the gene can discriminate between deregulated and physiological E2F activity. To address this issue, we examined regulation of the p27(Kip1) gene by E2F. Here we show that p27(Kip1) promoter specifically senses deregulated E2F activity through elements similar to typical E2F sites. This E2F-like elements were activated by deregulated E2F activity induced by forced inactivation of pRb but not by physiological E2F activity induced by serum stimulation, contrary to typical E2F sites activated by both E2F activity. The endogenous p27(Kip1) gene responded to deregulated and physiological E2F activity in the same manner to the E2F-like elements. Moreover, the E2F-like elements bound ectopically expressed E2F1 but not physiologically activated E2F1 or E2F4 in vivo. These results suggest that the p27(Kip1) gene specifically senses deregulated E2F activity through the E2F-like elements to suppress inappropriate cell cycle progression in response to loss of pRb function.


Assuntos
Inibidor de Quinase Dependente de Ciclina p27/genética , Fator de Transcrição E2F1/genética , Regulação da Expressão Gênica , Regiões Promotoras Genéticas/genética , Linhagem Celular Tumoral , Fator de Transcrição E2F4/metabolismo , Humanos , Ligação Proteica , Elementos de Resposta/genética , Proteína do Retinoblastoma/metabolismo , Soro/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...