Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
1.
JCI Insight ; 6(7)2021 04 08.
Artigo em Inglês | MEDLINE | ID: mdl-33830081

RESUMO

Lung cancer with oncogenic KRAS makes up a significant proportion of lung cancers and is accompanied by a poor prognosis. Recent advances in understanding the molecular pathogenesis of lung cancer with oncogenic KRAS have enabled the development of drugs, yet mutated KRAS remains undruggable. We performed small-molecule library screening and identified verteporfin, a yes-associated protein 1 (YAP1) inhibitor; verteporfin treatment markedly reduced cell viability in KRAS-mutant lung cancer cells in vitro and suppressed KRAS-driven lung tumorigenesis in vivo. Comparative functional analysis of verteporfin treatment and YAP1 knockdown with siRNA revealed that the cytotoxic effect of verteporfin was at least partially independent of YAP1 inhibition. A whole-transcriptome approach revealed the distinct expression profiles in KRAS-mutant lung cancer cells between verteporfin treatment and YAP1 knockdown and identified the selective involvement of the ER stress pathway in the effects of verteporfin treatment in KRAS-mutant lung cancer, leading to apoptotic cell death. These data provide novel insight to uncover vulnerabilities in KRAS-driven lung tumorigenesis.


Assuntos
Estresse do Retículo Endoplasmático/fisiologia , Neoplasias Pulmonares/patologia , Proteínas Proto-Oncogênicas p21(ras)/genética , Verteporfina/farmacologia , Animais , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Apoptose/fisiologia , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/genética , Camundongos Endogâmicos BALB C , Mutação , Proteínas Proto-Oncogênicas p21(ras)/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto , Proteínas de Sinalização YAP/genética
2.
Biochem Biophys Res Commun ; 525(2): 483-490, 2020 04 30.
Artigo em Inglês | MEDLINE | ID: mdl-32111352

RESUMO

The oncogenic Ras mutation is one of the most common genomic abnormalities having the highest incidence in cancer; it has three isoforms: Hras, Kras, and Nras. Although the Ras isoforms are highly similar in the primary sequence, each mutational frequency is clearly distinct according to tissue- or cell-type. Regarding non-small-cell lung carcinoma, almost all Kras mutations have been detected in lung adenocarcinoma, whereas lung squamous cell carcinoma is extremely rare. Here, we focus on the cell-type specific tumorigenesis of mutant Ras isoforms and determine the mechanisms of oncogenic signaling outputs between lung adenocarcinoma and squamous cell carcinoma. An in vitro transformation model with mutant Ras isoforms in immortalized bronchial epithelial cells (BEC-E6E7/myc) and immortalized small airway epithelial cells (SAEC-E6E7/myc) revealed that only the HrasG12V mutation, not the KrasG12V mutation, could induce tumorigenesis in BEC-E6E7/myc. In contrast, SAEC-E6E7/myc showed high sensitivity to the KrasG12V mutation compared with the HrasG12V mutation. The transformation of BEC-E6E7/myc and SAEC-E6E7/myc with mutant Ras isoforms was confirmed by soft agar assay and migration assay. HrasG12V-expressing BEC-E6E7/myc significantly increased MAPK/ERK signaling, whereas PI3K/AKT signaling was significantly elevated in KrasG12V-expressing SAEC-E6E7/myc. These results suggest a context dependency with oncogenic Ras mutations in tumorigenesis between lung adenocarcinoma and squamous cell carcinoma.


Assuntos
Carcinogênese/genética , Neoplasias Pulmonares/genética , Proteínas Proto-Oncogênicas p21(ras)/genética , Adenocarcinoma de Pulmão/genética , Carcinoma de Células Escamosas/genética , Linhagem Celular , Transformação Celular Neoplásica/genética , Genes ras , Humanos , Mutação , Mucosa Respiratória/citologia , Mucosa Respiratória/metabolismo
3.
Cancer Sci ; 111(2): 395-405, 2020 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-31854049

RESUMO

The onset of breast cancer among young patients is a major issue in cancer etiology. Our previous study has shown that poor prognosis in young women with breast cancer is associated with lower expression of the microRNA miR-1285-5p. In this study, we showed that the expression of miR-1285-5p is lower in tumor tissues than in normal tissues. Accumulating evidence suggests that miR-1285-5p plays critical roles in various types of cancers. However, the functional role of miR-1285-5p in breast cancer remains to be elucidated. Here, we showed the tumor-suppressive role of miR-1285-5p and detailed its mechanism of action in breast cancer. Overexpression of miR-1285-5p significantly inhibited cell proliferation in breast cancer cells regardless of the tumor subtype. Among the target genes of miR-1285-5p, we found that transmembrane protein 194A (TMEM194A) was directly regulated by miR-1285-5p. Notably, separation of centrosomes from the nuclear envelope was observed upon knockdown of TMEM194A or overexpression of miR-1285-5p. In conclusion, our findings show that miR-1285-5p is a tumor suppressor via TMEM194A inhibition in breast cancer.


Assuntos
Neoplasias da Mama/genética , MicroRNAs/genética , Proteínas Nucleares/genética , Proteína ran de Ligação ao GTP/genética , Regiões 3' não Traduzidas , Neoplasias da Mama/metabolismo , Linhagem Celular Tumoral , Núcleo Celular/metabolismo , Proliferação de Células , Centrossomo/metabolismo , Regulação para Baixo , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Células MCF-7 , Proteínas Nucleares/metabolismo , Proteína ran de Ligação ao GTP/metabolismo
4.
Oncogene ; 38(28): 5566-5579, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-31147602

RESUMO

Cancer-associated fibroblasts (CAFs), one of the major components of a tumour microenvironment, comprise heterogeneous populations involved in tumour progression. However, it remains obscure how CAF heterogeneity is governed by cancer cells. Here, we show that cancer extracellular vesicles (EVs) induce a series of chemokines in activated fibroblasts and contribute to the formation of the heterogeneity. In a xenograft model of diffuse-type gastric cancer, we showed two distinct fibroblast subpopulations with alpha-smooth muscle actin (α-SMA) expression or chemokine expression. MicroRNAs (miRNAs) profiling of the EVs and the transfection experiment suggested that several miRNAs played a role in the induction of chemokines such as CXCL1 and CXCL8 in fibroblasts, but not for the myofibroblastic differentiation. Clinically, aberrant activation of CXCL1 and CXCL8 in CAFs correlated with poorer survival in gastric cancer patients. Thus, this link between chemokine expression in CAFs and tumour progression may provide novel targets for anticancer therapy.


Assuntos
Fibroblastos Associados a Câncer/metabolismo , Quimiocina CXCL1/biossíntese , Vesículas Extracelulares/metabolismo , Interleucina-8/biossíntese , Neoplasias Gástricas/patologia , Células Estromais/patologia , Actinas/metabolismo , Animais , Linhagem Celular Tumoral , Xenoenxertos , Humanos , Camundongos , Neoplasias Gástricas/metabolismo , Microambiente Tumoral
5.
Cancer Lett ; 451: 11-22, 2019 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-30862488

RESUMO

KRAS is one of the most frequently mutated oncogenes in human non-small cell lung cancer (NSCLC). Mutations in KRAS are detected in 30% of NSCLC cases, with most of them occurring in codons 12 and 13 and less commonly in others. Despite intense efforts to develop drugs targeting mutant KRAS, no effective therapeutic strategies have been successfully tested in clinical trials. Here, we investigated molecular targets for KRAS-activated lung cancer cells using a drug library. A total of 1271 small molecules were screened in KRAS-mutant and wild-type lung cancer cell lines. The screening identified the cytotoxic effects of benzimidazole derivatives on KRAS-mutant lung cancer cells. Treatments with two benzimidazole derivatives, methiazole and fenbendazole-both of which are structurally specific-yielded significant suppression of the RAS-related signaling pathways in KRAS-mutated cells. Moreover, combinatorial therapy with methiazole and trametinib, a MEK inhibitor, induced synergistic effects in KRAS-mutant lung cancer cells. Our study demonstrates that these benzimidazole derivatives play an important role in suppressing KRAS-mutant lung cancer cells, thus offering a novel combinatorial therapeutic approach against such cancer cells.


Assuntos
Antineoplásicos/farmacologia , Benzimidazóis/farmacologia , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Genes ras , Neoplasias Pulmonares/tratamento farmacológico , Mutação , Antineoplásicos/química , Benzimidazóis/química , Carcinoma Pulmonar de Células não Pequenas/genética , Linhagem Celular Tumoral , Humanos , Neoplasias Pulmonares/genética , Transdução de Sinais/efeitos dos fármacos
6.
Cancer Sci ; 109(5): 1455-1467, 2018 May.
Artigo em Inglês | MEDLINE | ID: mdl-29498789

RESUMO

Despite considerable research on K-Ras inhibitors, none had been established until now. We synthesized nuclease-resistant synthetic miR-143 (miR-143#12), which strongly silenced K-Ras, its effector signal molecules AKT and ERK, and the K-Ras activator Sos1. We examined the anti-proliferative effect of miR-143#12 and the mechanism in human colon cancer DLD-1 cell (G13D) and other cell types harboring K-Ras mutations. Cell growth was markedly suppressed in a concentration-dependent manner by miR-143#12 (IC50 : 1.32 nmol L-1 ) with a decrease in the K-Ras mRNA level. Interestingly, this mRNA level was also downregulated by either a PI3K/AKT or MEK inhibitor, which indicates a positive circuit of K-Ras mRNA expression. MiR-143#12 silenced cytoplasmic K-Ras mRNA expression and impaired the positive circuit by directly targeting AKT and ERK mRNA. Combination treatment with miR-143#12 and a low-dose EGFR inhibitor induced a synergistic inhibition of growth with a marked inactivation of both PI3K/AKT and MAPK/ERK signaling pathways. However, silencing K-Ras by siR-KRas instead of miR-143#12 did not induce this synergism through the combined treatment with the EGFR inhibitor. Thus, miR-143#12 perturbed the K-Ras expression system and K-Ras activation by silencing Sos1 and, resultantly, restored the efficacy of the EGFR inhibitors. The in vivo results also supported those of the in vitro experiments. The extremely potent miR-143#12 enabled us to understand K-Ras signaling networks and shut them down by combination treatment with this miRNA and EGFR inhibitor in K-Ras-driven colon cancer cell lines.


Assuntos
Neoplasias do Colo/tratamento farmacológico , MicroRNAs/administração & dosagem , MicroRNAs/síntese química , Inibidores de Proteínas Quinases/administração & dosagem , Proteínas Proto-Oncogênicas p21(ras)/genética , Animais , Benzimidazóis/administração & dosagem , Benzimidazóis/farmacologia , Benzotiazóis/administração & dosagem , Benzotiazóis/farmacologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Neoplasias do Colo/genética , Sinergismo Farmacológico , Flavonoides/administração & dosagem , Flavonoides/farmacologia , Células HT29 , Humanos , Camundongos , MicroRNAs/antagonistas & inibidores , MicroRNAs/química , MicroRNAs/farmacologia , Mutação , Transplante de Neoplasias , Inibidores de Proteínas Quinases/farmacologia , Transdução de Sinais/efeitos dos fármacos
7.
J Immunol ; 199(4): 1505-1515, 2017 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-28696255

RESUMO

Macrophages are polarized into functional classically activated and alternatively activated (M2) phenotypes depending on their microenvironment, and these cells play an important role in the immune system. M2-like polarization of tumor-associated macrophages (TAMs) is activated by various secretions from cancer cells; however, the interaction between cancer cells and TAMs is not well understood. Recent studies showed that cancer cell-derived extracellular vesicles (EVs) contribute to tumor development and modulation of the tumor microenvironment. In the current study, we investigated colorectal cancer-derived EVs containing miR-145 with respect to the polarization of TAMs. Colorectal cancer cells positively secreted miR-145 via EVs, which were taken up by macrophage-like cells. Interestingly, colorectal cancer-derived EVs polarized macrophage-like cells into the M2-like phenotype through the downregulation of histone deacetylase 11 An in vivo study showed that EV-treated macrophages caused significant enlargement of the tumor volumes. These findings suggest that colorectal cancer cells use miR-145 within EVs to efficiently modulate M2-like macrophage polarization and tumor progression.


Assuntos
Neoplasias Colorretais/imunologia , Vesículas Extracelulares/fisiologia , Macrófagos/imunologia , MicroRNAs/metabolismo , Microambiente Tumoral/imunologia , Animais , Linhagem Celular Tumoral , Neoplasias Colorretais/genética , Regulação para Baixo , Vesículas Extracelulares/genética , Histona Desacetilases/genética , Histona Desacetilases/metabolismo , Humanos , Ativação de Macrófagos , Camundongos , Camundongos Nus , MicroRNAs/genética , Fenótipo , Microambiente Tumoral/genética
8.
Int J Mol Sci ; 18(1)2017 Jan 17.
Artigo em Inglês | MEDLINE | ID: mdl-28106737

RESUMO

Bladder cancer is one of the most difficult malignancies to control. We explored the use of a novel RNA-interference method for a driver oncogene regulating cancer specific energy metabolism by the combination treatment with a small interfering RNA (siRNA) and a microRNA. After transfection of T24 and 253JB-V cells with miR-145 and/or siR-PTBP1, we examined the effects of cell growth and gene expression by performing the trypan blue dye exclusion test, Western blot, Hoechst 33342 staining, reverse transcription polymerase chain reaction (RT-PCR), and electron microscopy. The anti-cancer effects of xenograft model mice with miR-145 and/or siR-PTBP1 were then assessed. The combination treatment induced the deeper and longer growth inhibition and reduced the levels of both mRNA and protein expression of c-Myc and polypyrimidine tract-binding protein 1 (PTBP1) more than each single treatment. Notably, the combination treatment not only impaired the cancer specific energy metabolism by inhibiting c-Myc/PTBP1/PKMs axis but also inactivated MAPK/ERK and PI3K/AKT pathways examined in vitro and in vivo. Furthermore, the combination treatment induced apoptosis or autophagy; but, in some cells, apoptotic cell death was accompanied by autophagy, because the condensation of chromatin and many autophagosomes were coexistent. This combination treatment could be a novel RNA-interference strategy through the systemic silencing of the Warburg effect-promoting driver oncogene PTBP1 in bladder cancer cells.


Assuntos
Apoptose/genética , Glicólise/genética , Ribonucleoproteínas Nucleares Heterogêneas/genética , MicroRNAs/genética , Proteína de Ligação a Regiões Ricas em Polipirimidinas/genética , Interferência de RNA , Neoplasias da Bexiga Urinária/genética , Idoso , Idoso de 80 Anos ou mais , Animais , Western Blotting , Linhagem Celular Tumoral , Proliferação de Células/genética , Feminino , Regulação Neoplásica da Expressão Gênica , Ribonucleoproteínas Nucleares Heterogêneas/metabolismo , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Pessoa de Meia-Idade , Modelos Genéticos , Proteína de Ligação a Regiões Ricas em Polipirimidinas/metabolismo , Terapêutica com RNAi/métodos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/genética , Neoplasias da Bexiga Urinária/metabolismo , Neoplasias da Bexiga Urinária/terapia , Ensaios Antitumorais Modelo de Xenoenxerto/métodos
9.
Int J Mol Sci ; 17(11)2016 Nov 05.
Artigo em Inglês | MEDLINE | ID: mdl-27827955

RESUMO

Dickkopf-related protein 3 (Dkk-3) is a potential tumor suppressor reported in various cancer entities. However, we found that Dkk-3 was exceptionally upregulated in bladder cancer T24 cells. To validate the biological role of Dkk-3 other than a tumor suppressor, we examined the function of Dkk-3 in T24 cells. Gene silencing of Dkk-3 inhibited cell growth through inducing G0/G1 cell-cycle arrest. Furthermore, Dkk-3 knock-down caused macropinocytosis accompanied by autophagy, which were canceled in part by their inhibitors 5-(N-ethyl-N-isopropyl) amiloride (EIPA) and 3-methyladenine (3-MA). The macropinocytosis was induced by the Dkk-3 knock-down when there were sufficient extracellular nutrients. On the other hand, when the nutritional condition was poor, the autophagy was mainly induced by the Dkk-3 knock-down. These data indicated that Dkk-3 has a role in modulating macropinocytotic and autophagic pathways, a distinct function other than a Wnt antagonist.


Assuntos
Células Epiteliais/metabolismo , Pontos de Checagem da Fase G1 do Ciclo Celular/genética , Regulação Neoplásica da Expressão Gênica , Peptídeos e Proteínas de Sinalização Intercelular/genética , Pinocitose/genética , Proteínas Adaptadoras de Transdução de Sinal , Adenina/análogos & derivados , Adenina/farmacologia , Amilorida/análogos & derivados , Amilorida/farmacologia , Autofagia/efeitos dos fármacos , Autofagia/genética , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Quimiocinas , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/patologia , Pontos de Checagem da Fase G1 do Ciclo Celular/efeitos dos fármacos , Inativação Gênica/efeitos dos fármacos , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Proteínas de Membrana Lisossomal/genética , Proteínas de Membrana Lisossomal/metabolismo , MAP Quinase Quinase 4/genética , MAP Quinase Quinase 4/metabolismo , MicroRNAs/genética , MicroRNAs/metabolismo , Proteína Quinase 1 Ativada por Mitógeno/genética , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/genética , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Pinocitose/efeitos dos fármacos , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Transdução de Sinais , Bexiga Urinária/metabolismo , Bexiga Urinária/patologia , Proteínas Quinases p38 Ativadas por Mitógeno/genética , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Proteínas rab5 de Ligação ao GTP/genética , Proteínas rab5 de Ligação ao GTP/metabolismo , Proteínas rac1 de Ligação ao GTP/genética , Proteínas rac1 de Ligação ao GTP/metabolismo
10.
Exp Cell Res ; 347(1): 133-142, 2016 09 10.
Artigo em Inglês | MEDLINE | ID: mdl-27453209

RESUMO

Tumor necrosis-factor (TNF)-related apoptosis-inducing ligand (TRAIL) is a member of the TNF-superfamily that selectively induces apoptosis through death receptors (DRs) 4 and/ or DR5 in cancer cells, without affecting normal cells. Unfortunately, many clinical studies have shown that cancer cells acquire TRAIL-resistance and thus avoid TRAIL-induced apoptosis. In the current study, we newly found that PTBP1, a splicer protein that plays an important role in energy metabolism is highly expressed in TRAIL-resistant human colon cancer DLD-1. Interestingly, silencing PTBP1 by using siRNA for PTBP1 (siR-PTBP1) resulted in a significant increase in TRAIL-sensitivity along with the switching of pyruvate kinase muscle (PKM) isoforms from PKM2 to PKM1, leading to impaired Warburg effect, because the intracellular ATP levels were significantly increased and the production of lactate decreased. Notably, siR-PTBP1 canceled the resistance by increasing the expression level of DR5 and effectively inducing the translocation of DR5 to the cell surface membrane. Also, siR-PTBP1 up-regulated the expression level of CCN1, which contributed to the enhanced sensitivity to TRAIL-induced apoptosis. These findings indicate that silencing PTBP1, thus impairing the Warburg effect positively affected TRAIL-induced apoptosis and that this splicer protein may thus serve as a possible target molecule to cancel the resistance of cancer cells to TRAIL.


Assuntos
Glicólise/efeitos dos fármacos , Neoplasias/patologia , Ligante Indutor de Apoptose Relacionado a TNF/farmacologia , Acetilcisteína/farmacologia , Morte Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Proliferação de Células/efeitos dos fármacos , Proteína Rica em Cisteína 61/metabolismo , Técnicas de Silenciamento de Genes , Inativação Gênica/efeitos dos fármacos , Ribonucleoproteínas Nucleares Heterogêneas/metabolismo , Humanos , Proteína de Ligação a Regiões Ricas em Polipirimidinas/metabolismo , Multimerização Proteica/efeitos dos fármacos , RNA Interferente Pequeno/metabolismo , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Regulação para Cima/efeitos dos fármacos
11.
Oncotarget ; 7(19): 27033-43, 2016 May 10.
Artigo em Inglês | MEDLINE | ID: mdl-27081032

RESUMO

Emerging studies on tumor cell-derived extracellular vesicles (EVs) have shown the biological significance in tumor development and microenvironment through reprogramming immune cells around cancer cells. In this study, we used colorectal cancer cells as EVs donor, and T cells as recipients to examine whether EVs impair the T cell function. As a result, we found that colorectal cancer cell-derived EVs (CRC-EVs) were enriched with TGF-ß1. Interestingly, CRC-EVs induced phenotypic alteration of the T cells to Treg-like cells through activating TGF-ß/Smad signaling and inactivating SAPK signaling. Furthermore, the CRC-EVs-induced-Treg-like cells had a remarkable tumor-growth promoting activity in vitro and in vivo. These results suggest that colorectal cancer cells utilize EVs to tame immune cells for their prosperity.


Assuntos
Neoplasias Colorretais/imunologia , Vesículas Extracelulares/imunologia , Linfócitos T Reguladores/imunologia , Linfócitos T/imunologia , Fator de Crescimento Transformador beta1/imunologia , Animais , Western Blotting , Linhagem Celular Tumoral , Proliferação de Células/genética , Células Cultivadas , Neoplasias Colorretais/genética , Neoplasias Colorretais/metabolismo , Vesículas Extracelulares/genética , Vesículas Extracelulares/metabolismo , Feminino , Humanos , Células Jurkat , Camundongos Nus , Interferência de RNA , Transdução de Sinais/genética , Transdução de Sinais/imunologia , Linfócitos T/metabolismo , Linfócitos T Reguladores/metabolismo , Fator de Crescimento Transformador beta1/genética , Fator de Crescimento Transformador beta1/metabolismo , Transplante Heterólogo
12.
Biochem Biophys Res Commun ; 473(1): 174-180, 2016 Apr 22.
Artigo em Inglês | MEDLINE | ID: mdl-27012213

RESUMO

Resistance to chemotherapy is a crucial problem in the clinical situation. To overcome this issue, many mechanisms of chemoresistance have been elucidated so far. However, this problem still has not been solved completely. In this study, we investigated the mechanism of chemoresistance from the view of cancer metabolism-related genes, especially focusing on the expression profile of pyruvate kinase muscle (PKM) isoforms, which are rate-limiting enzymes in cancer-specific metabolism (Warburg effect). Herein, we showed that PKM1, which promotes oxidative phosphorylation (OXPHOS), was commonly up-regulated in various chemoresistant cells. To clarify the functions of PKM1 in chemoresistance, we investigated effects of PKM1 expression in DLD-1 parental, 5-FU-resistant and oxaliplatin-resistant DLD-1 cells. The overexpression of PKM1 resulted in resistance of the parental cells to 5-FU and oxaliplatin. Moreover, gene-silencing of PKM1 induced apoptosis in these cells including the resistant cells by causing a decrease in the mitochondrial membrane potential. Furthermore, combination therapy using 5-FU or oxaliplatin with siR-PKM1 was also effective against the resistant cells. Our findings should lead to the development of new agents that can cancel the chemoresistance from the view of cancer energy metabolism.


Assuntos
Antineoplásicos/química , Proteínas de Transporte/metabolismo , Resistencia a Medicamentos Antineoplásicos , Proteínas de Membrana/metabolismo , Neoplasias/tratamento farmacológico , Neoplasias/metabolismo , Hormônios Tireóideos/metabolismo , Apoptose , Linhagem Celular Tumoral , Fluoruracila/química , Regulação Neoplásica da Expressão Gênica , Glicólise/efeitos dos fármacos , Humanos , Concentração Inibidora 50 , Células K562 , Compostos Organoplatínicos/química , Oxaliplatina , Fosforilação Oxidativa , Fenótipo , Fase de Repouso do Ciclo Celular , Transfecção , Proteínas de Ligação a Hormônio da Tireoide
13.
Oncotarget ; 7(14): 18940-52, 2016 Apr 05.
Artigo em Inglês | MEDLINE | ID: mdl-26980745

RESUMO

It is known that pyruvate kinase in muscle (PKM), which is a rate-limiting glycolytic enzyme, has essential roles in the Warburg effect and that expression of cancer-dominant PKM2 is increased by polypyrimidine tract-binding protein 1 (PTBP1), which is a splicer of the PKM gene. In other words, PKM2 acts as a promoter of the Warburg effect. Previously, we demonstrated that the Warburg effect was partially established by down-regulation of several microRNAs (miRs) that bind to PTBP1 and that ectopic expression of these miRs suppressed the Warburg effect. In this study, we investigated the functions of miR-1 and -133b, which are well known as muscle-specific miRs, from the viewpoint of the Warburg effect in colorectal tumors. The expression levels of miR-1 and -133b were relatively high in colon tissue except muscle and very frequently down-regulated in 75 clinical colorectal tumors samples, even in adenomas, compared with those of the adjacent normal tissue samples. The ectopic expression of these miRs induced growth suppression and autophagic cell death through the switching of PKM isoform expression from PKM2 to PKM1 by silencing PTBP1 expression both in vitro and in vivo. Also, we showed that the resultant increase in the intracellular level of reactive oxygen species (ROS) was involved in this mechanism. Furthermore, PTBP1 was highly expressed in most of the 30 clinical colorectal tumor samples examined, even in adenomas. Our results suggested that PTBP1 and PTBP1-associated miR-1 and -133b are crucial molecules for the maintenance of the Warburg effect in colorectal tumors.


Assuntos
Neoplasias Colorretais/genética , Neoplasias Colorretais/metabolismo , Ribonucleoproteínas Nucleares Heterogêneas/genética , Ribonucleoproteínas Nucleares Heterogêneas/metabolismo , MicroRNAs/biossíntese , Proteína de Ligação a Regiões Ricas em Polipirimidinas/genética , Proteína de Ligação a Regiões Ricas em Polipirimidinas/metabolismo , Animais , Linhagem Celular Tumoral , Neoplasias Colorretais/patologia , Regulação para Baixo , Feminino , Xenoenxertos , Humanos , Masculino , Camundongos , Camundongos Nus , MicroRNAs/genética , MicroRNAs/metabolismo , Transfecção
14.
Int J Mol Sci ; 17(1)2016 Jan 08.
Artigo em Inglês | MEDLINE | ID: mdl-26760996

RESUMO

Organic gem-dihydroperoxides (DHPs) and their derived peroxides have attracted a great deal of attention as potential anti-cancer agents. However, the precise mechanism of their inhibitory effect on tumors is unknown. To determine the mechanism of the inhibitory effects of DHPs, we examined the effects of DHPs on leukemia K562 cells. As a result, certain DHPs used in this study exhibited growth-inhibitory activity according to a clear structure-activity relationship. The most potent DHP, 12AC3O, induced apoptosis in K562 cells, but not in peripheral blood monocytes (PBMCs) or fibroblast cells. 12AC3O induced apoptosis through the intrinsic mitochondrial pathway and thereafter through the extrinsic pathway. The activity of the former pathway was partly attenuated by a JNK inhibitor. Interestingly, 12AC3O induced apoptosis by trapping a large amount of ROS, leading to an extremely lower intracellular ROS level compared with that in the cells in the steady-state condition. These results suggest that an appropriate level of intracellular ROS was necessary for the maintenance of cancer cell growth. DHPs may have a potential to be a novel anti-cancer agent with minimum adverse effects on normal cells.


Assuntos
Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Peróxido de Hidrogênio/farmacologia , Leucemia/tratamento farmacológico , Espécies Reativas de Oxigênio/metabolismo , Antineoplásicos/química , Humanos , Peróxido de Hidrogênio/química , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Células K562 , Leucemia/metabolismo
15.
Cancer Lett ; 371(1): 1-11, 2016 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-26607903

RESUMO

In Ph-positive leukemia, imatinib brought marked clinical improvement; however, further improvement is needed to prevent relapse. Cancer cells efficiently use limited energy sources, and drugs targeting cellular metabolism improve the efficacy of therapy. In this study, we characterized the effects of novel anti-cancer fatty-acid derivative AIC-47 and imatinib, focusing on cancer-specific energy metabolism in chronic myeloid leukemia cells. AIC-47 and imatinib in combination exhibited a significant synergic cytotoxicity. Imatinib inhibited only the phosphorylation of BCR-ABL; whereas AIC-47 suppressed the expression of the protein itself. Both AIC-47 and imatinib modulated the expression of pyruvate kinase M (PKM) isoforms from PKM2 to PKM1 through the down-regulation of polypyrimidine tract-binding protein 1 (PTBP1). PTBP1 functions as alternative splicing repressor of PKM1, resulting in expression of PKM2, which is an inactive form of pyruvate kinase for the last step of glycolysis. Although inactivation of BCR-ABL by imatinib strongly suppressed glycolysis, compensatory fatty-acid oxidation (FAO) activation supported glucose-independent cell survival by up-regulating CPT1C, the rate-limiting FAO enzyme. In contrast, AIC-47 inhibited the expression of CPT1C and directly fatty-acid metabolism. These findings were also observed in the CD34(+) fraction of Ph-positive acute lymphoblastic leukemia cells. These results suggest that AIC-47 in combination with imatinib strengthened the attack on cancer energy metabolism, in terms of both glycolysis and compensatory activation of FAO.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Metabolismo Energético/efeitos dos fármacos , Ácidos Graxos/farmacologia , Compostos Heterocíclicos com 1 Anel/farmacologia , Mesilato de Imatinib/farmacologia , Cetonas/farmacologia , Leucemia Mielogênica Crônica BCR-ABL Positiva/tratamento farmacológico , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamento farmacológico , Inibidores de Proteínas Quinases/farmacologia , Antígenos CD34/metabolismo , Relação Dose-Resposta a Droga , Sinergismo Farmacológico , Proteínas de Fusão bcr-abl/genética , Proteínas de Fusão bcr-abl/metabolismo , Glicólise/efeitos dos fármacos , Ribonucleoproteínas Nucleares Heterogêneas/genética , Ribonucleoproteínas Nucleares Heterogêneas/metabolismo , Humanos , Células K562 , Leucemia Mielogênica Crônica BCR-ABL Positiva/genética , Leucemia Mielogênica Crônica BCR-ABL Positiva/metabolismo , MicroRNAs/genética , MicroRNAs/metabolismo , Células-Tronco Neoplásicas/efeitos dos fármacos , Células-Tronco Neoplásicas/metabolismo , Oxirredução , Proteína de Ligação a Regiões Ricas em Polipirimidinas/genética , Proteína de Ligação a Regiões Ricas em Polipirimidinas/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/metabolismo , Piruvato Quinase/genética , Piruvato Quinase/metabolismo , Interferência de RNA , Transfecção
16.
Am J Physiol Heart Circ Physiol ; 309(11): H1813-26, 2015 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-26432843

RESUMO

We investigated whether microRNA-145 (miR-145) has a cardioprotective effect in a rabbit model of myocardial infarction (MI) and in H9c2 rat cardiomyoblasts. Rabbits underwent 30 min of coronary occlusion, followed by 2 days or 2 wk of reperfusion. Control microRNA (control group; 2.5 nmol/kg, n = 10) or miR-145 (miR-145 group, 2.5 nmol/kg, n = 10) encapsulated in liposomes was intravenously administered immediately after the start of reperfusion. H9c2 rat cardiomyoblasts were transfected with miR-145. The MI size was significantly smaller in the miR-145 group than in the control group at 2 days and 2 wk post-MI. miR-145 had improved the cardiac function and remodeling at 2 wk post-MI. These effects were reversed by chloroquine. Western blot analysis showed that miR-145 accelerated the transition of LC3B I to II and downregulated p62/SQSTM1 at 2 days or 2 wk after MI, but not at 4 wk, and activated Akt in the ischemic area at 2 days after MI. miR-145 inhibited the growth of H9c2 cells, accelerated the transition of LC3B I to II, and increased phosphorylated Akt in the H9c2 cells at 2 days after miR-145 transfection. Antagomir-145 significantly abolished the morphological change, the transition of LC3B I to II, and the increased phosphorylated Akt induced by miR-145 in H9c2 cells. We determined fibroblast growth factor receptor substrate 2 mRNA to be a target of miR-145, both in an in vivo model and in H9c2 cells. In conclusion, post-MI treatment with miR-145 protected the heart through the induction of cardiomyocyte autophagy by targeting fibroblast growth factor receptor substrate 2.


Assuntos
Autofagia , Terapia Genética/métodos , MicroRNAs/metabolismo , Infarto do Miocárdio/terapia , Miócitos Cardíacos/metabolismo , Função Ventricular Esquerda , Remodelação Ventricular , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Linhagem Celular , Modelos Animais de Doenças , Fator 4 Semelhante a Kruppel , Fatores de Transcrição Kruppel-Like/metabolismo , Masculino , MicroRNAs/administração & dosagem , MicroRNAs/genética , Proteínas Associadas aos Microtúbulos/metabolismo , Infarto do Miocárdio/genética , Infarto do Miocárdio/metabolismo , Infarto do Miocárdio/patologia , Infarto do Miocárdio/fisiopatologia , Miócitos Cardíacos/patologia , Fosforilação , Proteínas Proto-Oncogênicas c-akt/metabolismo , Interferência de RNA , RNA Mensageiro/metabolismo , Coelhos , Ratos , Transdução de Sinais , Fatores de Tempo , Transfecção
17.
Oncotarget ; 6(28): 25828-42, 2015 Sep 22.
Artigo em Inglês | MEDLINE | ID: mdl-26304927

RESUMO

Tumor necrosis-factor (TNF)-related apoptosis-inducing ligand (TRAIL) is a member of the TNF-superfamily that selectively induces apoptosis through death receptors (DRs) 4 and/or 5 in cancer cells. These receptors are expressed on the cancer cell surface, without affecting normal cells. Unfortunately, many clinical studies have shown that cancer cells acquire TRAIL-resistance and finally avoid TRAIL-induced apoptosis. The detailed mechanisms of this resistance are not well understood. In the current study, we established a TRAIL-resistant human colon cancer DLD-1 cell line to clarify the mechanisms of TRAIL-resistance and developed agents to cancel its machinery. Also, we found that cancer stem-like cells from breast epithelial proliferating MCF10A cells were also sensitive to TRAIL-induced apoptosis. The enforced expression of DR5 in both TRAIL-resistant cells partially recovered the sensitivity to the TRAIL ligand, which was judged by the activation of caspase-8. As a result, we newly found that the mechanisms of TRAIL-resistance comprised co-existence of a decrease in the expression level of DR5 along with malfunction of its recruitment to the cell surface, as evidenced by Western blot and immunocytological analysis, respectively. Interestingly, α-mangostin, which is a xanthone derivative, canceled the resistance by increasing the expression level of DR5 through down-regulation of miR-133b and effectively induced the translocation of DR5 to the cancer cell surface membrane in TRAIL-resistant DLD-1 cells. These findings indicate that α-mangostin functioned as a sensitizer of TRAIL-induced apoptosis and may thus serve as a possible adjuvant compound for cytokine therapy to conquer TRAIL-resistance.


Assuntos
Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Neoplasias da Mama/tratamento farmacológico , Neoplasias do Colo/tratamento farmacológico , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Células-Tronco Neoplásicas/efeitos dos fármacos , Ligante Indutor de Apoptose Relacionado a TNF/farmacologia , Xantonas/farmacologia , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Caspase 8/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Neoplasias do Colo/genética , Neoplasias do Colo/metabolismo , Neoplasias do Colo/patologia , Relação Dose-Resposta a Droga , Ativação Enzimática , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , MicroRNAs/genética , MicroRNAs/metabolismo , Células-Tronco Neoplásicas/metabolismo , Células-Tronco Neoplásicas/patologia , Transporte Proteico , Interferência de RNA , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/agonistas , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/genética , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transfecção
18.
Biochim Biophys Acta ; 1852(9): 1971-80, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26144048

RESUMO

The human DEAD/H-box RNA helicase gene DDX6 is a target of the t(11;14)(q23;q32) chromosomal translocation observed in human B-cell lymphoma, and the overexpression of its protein has been shown to cause malignant transformation. DDX6 has a variety of functions such as translation initiation, pre-mRNA splicing, ribosome assembly, and more. However, details of the regulatory mechanism of DDX6 and functions of DDX6 in cancer cells are largely unknown. On the other hand, the Warburg effect is a well-known feature of cancer cells. Pyruvate kinase in muscle (PKM), which is a rate-limiting glycolytic enzyme, has 2 isoforms, PKM1 and PKM2. It has been frequently reported that PKM2 is a tumor-specific isoform and promotes the Warburg effect. However, the functions of the PKM1 gene have been hardly mentioned. Here, we showed that DDX6 was overexpressed in colorectal cancer specimens and regulated by microRNA (miR)-124 in colon cancer cells. Also, a DDX6/c-Myc/PTB1 positive feedback circuit regulated by miR-124 was shown to be established and to contribute to maintenance of the Warburg effect. Moreover, we showed that knockdown of DDX6 induced mainly apoptosis through an imbalance of PKM gene expression, especially causing down-regulation of PKM1 in colon cancer cells. These results suggest that miR-124 is a fine tuner of the Warburg effect and that DDX6 is one of the key molecules in Warburg effect-related miR-124 targeting various genes.

19.
Cancer Lett ; 363(1): 17-27, 2015 Jul 10.
Artigo em Inglês | MEDLINE | ID: mdl-25818238

RESUMO

Altered levels and functions of microRNAs (miRs) have been associated with carcinogenesis. In this study, we investigated the role of miR-124 in colorectal adenoma (CRA) and cancer (CRC). The expression levels of miR-124 were decreased in CRA (81.8%) and CRC (57.6%) in 55 clinical samples. The ectopic expression of miR-124 induced apoptosis and autophagy in colon cancer cells. Also, miR-124 targeted polypyrimidine tract-binding protein 1 (PTB1), which is a splicer of pyruvate kinase muscles 1 and 2 (PKM1 and PKM2) and induced the switching of PKM isoform expression from PKM2 to PKM1. Also, siR-PTB1 induced drastic apoptosis in colon cancer cells. Furthermore, we found that the ectopic expression of miR-124 enhanced oxidative stress and the miR-124/PTB1/PKM1/PKM2 axis constituted a feedback cascade. Finally, we showed that intratumor injection of miR-124 and siR-PTB1 induced a tumor-suppressive effect in xenografted mice. The axis was established by both in vitro and in vivo experiments to function in human colorectal cancer cells. These findings suggest that miR-124 acts as a tumor-suppressor and a modulator of energy metabolism through a PTB1/PKM1/PKM2 feedback cascade in human colorectal tumor cells.


Assuntos
Proteínas de Transporte/metabolismo , Neoplasias Colorretais/enzimologia , Ribonucleoproteínas Nucleares Heterogêneas/metabolismo , Proteínas de Membrana/metabolismo , MicroRNAs/metabolismo , Proteína de Ligação a Regiões Ricas em Polipirimidinas/metabolismo , Hormônios Tireóideos/metabolismo , Regiões 3' não Traduzidas , Animais , Apoptose , Autofagia , Proteínas de Transporte/genética , Estudos de Casos e Controles , Linhagem Celular Tumoral , Neoplasias Colorretais/genética , Neoplasias Colorretais/patologia , Neoplasias Colorretais/terapia , Metabolismo Energético , Retroalimentação Fisiológica , Feminino , Regulação Enzimológica da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Terapia Genética/métodos , Ribonucleoproteínas Nucleares Heterogêneas/genética , Humanos , Masculino , Proteínas de Membrana/genética , Camundongos Endogâmicos BALB C , Camundongos Nus , MicroRNAs/administração & dosagem , MicroRNAs/genética , Estresse Oxidativo , Proteína de Ligação a Regiões Ricas em Polipirimidinas/genética , Interferência de RNA , Hormônios Tireóideos/genética , Fatores de Tempo , Transfecção , Carga Tumoral , Ensaios Antitumorais Modelo de Xenoenxerto , Proteínas de Ligação a Hormônio da Tireoide
20.
Cancer Lett ; 360(1): 28-38, 2015 Apr 28.
Artigo em Inglês | MEDLINE | ID: mdl-25644089

RESUMO

The fusion gene bcr-abl develops chronic myeloid leukemia (CML), and stimulates PI3K/Akt/mTOR signaling, leading to impaired autophagy. PI3K/Akt/mTOR signaling also plays an important role in cell metabolism. The Warburg effect is a well-recognized hallmark of cancer energy metabolism, and is regulated by the mTOR/c-Myc/hnRNP/PKM signaling cascade. To develop a new strategy for the treatment of CML, we investigated the associations among bcr-abl, the cascade related to cancer energy metabolism, and autophagy induced by a fatty-acid derivative that we had previously reported as being an autophagy inducer. Here we report that a fatty-acid derivative, AIC-47, induced transcriptional repression of the bcr-abl gene and modulated the expression profile of PKM isoforms, resulting in autophagic cell death. We show that c-Myc functioned as a transcriptional activator of bcr-abl, and regulated the hnRNP/PKM cascade. AIC-47, acting through the PPARγ/ß-catenin pathway, induced down-regulation of c-Myc, leading to the disruption of the bcr-abl/mTOR/hnRNP signaling pathway, and switching of the expression of PKM2 to PKM1. This switching caused autophagic cell death through an increase in the ROS level. Our findings suggest that AIC-47 induced autophagic cell death through the PPARγ/ß-catenin/bcr-abl/mTOR/hnRNP/PKM cascade.


Assuntos
Antineoplásicos/farmacologia , Autofagia/efeitos dos fármacos , Proteínas de Transporte/metabolismo , Ácidos Graxos/farmacologia , Proteínas de Fusão bcr-abl/metabolismo , Leucemia Mielogênica Crônica BCR-ABL Positiva/enzimologia , Leucemia Mielogênica Crônica BCR-ABL Positiva/patologia , Proteínas de Membrana/metabolismo , Piruvato Quinase/metabolismo , Hormônios Tireóideos/metabolismo , Proteínas de Transporte/genética , Relação Dose-Resposta a Droga , Metabolismo Energético/efeitos dos fármacos , Proteínas de Fusão bcr-abl/genética , Regulação Neoplásica da Expressão Gênica , Ribonucleoproteínas Nucleares Heterogêneas/metabolismo , Humanos , Isoenzimas , Células K562 , Leucemia Mielogênica Crônica BCR-ABL Positiva/genética , Proteínas de Membrana/genética , PPAR gama/metabolismo , Proteínas Proto-Oncogênicas c-myc/genética , Proteínas Proto-Oncogênicas c-myc/metabolismo , Piruvato Quinase/genética , Interferência de RNA , Transdução de Sinais/efeitos dos fármacos , Serina-Treonina Quinases TOR/metabolismo , Hormônios Tireóideos/genética , Transcrição Gênica , Transfecção , beta Catenina/genética , beta Catenina/metabolismo , Proteínas de Ligação a Hormônio da Tireoide
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...