Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
JCI Insight ; 6(7)2021 04 08.
Article in English | MEDLINE | ID: mdl-33830081

ABSTRACT

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.


Subject(s)
Endoplasmic Reticulum Stress/physiology , Lung Neoplasms/pathology , Proto-Oncogene Proteins p21(ras)/genetics , Verteporfin/pharmacology , Animals , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Apoptosis/physiology , Endoplasmic Reticulum Stress/drug effects , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Mice, Inbred BALB C , Mutation , Proto-Oncogene Proteins p21(ras)/metabolism , Xenograft Model Antitumor Assays , YAP-Signaling Proteins/genetics
2.
Biochem Biophys Res Commun ; 525(2): 483-490, 2020 04 30.
Article in English | MEDLINE | ID: mdl-32111352

ABSTRACT

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.


Subject(s)
Carcinogenesis/genetics , Lung Neoplasms/genetics , Proto-Oncogene Proteins p21(ras)/genetics , Adenocarcinoma of Lung/genetics , Carcinoma, Squamous Cell/genetics , Cell Line , Cell Transformation, Neoplastic/genetics , Genes, ras , Humans , Mutation , Respiratory Mucosa/cytology , Respiratory Mucosa/metabolism
3.
Cancer Sci ; 111(2): 395-405, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31854049

ABSTRACT

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.


Subject(s)
Breast Neoplasms/genetics , MicroRNAs/genetics , Nuclear Proteins/genetics , ran GTP-Binding Protein/genetics , 3' Untranslated Regions , Breast Neoplasms/metabolism , Cell Line, Tumor , Cell Nucleus/metabolism , Cell Proliferation , Centrosome/metabolism , Down-Regulation , Female , Gene Expression Regulation, Neoplastic , Humans , MCF-7 Cells , Nuclear Proteins/metabolism , ran GTP-Binding Protein/metabolism
4.
Oncogene ; 38(28): 5566-5579, 2019 07.
Article in English | MEDLINE | ID: mdl-31147602

ABSTRACT

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.


Subject(s)
Cancer-Associated Fibroblasts/metabolism , Chemokine CXCL1/biosynthesis , Extracellular Vesicles/metabolism , Interleukin-8/biosynthesis , Stomach Neoplasms/pathology , Stromal Cells/pathology , Actins/metabolism , Animals , Cell Line, Tumor , Heterografts , Humans , Mice , Stomach Neoplasms/metabolism , Tumor Microenvironment
5.
Cancer Lett ; 451: 11-22, 2019 06 01.
Article in English | MEDLINE | ID: mdl-30862488

ABSTRACT

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.


Subject(s)
Antineoplastic Agents/pharmacology , Benzimidazoles/pharmacology , Carcinoma, Non-Small-Cell Lung/drug therapy , Genes, ras , Lung Neoplasms/drug therapy , Mutation , Antineoplastic Agents/chemistry , Benzimidazoles/chemistry , Carcinoma, Non-Small-Cell Lung/genetics , Cell Line, Tumor , Humans , Lung Neoplasms/genetics , Signal Transduction/drug effects
6.
Cancer Sci ; 109(5): 1455-1467, 2018 May.
Article in English | MEDLINE | ID: mdl-29498789

ABSTRACT

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.


Subject(s)
Colonic Neoplasms/drug therapy , MicroRNAs/administration & dosage , MicroRNAs/chemical synthesis , Protein Kinase Inhibitors/administration & dosage , Proto-Oncogene Proteins p21(ras)/genetics , Animals , Benzimidazoles/administration & dosage , Benzimidazoles/pharmacology , Benzothiazoles/administration & dosage , Benzothiazoles/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Colonic Neoplasms/genetics , Drug Synergism , Flavonoids/administration & dosage , Flavonoids/pharmacology , HT29 Cells , Humans , Mice , MicroRNAs/antagonists & inhibitors , MicroRNAs/chemistry , MicroRNAs/pharmacology , Mutation , Neoplasm Transplantation , Protein Kinase Inhibitors/pharmacology , Signal Transduction/drug effects
7.
J Immunol ; 199(4): 1505-1515, 2017 08 15.
Article in English | MEDLINE | ID: mdl-28696255

ABSTRACT

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.


Subject(s)
Colorectal Neoplasms/immunology , Extracellular Vesicles/physiology , Macrophages/immunology , MicroRNAs/metabolism , Tumor Microenvironment/immunology , Animals , Cell Line, Tumor , Colorectal Neoplasms/genetics , Down-Regulation , Extracellular Vesicles/genetics , Histone Deacetylases/genetics , Histone Deacetylases/metabolism , Humans , Macrophage Activation , Mice , Mice, Nude , MicroRNAs/genetics , Phenotype , Tumor Microenvironment/genetics
8.
Int J Mol Sci ; 18(1)2017 Jan 17.
Article in English | MEDLINE | ID: mdl-28106737

ABSTRACT

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.


Subject(s)
Apoptosis/genetics , Glycolysis/genetics , Heterogeneous-Nuclear Ribonucleoproteins/genetics , MicroRNAs/genetics , Polypyrimidine Tract-Binding Protein/genetics , RNA Interference , Urinary Bladder Neoplasms/genetics , Aged , Aged, 80 and over , Animals , Blotting, Western , Cell Line, Tumor , Cell Proliferation/genetics , Female , Gene Expression Regulation, Neoplastic , Heterogeneous-Nuclear Ribonucleoproteins/metabolism , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Middle Aged , Models, Genetic , Polypyrimidine Tract-Binding Protein/metabolism , RNAi Therapeutics/methods , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/genetics , Urinary Bladder Neoplasms/metabolism , Urinary Bladder Neoplasms/therapy , Xenograft Model Antitumor Assays/methods
9.
Int J Mol Sci ; 17(11)2016 Nov 05.
Article in English | MEDLINE | ID: mdl-27827955

ABSTRACT

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.


Subject(s)
Epithelial Cells/metabolism , G1 Phase Cell Cycle Checkpoints/genetics , Gene Expression Regulation, Neoplastic , Intercellular Signaling Peptides and Proteins/genetics , Pinocytosis/genetics , Adaptor Proteins, Signal Transducing , Adenine/analogs & derivatives , Adenine/pharmacology , Amiloride/analogs & derivatives , Amiloride/pharmacology , Autophagy/drug effects , Autophagy/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Chemokines , Epithelial Cells/drug effects , Epithelial Cells/pathology , G1 Phase Cell Cycle Checkpoints/drug effects , Gene Silencing/drug effects , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Lysosomal Membrane Proteins/genetics , Lysosomal Membrane Proteins/metabolism , MAP Kinase Kinase 4/genetics , MAP Kinase Kinase 4/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/metabolism , Pinocytosis/drug effects , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction , Urinary Bladder/metabolism , Urinary Bladder/pathology , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/metabolism , rab5 GTP-Binding Proteins/genetics , rab5 GTP-Binding Proteins/metabolism , rac1 GTP-Binding Protein/genetics , rac1 GTP-Binding Protein/metabolism
10.
Exp Cell Res ; 347(1): 133-142, 2016 09 10.
Article in English | MEDLINE | ID: mdl-27453209

ABSTRACT

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.


Subject(s)
Glycolysis/drug effects , Neoplasms/pathology , TNF-Related Apoptosis-Inducing Ligand/pharmacology , Acetylcysteine/pharmacology , Cell Death/drug effects , Cell Line, Tumor , Cell Membrane/drug effects , Cell Membrane/metabolism , Cell Proliferation/drug effects , Cysteine-Rich Protein 61/metabolism , Gene Knockdown Techniques , Gene Silencing/drug effects , Heterogeneous-Nuclear Ribonucleoproteins/metabolism , Humans , Polypyrimidine Tract-Binding Protein/metabolism , Protein Multimerization/drug effects , RNA, Small Interfering/metabolism , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , Up-Regulation/drug effects
11.
Oncotarget ; 7(19): 27033-43, 2016 May 10.
Article in English | MEDLINE | ID: mdl-27081032

ABSTRACT

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.


Subject(s)
Colorectal Neoplasms/immunology , Extracellular Vesicles/immunology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes/immunology , Transforming Growth Factor beta1/immunology , Animals , Blotting, Western , Cell Line, Tumor , Cell Proliferation/genetics , Cells, Cultured , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Extracellular Vesicles/genetics , Extracellular Vesicles/metabolism , Female , Humans , Jurkat Cells , Mice, Nude , RNA Interference , Signal Transduction/genetics , Signal Transduction/immunology , T-Lymphocytes/metabolism , T-Lymphocytes, Regulatory/metabolism , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/metabolism , Transplantation, Heterologous
12.
Biochem Biophys Res Commun ; 473(1): 174-180, 2016 Apr 22.
Article in English | MEDLINE | ID: mdl-27012213

ABSTRACT

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.


Subject(s)
Antineoplastic Agents/chemistry , Carrier Proteins/metabolism , Drug Resistance, Neoplasm , Membrane Proteins/metabolism , Neoplasms/drug therapy , Neoplasms/metabolism , Thyroid Hormones/metabolism , Apoptosis , Cell Line, Tumor , Fluorouracil/chemistry , Gene Expression Regulation, Neoplastic , Glycolysis/drug effects , Humans , Inhibitory Concentration 50 , K562 Cells , Organoplatinum Compounds/chemistry , Oxaliplatin , Oxidative Phosphorylation , Phenotype , Resting Phase, Cell Cycle , Transfection , Thyroid Hormone-Binding Proteins
13.
Oncotarget ; 7(14): 18940-52, 2016 Apr 05.
Article in English | MEDLINE | ID: mdl-26980745

ABSTRACT

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.


Subject(s)
Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Heterogeneous-Nuclear Ribonucleoproteins/genetics , Heterogeneous-Nuclear Ribonucleoproteins/metabolism , MicroRNAs/biosynthesis , Polypyrimidine Tract-Binding Protein/genetics , Polypyrimidine Tract-Binding Protein/metabolism , Animals , Cell Line, Tumor , Colorectal Neoplasms/pathology , Down-Regulation , Female , Heterografts , Humans , Male , Mice , Mice, Nude , MicroRNAs/genetics , MicroRNAs/metabolism , Transfection
14.
Int J Mol Sci ; 17(1)2016 Jan 08.
Article in English | MEDLINE | ID: mdl-26760996

ABSTRACT

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.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Hydrogen Peroxide/pharmacology , Leukemia/drug therapy , Reactive Oxygen Species/metabolism , Antineoplastic Agents/chemistry , Humans , Hydrogen Peroxide/chemistry , JNK Mitogen-Activated Protein Kinases/metabolism , K562 Cells , Leukemia/metabolism
15.
Cancer Lett ; 371(1): 1-11, 2016 Feb 01.
Article in English | MEDLINE | ID: mdl-26607903

ABSTRACT

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.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Energy Metabolism/drug effects , Fatty Acids/pharmacology , Heterocyclic Compounds, 1-Ring/pharmacology , Imatinib Mesylate/pharmacology , Ketones/pharmacology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Protein Kinase Inhibitors/pharmacology , Antigens, CD34/metabolism , Dose-Response Relationship, Drug , Drug Synergism , Fusion Proteins, bcr-abl/genetics , Fusion Proteins, bcr-abl/metabolism , Glycolysis/drug effects , Heterogeneous-Nuclear Ribonucleoproteins/genetics , Heterogeneous-Nuclear Ribonucleoproteins/metabolism , Humans , K562 Cells , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Oxidation-Reduction , Polypyrimidine Tract-Binding Protein/genetics , Polypyrimidine Tract-Binding Protein/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Pyruvate Kinase/genetics , Pyruvate Kinase/metabolism , RNA Interference , Transfection
16.
Am J Physiol Heart Circ Physiol ; 309(11): H1813-26, 2015 Dec 01.
Article in English | MEDLINE | ID: mdl-26432843

ABSTRACT

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.


Subject(s)
Autophagy , Genetic Therapy/methods , MicroRNAs/metabolism , Myocardial Infarction/therapy , Myocytes, Cardiac/metabolism , Ventricular Function, Left , Ventricular Remodeling , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cell Line , Disease Models, Animal , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/metabolism , Male , MicroRNAs/administration & dosage , MicroRNAs/genetics , Microtubule-Associated Proteins/metabolism , Myocardial Infarction/genetics , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myocytes, Cardiac/pathology , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , RNA Interference , RNA, Messenger/metabolism , Rabbits , Rats , Signal Transduction , Time Factors , Transfection
17.
Oncotarget ; 6(28): 25828-42, 2015 Sep 22.
Article in English | MEDLINE | ID: mdl-26304927

ABSTRACT

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.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Breast Neoplasms/drug therapy , Colonic Neoplasms/drug therapy , Drug Resistance, Neoplasm/drug effects , Neoplastic Stem Cells/drug effects , TNF-Related Apoptosis-Inducing Ligand/pharmacology , Xanthones/pharmacology , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Caspase 8/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Colonic Neoplasms/genetics , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Dose-Response Relationship, Drug , Enzyme Activation , Female , Gene Expression Regulation, Neoplastic , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Protein Transport , RNA Interference , Receptors, TNF-Related Apoptosis-Inducing Ligand/agonists , Receptors, TNF-Related Apoptosis-Inducing Ligand/genetics , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , Signal Transduction/drug effects , Transfection
18.
Biochim Biophys Acta ; 1852(9): 1971-80, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26144048

ABSTRACT

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.
Article in English | MEDLINE | ID: mdl-25818238

ABSTRACT

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.


Subject(s)
Carrier Proteins/metabolism , Colorectal Neoplasms/enzymology , Heterogeneous-Nuclear Ribonucleoproteins/metabolism , Membrane Proteins/metabolism , MicroRNAs/metabolism , Polypyrimidine Tract-Binding Protein/metabolism , Thyroid Hormones/metabolism , 3' Untranslated Regions , Animals , Apoptosis , Autophagy , Carrier Proteins/genetics , Case-Control Studies , Cell Line, Tumor , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Colorectal Neoplasms/therapy , Energy Metabolism , Feedback, Physiological , Female , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Genetic Therapy/methods , Heterogeneous-Nuclear Ribonucleoproteins/genetics , Humans , Male , Membrane Proteins/genetics , Mice, Inbred BALB C , Mice, Nude , MicroRNAs/administration & dosage , MicroRNAs/genetics , Oxidative Stress , Polypyrimidine Tract-Binding Protein/genetics , RNA Interference , Thyroid Hormones/genetics , Time Factors , Transfection , Tumor Burden , Xenograft Model Antitumor Assays , Thyroid Hormone-Binding Proteins
20.
Cancer Lett ; 360(1): 28-38, 2015 Apr 28.
Article in English | MEDLINE | ID: mdl-25644089

ABSTRACT

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.


Subject(s)
Antineoplastic Agents/pharmacology , Autophagy/drug effects , Carrier Proteins/metabolism , Fatty Acids/pharmacology , Fusion Proteins, bcr-abl/metabolism , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/enzymology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Membrane Proteins/metabolism , Pyruvate Kinase/metabolism , Thyroid Hormones/metabolism , Carrier Proteins/genetics , Dose-Response Relationship, Drug , Energy Metabolism/drug effects , Fusion Proteins, bcr-abl/genetics , Gene Expression Regulation, Neoplastic , Heterogeneous-Nuclear Ribonucleoproteins/metabolism , Humans , Isoenzymes , K562 Cells , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Membrane Proteins/genetics , PPAR gamma/metabolism , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , Pyruvate Kinase/genetics , RNA Interference , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/metabolism , Thyroid Hormones/genetics , Transcription, Genetic , Transfection , beta Catenin/genetics , beta Catenin/metabolism , Thyroid Hormone-Binding Proteins
SELECTION OF CITATIONS
SEARCH DETAIL
...