Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Cancer Res ; 81(21): 5451-5463, 2021 11 01.
Article in English | MEDLINE | ID: mdl-34462275

ABSTRACT

Ionizing radiation (IR) and chemotherapy are mainstays of treatment for patients with rhabdomyosarcoma, yet the molecular mechanisms that underlie the success or failure of radiotherapy remain unclear. The transcriptional repressor SNAI2 was previously identified as a key regulator of IR sensitivity in normal and malignant stem cells through its repression of the proapoptotic BH3-only gene PUMA/BBC3. Here, we demonstrate a clear correlation between SNAI2 expression levels and radiosensitivity across multiple rhabdomyosarcoma cell lines. Modulating SNAI2 levels in rhabdomyosarcoma cells through its overexpression or knockdown altered radiosensitivity in vitro and in vivo. SNAI2 expression reliably promoted overall cell growth and inhibited mitochondrial apoptosis following exposure to IR, with either variable or minimal effects on differentiation and senescence, respectively. Importantly, SNAI2 knockdown increased expression of the proapoptotic BH3-only gene BIM, and chromatin immunoprecipitation sequencing experiments established that SNAI2 is a direct repressor of BIM/BCL2L11. Because the p53 pathway is nonfunctional in the rhabdomyosarcoma cells used in this study, we have identified a new, p53-independent SNAI2/BIM signaling axis that could potentially predict clinical responses to IR treatment and be exploited to improve rhabdomyosarcoma therapy. SIGNIFICANCE: SNAI2 is identified as a major regulator of radiation-induced apoptosis in rhabdomyosarcoma through previously unknown mechanisms independent of p53.


Subject(s)
Bcl-2-Like Protein 11/antagonists & inhibitors , Biomarkers, Tumor/metabolism , Gene Expression Regulation, Neoplastic/radiation effects , Radiation, Ionizing , Rhabdomyosarcoma/prevention & control , Snail Family Transcription Factors/metabolism , Animals , Apoptosis , Bcl-2-Like Protein 11/genetics , Bcl-2-Like Protein 11/metabolism , Biomarkers, Tumor/genetics , Cell Cycle , Cell Movement , Cell Proliferation , Female , Humans , Mice , Mice, SCID , RNA-Seq , Rhabdomyosarcoma/etiology , Rhabdomyosarcoma/pathology , Snail Family Transcription Factors/genetics , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
2.
Leukemia ; 35(10): 2862-2874, 2021 10.
Article in English | MEDLINE | ID: mdl-34007045

ABSTRACT

Herein, we screened a novel inhibitor of the Hsp70-Bim protein-protein interaction (PPI), S1g-2, from a Bcl-2 inhibitor library; this compound specifically disrupted the Hsp70-Bim PPI by direct binding to an unknown site adjacent to that of an allosteric Hsp70 inhibitor MKT-077, showing binding affinity in sub-µM concentration range. S1g-2 exhibited overall 5-10-fold higher apoptosis-inducing activity in CML cells, primary CML blasts, and BCR-ABL-transformed BaF3 cells than other cancer cells, normal lymphocytes, and BaF3 cells, illustrating Hsp70-Bim PPI driven by BCR-ABL protects CML through oncoclient proteins that enriched in three pathways: eIF2 signaling, the regulation of eIF4E and p70S6K signaling, and the mTOR signaling pathways. Moreover, S1g-2 progressively enhanced lethality along with the increase in BCR-ABL-independent TKI resistance in the K562 cell lines and is more effective in primary samples from BCR-ABL-independent TKI-resistant patients than those from TKI-sensitive patients. By comparing the underlying mechanisms of S1g-2, MKT-077, and an ATP-competitive Hsp70 inhibitor VER-155008, the Hsp70-Bim PPI was identified to be a CML-specific target to protect from TKIs through the above three oncogenic signaling pathways. The in vivo activity against CML and low toxicity endows S1g-2 a first-in-class promising drug candidate for both TKI-sensitive and resistant CML.


Subject(s)
Bcl-2-Like Protein 11/antagonists & inhibitors , Drug Resistance, Neoplasm , Fusion Proteins, bcr-abl/antagonists & inhibitors , Gene Expression Regulation, Leukemic/drug effects , HSP70 Heat-Shock Proteins/antagonists & inhibitors , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Protein Interaction Domains and Motifs , Small Molecule Libraries/pharmacology , Animals , Antineoplastic Agents/pharmacology , Apoptosis , Bcl-2-Like Protein 11/genetics , Bcl-2-Like Protein 11/metabolism , Cell Proliferation , Fusion Proteins, bcr-abl/genetics , HSP70 Heat-Shock Proteins/genetics , HSP70 Heat-Shock Proteins/metabolism , Humans , Imatinib Mesylate/pharmacology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Male , Mice , Mice, Nude , Prognosis , Protein Kinase Inhibitors/pharmacology , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
3.
Eur J Med Chem ; 220: 113452, 2021 Aug 05.
Article in English | MEDLINE | ID: mdl-33906046

ABSTRACT

Targeting cancer-related Hsp70-Bim protein-protein interactions (PPIs) offers a new strategy for the design of Hsp70 inhibitors. Herein, we discovered a novel Hsp70 inhibitor, S1g-6, based on the established BH3 mimetics. S1g-6 exhibited sub-µM binding affinity toward Hsp70 and selectively disrupted Hsp70-Bim PPI. The target specificity of S1g-6in situ was validated by affinity-based protein profiling, co-immunoprecipitation, and cell-based shRNA assays. S1g-6 specifically antagonized the ATPase activity of Hsp70 upon recruiting Bim and showed selective apoptosis induction in some cancer cell lines over normal ones through suppression of some oncogenic clients of Hsp70, representing a new class of antitumor candidates. Hsp70-Bim PPI exhibited cancer-dependent role as a potential anti-cancer target.


Subject(s)
Antineoplastic Agents/pharmacology , Bcl-2-Like Protein 11/antagonists & inhibitors , HSP70 Heat-Shock Proteins/antagonists & inhibitors , Antineoplastic Agents/chemistry , Bcl-2-Like Protein 11/chemistry , Dose-Response Relationship, Drug , HSP70 Heat-Shock Proteins/chemistry , Humans , Molecular Structure , Protein Binding , Structure-Activity Relationship
4.
Acta Pharmacol Sin ; 41(4): 561-571, 2020 Apr.
Article in English | MEDLINE | ID: mdl-31685975

ABSTRACT

Proximal renal tubular damage is a critical process underlying diabetic kidney disease (DKD). Our previous study shows that prostaglandin E1 (PGE1) reduces the apoptosis of renal tubular cells in DKD rats. But its underlying mechanisms remain unclear. In this study we investigated the protective effects of PGE1 in DKD rats and high glucose (HG, 30 mM)-treated HK-2 proximal tubular cells. Four weeks after uninephrectomized streptozotocin-induced diabetic rats were established, the DKD rats were administered PGE1 (10 µg· kg-1· d-1, iv.) for 10 consecutive days. We showed that PGE1 administration did not change blood glucose levels, but alleviated diabetic kidney injury in the DKD rats, evidenced by markedly reduced proteinuria and renal tubular apoptosis. In the in vitro experiments, PGE1 (0.1-100 µM) significantly enhanced HG-reduced HK-2 cell viability. In HG-treated HK-2 cells, PGE1 (10 µM) significantly suppressed the c-Jun N-terminal kinase (JNK) and the mitochondrial apoptosis-related protein expressions such as Bim, Bax, caspase-3 and cleaved caspase-3; similar changes were also observed in the kidney of PGE1-treated DKD rats. By using two pharmacological tools-JNK activator anisomycin (AM) and JNK inhibitor SP600125, we revealed that PGE1 blocked HG-triggered activation of JNK/Bim pathway in HK-2 cells; JNK was an upstream regulator of Bim. In conclusion, our results demonstrate that the nephroprotective effects of PGE1 against apoptosis of proximal renal tubule in DKD rats via suppressing JNK-related Bim signaling pathway.


Subject(s)
Alprostadil/pharmacology , Apoptosis/drug effects , Diabetic Nephropathies/drug therapy , Glucose/antagonists & inhibitors , Kidney Tubules, Proximal/drug effects , Protective Agents/pharmacology , Alprostadil/administration & dosage , Animals , Bcl-2-Like Protein 11/antagonists & inhibitors , Bcl-2-Like Protein 11/metabolism , Cells, Cultured , Diabetic Nephropathies/chemically induced , Diabetic Nephropathies/pathology , Glucose/pharmacology , Humans , Injections, Intravenous , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , JNK Mitogen-Activated Protein Kinases/metabolism , Kidney Tubules, Proximal/metabolism , Kidney Tubules, Proximal/pathology , Male , Protective Agents/administration & dosage , Rats , Rats, Wistar , Streptozocin
5.
Br J Pharmacol ; 176(14): 2465-2481, 2019 07.
Article in English | MEDLINE | ID: mdl-30932177

ABSTRACT

BACKGROUND AND PURPOSE: Expression of the pro-fibrotic galectin-3 and the pro-apoptotic BIM is elevated in diseased heart or after ß-adrenoceptor stimulation, but the underlying mechanisms are unclear. This question was addressed in the present study. EXPERIMENTAL APPROACH: Wild-type mice and mice with cardiac transgenic expression of ß2 -adrenoceptors, mammalian sterile-20 like kinase 1 (Mst1) or dominant-negative Mst1, and non-specific galectin-3 knockout mice were used. Effects of the ß-adrenoceptor agonist isoprenaline or ß-adrenoceptor antagonists were studied. Rat cardiomyoblasts (H9c2) were used for mechanistic exploration. Biochemical assays were performed. KEY RESULTS: Isoprenaline treatment up-regulated expression of galectin-3 and BIM, and this was inhibited by non-selective or selective ß-adrenoceptor antagonists (by 60-70%). Cardiac expression of galectin-3 and BIM was increased in ß2 -adrenoceptor transgenic mice. Isoprenaline-induced up-regulation of galectin-3 and BIM was attenuated by Mst1 inactivation, but isoprenaline-induced galectin-3 expression was exaggerated by transgenic Mst1 activation. Pharmacological or genetic activation of ß-adrenoceptors induced Mst1 expression and yes-associated protein (YAP) phosphorylation. YAP hyper-phosphorylation was also evident in Mst1 transgenic hearts with up-regulated expression of galectin-3 (40-fold) and BIM as well as up-regulation of many YAP-target genes by RNA sequencing. In H9c2 cells, isoprenaline induced YAP phosphorylation and expression of galectin-3 and BIM, effects simulated by forskolin but abolished by PKA inhibitors, and YAP knockdown induced expression of galectin-3 and BIM. CONCLUSIONS AND IMPLICATIONS: Stimulation of cardiac ß-adrenoceptors activated the Mst1/Hippo pathway leading to YAP hyper-phosphorylation with enhanced expression of galectin-3 and BIM. This signalling pathway would have therapeutic potential. LINKED ARTICLES: This article is part of a themed section on Adrenoceptors-New Roles for Old Players. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v176.14/issuetoc.


Subject(s)
Bcl-2-Like Protein 11/metabolism , Cardiovascular Diseases/metabolism , Galectin 3/metabolism , Receptors, Adrenergic, beta-3/metabolism , Signal Transduction , Up-Regulation , Adrenergic beta-Agonists/pharmacology , Adrenergic beta-Antagonists/pharmacology , Animals , Bcl-2-Like Protein 11/antagonists & inhibitors , Cardiovascular Diseases/drug therapy , Carvedilol/pharmacology , Cell Line , Dose-Response Relationship, Drug , Galectin 3/antagonists & inhibitors , Galectin 3/deficiency , Isoproterenol/administration & dosage , Isoproterenol/pharmacology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Propanolamines/pharmacology , Propranolol/pharmacology , Rats , Receptors, Adrenergic, beta-3/genetics , Signal Transduction/drug effects , Up-Regulation/drug effects
6.
Mol Med Rep ; 19(5): 4457-4467, 2019 May.
Article in English | MEDLINE | ID: mdl-30942414

ABSTRACT

Apoptosis of vascular smooth muscle cells (VSMCs) is a process that regulates vessel remodeling in various cardiovascular diseases. The specific mechanisms that control VSMC apoptosis remain unclear. The present study aimed to investigate whether microRNA­494 (miR­494) is involved in regulating VSMC apoptosis and its underlying mechanisms. Cell death ELISA and terminal deoxynucleotidyl­transferase­mediated dUTP nick end labeling assays were used to detect apoptosis of murine VSMCs following stimulation with tumor necrosis factor­α (TNF­α). The results indicated that TNF­α upregulated VSMC apoptosis in a dose­dependent manner. Microarray analysis was used to evaluate the expression profile of microRNAs following TNF­α stimulation in murine VSMCs. The expression of miR­494 was downregulated, whereas B­cell lymphoma-2­like 11 (BCL2L11) protein expression levels were upregulated in VSMCs following treatment with TNF­α. Luciferase reporter assays confirmed that BCL2L11 was a direct target of miR­494. Transfection with miR­494 mimics decreased VSMC apoptosis and downregulated BCL2L11 protein levels. Conversely, transfection with miR­494 inhibitors increased cell apoptosis and upregulated BCL2L11 protein levels, suggesting that miR­494 may function as an essential regulator of BCL2L11. The increase in apoptosis caused by miR­494 inhibitors was abolished in cells co­transfected with BCL2L11­targeting small interfering RNA. The findings of the present study revealed that miR­494 inhibited TNF­α­induced VSMC apoptosis by downregulating the expression of BCL2L11.


Subject(s)
Apoptosis , MicroRNAs/metabolism , 3' Untranslated Regions , Animals , Antagomirs/metabolism , Apoptosis/drug effects , Bcl-2-Like Protein 11/antagonists & inhibitors , Bcl-2-Like Protein 11/genetics , Bcl-2-Like Protein 11/metabolism , Cells, Cultured , Female , Male , Mice , Mice, Inbred C57BL , MicroRNAs/antagonists & inhibitors , MicroRNAs/genetics , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Up-Regulation/drug effects
7.
Antioxid Redox Signal ; 31(5): 403-419, 2019 08 10.
Article in English | MEDLINE | ID: mdl-30860395

ABSTRACT

Aims: Cardiac-specific overexpression of metallothionein (MT) has been shown to be beneficial in ischemic heart disease, but the detailed mechanisms through which MT protects against myocardial infarction (MI) remain unknown. This study assessed the involvement of the mTORC2/FoxO3a/Bim pathway in the cardioprotective effects of MT. Results: MI was induced in wild-type (FVB) mice and in cardiac-specific MT-overexpressing transgenic (MT-TG) mice by ligation of the left anterior descending (LAD) coronary artery. Cardiac function was better; infarct size and cardiomyocyte apoptosis were lower in MT-TG mice than in FVB mice after MI. Moreover, MT-TG mice exhibited better phenotypes after LAD ligation than FVB mice treated with Mn(III)tetrakis (1-methyl-4-pyridyl) porphyrin pentachloride (MnTMPyP; a reactive oxygen species [ROS] scavenger) and cardiac-specific catalase-overexpressing transgenic (CAT-TG) mice, which showed the same ROS levels as MT-TG mice after MI. Activation of mechanistic target of rapamycin complex 2 (mTORC2) was essential for the cardioprotective effects of MT against MI. In addition, MT attenuated the downregulation of phospho-FoxO3a after MI, inhibiting the expression of the apoptosis-associated gene Bim, located downstream of FoxO3a, and reducing the level of apoptosis after MI. To mimic ischemic-injured FVB and MT-TG mice in vitro, H9c2 and MT-overexpressing H9c2 (H9c2MT7) cardiomyocytes were subjected to oxygen and glucose deprivation, with the results being consistent with those obtained in vivo. Innovation and Conclusion: The cardioprotective effects of MT against MI are not entirely dependent upon its ability to eliminate ROS. Rather, MT overexpression mostly protects against MI through the mTORC2-FoxO3a-Bim pathway.


Subject(s)
Bcl-2-Like Protein 11/antagonists & inhibitors , Cardiotonic Agents/metabolism , Forkhead Box Protein O3/metabolism , Mechanistic Target of Rapamycin Complex 2/metabolism , Metallothionein/metabolism , Myocardial Infarction/metabolism , Animals , Apoptosis , Bcl-2-Like Protein 11/genetics , Bcl-2-Like Protein 11/metabolism , Cells, Cultured , Heart , Mice , Mice, Inbred Strains , Mice, Transgenic , Rats , Reactive Oxygen Species/metabolism , Signal Transduction
8.
J Mol Biol ; 430(18 Pt B): 3041-3050, 2018 09 14.
Article in English | MEDLINE | ID: mdl-30036494

ABSTRACT

Diabetes is a metabolic disorder affecting more than 400 million individuals and their families worldwide. The major forms of diabetes (types 1 and 2) are characterized by pancreatic ß-cell dysfunction and, in some cases, loss of ß-cell mass causing hyperglycemia due to absolute or relative insulin deficiency. The BCL-2 homology 3 (BH3)-only protein BIM has a wide role in apoptosis induction in cells. In this review, we describe the apoptotic mechanisms mediated by BIM activation in ß cells in obesity and both forms of diabetes. We focus on molecular pathways triggered by inflammation, saturated fats, and high levels of glucose. Besides its role in cell death, BIM has been implicated in the regulation of mitochondrial oxidative phosphorylation and cellular metabolism in hepatocytes. BIM is both a key mediator of pancreatic ß-cell death and hepatic insulin resistance and is thus a potential therapeutic target for novel anti-diabetogenic drugs. We consider the implications and challenges of targeting BIM in the treatment of the disease.


Subject(s)
Bcl-2-Like Protein 11/metabolism , Diabetes Mellitus/etiology , Diabetes Mellitus/metabolism , Obesity/etiology , Obesity/metabolism , Peptide Fragments , Protein Interaction Domains and Motifs , Proto-Oncogene Proteins , Animals , Apoptosis , Autoimmunity , Bcl-2-Like Protein 11/antagonists & inhibitors , Bcl-2-Like Protein 11/chemistry , Diabetes Mellitus/diagnosis , Diabetes Mellitus/drug therapy , Hepatocytes/metabolism , Humans , Insulin/metabolism , Insulin Resistance , Insulin-Secreting Cells/immunology , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/pathology , Mitochondria/metabolism , Molecular Targeted Therapy , Obesity/drug therapy , Peptide Fragments/chemistry , Proto-Oncogene Proteins/chemistry , Signal Transduction
9.
Oncogene ; 37(16): 2122-2136, 2018 04.
Article in English | MEDLINE | ID: mdl-29391601

ABSTRACT

Phosphorylation of Ser/Thr residues is a well-established modulating mechanism of the pro-apoptotic function of the BH3-only protein Bim. However, nothing is known about the putative tyrosine phosphorylation of this Bcl-2 family member and its potential impact on Bim function and subsequent Bax/Bak-mediated cytochrome c release and apoptosis. As we have previously shown that the tyrosine kinase Lyn could behave as an anti-apoptotic molecule, we investigated whether this Src family member could directly regulate the pro-apoptotic function of Bim. In the present study, we show that Bim is phosphorylated onto tyrosine residues 92 and 161 by Lyn, which results in an inhibition of its pro-apoptotic function. Mechanistically, we show that Lyn-dependent tyrosine phosphorylation of Bim increases its interaction with anti-apoptotic members such as Bcl-xL, therefore limiting mitochondrial outer membrane permeabilization and subsequent apoptosis. Collectively, our data uncover one molecular mechanism through which the oncogenic tyrosine kinase Lyn negatively regulates the mitochondrial apoptotic pathway, which may contribute to the transformation and/or the chemotherapeutic resistance of cancer cells.


Subject(s)
Apoptosis/genetics , Bcl-2-Like Protein 11/physiology , src-Family Kinases/physiology , Animals , Bcl-2-Like Protein 11/antagonists & inhibitors , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Cells, Cultured , Drug Resistance, Neoplasm/genetics , HEK293 Cells , HeLa Cells , Humans , K562 Cells , Mice , Mitochondria/genetics , Mitochondria/metabolism , Oncogenes/physiology , Signal Transduction/genetics , src-Family Kinases/genetics
10.
Cell Death Differ ; 25(2): 241-254, 2018 02.
Article in English | MEDLINE | ID: mdl-28960205

ABSTRACT

While the association of Epstein-Barr virus (EBV) with Burkitt lymphoma (BL) has long been recognised, the precise role of the virus in BL pathogenesis is not fully resolved. EBV can be lost spontaneously from some BL cell lines, and these EBV-loss lymphoma cells reportedly have a survival disadvantage. Here we have generated an extensive panel of EBV-loss clones from multiple BL backgrounds and examined their phenotype comparing them to their isogenic EBV-positive counterparts. We report that, while loss of EBV from BL cells is rare, it is consistently associated with an enhanced predisposition to undergo apoptosis and reduced tumorigenicity in vivo. Importantly, reinfection of EBV-loss clones with EBV, but surprisingly not transduction with individual BL-associated latent viral genes, restored protection from apoptosis. Expression profiling and functional analysis of apoptosis-related proteins and transcripts in BL cells revealed that EBV inhibits the upregulation of the proapoptotic BH3-only proteins, BIM and PUMA. We conclude that latent EBV genes cooperatively enhance the survival of BL cells by suppression of the intrinsic apoptosis pathway signalling via inhibition of the potent apoptosis initiators, BIM and PUMA.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Bcl-2-Like Protein 11/metabolism , Burkitt Lymphoma/metabolism , Herpesvirus 4, Human/metabolism , Proto-Oncogene Proteins/metabolism , Apoptosis/genetics , Apoptosis Regulatory Proteins/antagonists & inhibitors , Bcl-2-Like Protein 11/antagonists & inhibitors , Burkitt Lymphoma/pathology , Cell Line , Herpesvirus 4, Human/genetics , Herpesvirus 4, Human/growth & development , Humans , Proto-Oncogene Proteins/antagonists & inhibitors
11.
Cell Death Dis ; 8(8): e3026, 2017 08 31.
Article in English | MEDLINE | ID: mdl-29048431

ABSTRACT

BIM, a pro-apoptotic BH3-only protein, is a key regulator of the intrinsic (or mitochondrial) apoptosis pathway. Here, we show that BIM induction by endoplasmic reticulum (ER) stress is suppressed in rat PC12 cells overexpressing heat shock protein B1 (HSPB1 or HSP27) and that this is due to enhanced proteasomal degradation of BIM. HSPB1 and BIM form a complex that immunoprecipitates with p-ERK1/2. We found that HSPB1-mediated proteasomal degradation of BIM is dependent on MEK-ERK signaling. Other studies have shown that several missense mutations in HSPB1 cause the peripheral neuropathy, Charcot-Marie-Tooth (CMT) disease, which is associated with nerve degeneration. Here we show that cells overexpressing CMT-related HSPB1 mutants exhibited increased susceptibility to ER stress-induced cell death and high levels of BIM. These findings identify a novel function for HSPB1 as a negative regulator of BIM protein stability leading to protection against ER stress-induced apoptosis, a function that is absent in CMT-associated HSPB1 mutants.


Subject(s)
Bcl-2-Like Protein 11/genetics , Endoplasmic Reticulum Stress/genetics , HSP27 Heat-Shock Proteins/genetics , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 3/genetics , Animals , Apoptosis/genetics , Bcl-2-Like Protein 11/antagonists & inhibitors , Bcl-2-Like Protein 11/metabolism , Endoplasmic Reticulum/metabolism , Gene Expression Regulation , HSP27 Heat-Shock Proteins/metabolism , Mitochondria/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , PC12 Cells , Phosphorylation , Proteasome Endopeptidase Complex/metabolism , Protein Binding , Proteolysis , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Rats , Signal Transduction
12.
Eur Rev Med Pharmacol Sci ; 21(13): 3088-3097, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28742197

ABSTRACT

OBJECTIVE: Ischemia hypoxia induces cardiomyocyte (CM) apoptosis in the process of acute myocardial infarction (AMI). It was showed that pro-apoptosis factor BIM participates in regulating tumor cell apoptosis under ischemia or hypoxia condition, while its role in CM apoptosis after AMI is still unclear. It was revealed that miR-24 expression was significantly reduced in myocardial tissue after AMI. Bioinformatics analysis exhibits that miR-24 is targeted to the 3'-UTR of BIM. This study aims to investigate the role of miR-24 in mediating BIM expression and CM apoptosis. PATIENTS AND METHODS: Dual-luciferase assay was used to confirm the targeted regulation between miR-24 and BIM. Cells were cultured under ischemia hypoxia for 12 h after transfection for 48 h. Cell apoptosis was tested by using flow cytometry. The caspase activity was detected by using spectrophotometry. Wistar rats were divided into four groups, including Sham, AMI, AMI + agomir-control, and AMI + agomir-24 groups. Cardiac function was evaluated by using echocardiography. CM apoptosis was determined by using TUNEL. Infarction area was measured by using evans blue staining. MiR-24 targeted suppressed BIM expression. RESULTS: MiR-24 mimic and/or si-BIM transfection significantly declined the BIM expression, inhibited caspase-9 and caspase-3 activities, and reduced cell apoptosis in H9C2 cells. MiR-24 expression was decreased, while BIM levels were up-regulated in myocardium after AMI. Agomir-24 injection down-regulated the BIM expression in myocardium, reduced CM apoptosis, narrowed infarction area, and improved cardiac function in rats. CONCLUSIONS: MiR-24 was reduced, whereas BIM was enhanced in the CM after AMI. MiR-24 up-regulation plays a critical role in decreasing BIM expression, reducing CM apoptosis, and improving cardiac function after AMI.


Subject(s)
Apoptosis , Bcl-2-Like Protein 11/metabolism , MicroRNAs/metabolism , 3' Untranslated Regions , Animals , Antagomirs/metabolism , Bcl-2-Like Protein 11/antagonists & inhibitors , Bcl-2-Like Protein 11/genetics , Caspase 3/metabolism , Caspase 9/metabolism , Cell Hypoxia , Cell Line , Down-Regulation , Echocardiography , Heart Ventricles/diagnostic imaging , Male , MicroRNAs/antagonists & inhibitors , MicroRNAs/genetics , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocardial Infarction/veterinary , Myocardium/metabolism , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Rats , Rats, Wistar , Up-Regulation
13.
Apoptosis ; 22(3): 381-392, 2017 Mar.
Article in English | MEDLINE | ID: mdl-27873035

ABSTRACT

3,5-Dimethyl-7H-furo[3,2-g]chromen-7-one (DMFC) is a coumarin derivative with anti-cancer activity against human hepatoma cells, but the mechanisms underlying DMFC function in cancer suppression is unknown. In this study, we aimed at elucidating the molecular mechanisms underlying DMFC anti-cancer activity and determining whether DMFC is effective in suppression of drug-resistant human hepatocellular carcinoma. We show here that DMFC effectively suppresses both the parent and the multidrug-resistant hepatoma cell growth in vitro and DMFC suppresses hepatoma cell growth at least in part through inducing tumor cell apoptosis. In the molecular level, we observed that DMFC treatment decreases Bcl-2 level by a post-transcriptional mechanism and activates Bim transcription to increase Bim mRNA and protein level in hepatoma cells. Furthermore, co-immunoprecipitation studies revealed that DMFC-induced Bim interrupts interactions between Bcl-2 and Bax and between Mcl-1 and Bak, resulting in dissociation of Bax from Bcl-2 and Bak from Mcl-1 and subsequent activation of both Bax and Bak. Activation of Bax and Bak leads to mitochondrial outer membrane permeabilization and cytochrome c release. Consistent with its potent apoptosis-inducing activity, DMFC exhibited potent activity against the multidrug-resistant hepatoma xenograft growth in vivo. Therefore, we determine that DMFC suppresses hepatoma growth through decreasing Bcl-2 and increasing Bim to induce tumor cell apoptosis and hold great promise for further development as a therapeutic agent to treat chemoresistant hepatoma.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Bcl-2-Like Protein 11/physiology , Benzofurans/pharmacology , Carcinoma, Hepatocellular/metabolism , Coumarins/pharmacology , Drug Resistance, Neoplasm/drug effects , Liver Neoplasms/metabolism , Neoplasm Proteins/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , bcl-2 Homologous Antagonist-Killer Protein/metabolism , bcl-2-Associated X Protein/metabolism , Animals , Antineoplastic Agents/chemistry , Bcl-2-Like Protein 11/antagonists & inhibitors , Bcl-2-Like Protein 11/biosynthesis , Bcl-2-Like Protein 11/genetics , Benzofurans/chemistry , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Coumarins/chemistry , Drug Resistance, Multiple/drug effects , Drug Resistance, Multiple/physiology , Drug Resistance, Neoplasm/physiology , Hep G2 Cells , Humans , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Mice , Mice, Nude , Molecular Structure , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Protein Conformation/drug effects , RNA Interference , Transcription, Genetic/drug effects , Xenograft Model Antitumor Assays
14.
Zhongguo Fei Ai Za Zhi ; 19(11): 789-792, 2016 Nov 20.
Article in Chinese | MEDLINE | ID: mdl-27866524

ABSTRACT

B-cell lymphoma 2 interacting mediator of cell death (BIM) plays an important role in the progress of cell apoptosis. The lowering expression level or functional defect of which may have an negative effect on the efficacy of anticancer drugs and the prognosis of postoperative patients with non-small cell lung cancer (NSCLC). This review aims to summarize the structure and function of BIM, as well as the relationship between BIM and the therapeutic efficacy of NSCLC.


Subject(s)
Bcl-2-Like Protein 11/metabolism , Carcinoma, Non-Small-Cell Lung/metabolism , Lung Neoplasms/metabolism , Animals , Bcl-2-Like Protein 11/antagonists & inhibitors , Bcl-2-Like Protein 11/chemistry , Bcl-2-Like Protein 11/genetics , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/therapy , Humans , Lung Neoplasms/genetics , Lung Neoplasms/therapy
15.
Cell Death Dis ; 7: e2177, 2016 Apr 07.
Article in English | MEDLINE | ID: mdl-27054332

ABSTRACT

B-cell acute lymphoblastic leukemia (B-ALL) is an aggressive hematological disease that kills ~50% of adult patients. With the exception of some BCR-ABL1(+) patients who benefit from tyrosine kinase inhibitors, there are no effective targeted therapies for adult B-ALL patients and chemotherapy remains first-line therapy despite adverse side effects and poor efficacy. We show that, although the MEK/ERK pathway is activated in B-ALL cells driven by different oncogenes, MEK inhibition does not suppress B-ALL cell growth. However, MEK inhibition synergized with BCL-2/BCL-XL family inhibitors to suppress proliferation and induce apoptosis in B-ALL cells. We show that this synergism is mediated by the pro-apoptotic factor BIM, which is dephosphorylated as a result of MEK inhibition, allowing it to bind to and neutralize MCL-1, thereby enhancing BCL-2/BCL-XL inhibitor-induced cell death. This cooperative effect is observed in B-ALL cells driven by a range of genetic abnormalities and therefore has significant therapeutic potential.


Subject(s)
Bcl-2-Like Protein 11/metabolism , MAP Kinase Kinase Kinases/antagonists & inhibitors , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Aniline Compounds/pharmacology , Aniline Compounds/therapeutic use , Animals , Apoptosis/drug effects , B-Lymphocytes/cytology , B-Lymphocytes/metabolism , Bcl-2-Like Protein 11/antagonists & inhibitors , Bcl-2-Like Protein 11/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Female , Humans , MAP Kinase Kinase Kinases/metabolism , MAP Kinase Signaling System/drug effects , Mice , Myeloid Cell Leukemia Sequence 1 Protein/antagonists & inhibitors , Myeloid Cell Leukemia Sequence 1 Protein/genetics , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/mortality , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Pyridones/pharmacology , Pyridones/therapeutic use , Pyrimidinones/pharmacology , Pyrimidinones/therapeutic use , RNA Interference , Sulfonamides/pharmacology , Sulfonamides/therapeutic use , Thioglycolates/pharmacology , Thioglycolates/therapeutic use , bcl-X Protein/antagonists & inhibitors , bcl-X Protein/genetics , bcl-X Protein/metabolism
16.
Cell Death Differ ; 23(9): 1483-92, 2016 09 01.
Article in English | MEDLINE | ID: mdl-27035620

ABSTRACT

Loss of cellular adhesion leads to the progression of breast cancer through acquisition of anchorage independence, also known as resistance to anoikis. Although inactivation of E-cadherin is essential for acquisition of anoikis resistance, it has remained unclear how metastatic breast cancer cells counterbalance the induction of apoptosis without E-cadherin-dependent cellular adhesion. We report here that E-cadherin inactivation in breast cancer cells induces PI3K/AKT-dependent FOXO3 inhibition and identify FOXO3 as a novel and direct transcriptional activator of the pro-apoptotic protein BMF. As a result, E-cadherin-negative breast fail to upregulate BMF upon transfer to anchorage independence, leading to anoikis resistance. Conversely, expression of BMF in E-cadherin-negative metastatic breast cancer cells is sufficient to inhibit tumour growth and dissemination in mice. In conclusion, we have identified repression of BMF as a major cue that underpins anoikis resistance and tumour dissemination in E-cadherin-deficient metastatic breast cancer.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Breast Neoplasms/pathology , Cadherins/metabolism , Forkhead Box Protein O3/metabolism , Adaptor Proteins, Signal Transducing/antagonists & inhibitors , Adaptor Proteins, Signal Transducing/genetics , Animals , Anoikis/drug effects , Apoptosis/drug effects , Bcl-2-Like Protein 11/antagonists & inhibitors , Bcl-2-Like Protein 11/genetics , Bcl-2-Like Protein 11/metabolism , Breast Neoplasms/drug therapy , Breast Neoplasms/mortality , Cadherins/genetics , Cell Line, Tumor , Doxycycline/pharmacology , Doxycycline/therapeutic use , Female , Humans , Lung Neoplasms/pathology , Lung Neoplasms/secondary , MCF-7 Cells , Mice , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , RNA Interference , Signal Transduction , Transcriptional Activation
17.
Blood ; 127(18): 2219-30, 2016 05 05.
Article in English | MEDLINE | ID: mdl-26851293

ABSTRACT

Two classes of novel agents, NEDD8-activating enzyme (NAE) and histone deacetylase (HDAC) inhibitors, have shown single-agent activity in acute myelogenous leukemia (AML)/myelodysplastic syndrome (MDS). Here we examined mechanisms underlying interactions between the NAE inhibitor pevonedistat (MLN4924) and the approved HDAC inhibitor belinostat in AML/MDS cells. MLN4924/belinostat coadministration synergistically induced AML cell apoptosis with or without p53 deficiency or FLT3-internal tandem duplication (ITD), whereas p53 short hairpin RNA (shRNA) knockdown or enforced FLT3-ITD expression significantly sensitized cells to the regimen. MLN4924 blocked belinostat-induced antiapoptotic gene expression through nuclear factor-κB inactivation. Each agent upregulated Bim, and Bim knockdown significantly attenuated apoptosis. Microarrays revealed distinct DNA damage response (DDR) genetic profiles between individual vs combined MLN4924/belinostat exposure. Whereas belinostat abrogated the MLN4924-activated intra-S checkpoint through Chk1 and Wee1 inhibition/downregulation, cotreatment downregulated multiple homologous recombination and nonhomologous end-joining repair proteins, triggering robust double-stranded breaks, chromatin pulverization, and apoptosis. Consistently, Chk1 or Wee1 shRNA knockdown significantly sensitized AML cells to MLN4924. MLN4924/belinostat displayed activity against primary AML or MDS cells, including those carrying next-generation sequencing-defined poor-prognostic cancer hotspot mutations, and CD34(+)/CD38(-)/CD123(+) populations, but not normal CD34(+) progenitors. Finally, combined treatment markedly reduced tumor burden and significantly prolonged animal survival (P < .0001) in AML xenograft models with negligible toxicity, accompanied by pharmacodynamic effects observed in vitro. Collectively, these findings argue that MLN4924 and belinostat interact synergistically by reciprocally disabling the DDR in AML/MDS cells. This strategy warrants further consideration in AML/MDS, particularly in disease with unfavorable genetic aberrations.


Subject(s)
Cyclopentanes/therapeutic use , DNA Damage , DNA Repair/drug effects , Histone Deacetylase Inhibitors/therapeutic use , Hydroxamic Acids/therapeutic use , Leukemia, Myeloid, Acute/drug therapy , Molecular Targeted Therapy , Myelodysplastic Syndromes/drug therapy , Pyrimidines/therapeutic use , Sulfonamides/therapeutic use , Ubiquitin-Activating Enzymes/antagonists & inhibitors , Animals , Apoptosis/drug effects , Bcl-2-Like Protein 11/antagonists & inhibitors , Bcl-2-Like Protein 11/genetics , Cell Cycle Proteins/antagonists & inhibitors , Cell Cycle Proteins/genetics , Cells, Cultured , Checkpoint Kinase 1/antagonists & inhibitors , Checkpoint Kinase 1/genetics , Cyclopentanes/pharmacology , Drug Synergism , Histone Deacetylase Inhibitors/pharmacology , Humans , Hydroxamic Acids/pharmacology , Kaplan-Meier Estimate , Leukemia, Myeloid, Acute/pathology , Mice , Myelodysplastic Syndromes/pathology , NF-kappa B/metabolism , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , Neoplasm Proteins/physiology , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/genetics , Protein Processing, Post-Translational/drug effects , Protein-Tyrosine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/genetics , Pyrimidines/pharmacology , RNA Interference , RNA, Small Interfering/genetics , S Phase Cell Cycle Checkpoints/drug effects , Sulfonamides/pharmacology , U937 Cells , Xenograft Model Antitumor Assays
18.
Cell Cycle ; 15(3): 394-402, 2016.
Article in English | MEDLINE | ID: mdl-26694174

ABSTRACT

Doxorubicin and other anthracycline compounds exert their anti-cancer effects by causing DNA damage and initiating cell cycle arrest in cancer cells, followed by apoptosis. DNA damage generally activates a p53-mediated pathway to initiate apoptosis by increasing the level of the BH3-only protein, Puma. However, p53-mediated apoptosis in response to DNA damage has not yet been validated in prostate cancers. In the current study, we used LNCaP and PC3 prostate cancer cells, representing wild type p53 and a p53-null model, to determine if DNA damage activates p53-mediated apoptosis in prostate cancers. Our results revealed that PC3 cells were 4 to 8-fold less sensitive than LNCaP cells to doxorubicin-inuced apoptosis. We proved that the differential response of LNCaP and PC3 to doxorubicin was p53-independent by introducing wild-type or dominant negative p53 into PC3 or LNCaP cells, respectively. By comparing several apoptosis-related proteins in both cell lines, we found that Bcl-xl proteins were much more abundant in PC3 cells than in LNCaP cells. We further demonstrated that Bcl-xl protects LNCaP and PC3 cells from doxorubicin-induced apoptosis by using ABT-263, an inhibitor of Bcl-xl, as a single agent or in combination with doxorubicin to treat LNCaP or PC3 cells. Bcl-xl rather than p53, likely contributes to the differential response of LNCaP and PC3 to doxorubicin in apoptosis. Finally, co-immunoprecipitation and siRNA analysis revealed that a BH3-only protein, Bim, is involved in doxorubicin-induced apoptosis by directly counteracting Bcl-xl.


Subject(s)
Apoptosis/drug effects , Bcl-2-Like Protein 11/metabolism , Doxorubicin/pharmacology , Tumor Suppressor Protein p53/metabolism , bcl-X Protein/metabolism , Aniline Compounds/pharmacology , Antineoplastic Agents/pharmacology , Apoptosis Regulatory Proteins/metabolism , Bcl-2-Like Protein 11/antagonists & inhibitors , Bcl-2-Like Protein 11/genetics , Cell Line, Tumor , Cyclin-Dependent Kinase Inhibitor p21/metabolism , DNA Damage/drug effects , Humans , Immunoblotting , Immunoprecipitation , Male , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Proto-Oncogene Proteins/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Sulfonamides/pharmacology , Transfection , Tumor Suppressor Protein p53/genetics , bcl-X Protein/antagonists & inhibitors
SELECTION OF CITATIONS
SEARCH DETAIL
...