Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
PLoS Pathog ; 18(6): e1010555, 2022 06.
Article in English | MEDLINE | ID: mdl-35666761

ABSTRACT

The reservoir of latently HIV-1 infected cells is heterogeneous. To achieve an HIV-1 cure, the reservoir of activatable proviruses must be eliminated while permanently silenced proviruses may be tolerated. We have developed a method to assess the proviral nuclear microenvironment in single cells. In latently HIV-1 infected cells, a zinc finger protein tethered to the HIV-1 promoter produced a fluorescent signal as a protein of interest came in its proximity, such as the viral transactivator Tat when recruited to the nascent RNA. Tat is essential for viral replication. In these cells we assessed the proviral activation and chromatin composition. By linking Tat recruitment to proviral activity, we dissected the mechanisms of HIV-1 latency reversal and the consequences of HIV-1 production. A pulse of promoter-associated Tat was identified that contrasted to the continuous production of viral proteins. As expected, promoter H3K4me3 led to substantial expression of the provirus following T cell stimulation. However, the activation-induced cell cycle arrest and death led to a surviving cell fraction with proviruses encapsulated in repressive chromatin. Further, this cellular model was used to reveal mechanisms of action of small molecules. In a proof-of-concept study we determined the effect of modifying enhancer chromatin on HIV-1 latency reversal. Only proviruses resembling active enhancers, associated with H3K4me1 and H3K27ac and subsequentially recognized by BRD4, efficiently recruited Tat upon cell stimulation. Tat-independent HIV-1 latency reversal of unknown significance still occurred. We present a method for single cell assessment of the microenvironment of the latent HIV-1 proviruses, used here to reveal how T cell stimulation modulates the proviral activity and how the subsequent fate of the infected cell depends on the chromatin context.


Subject(s)
HIV Infections , HIV Seropositivity , HIV-1 , CD4-Positive T-Lymphocytes , Cell Cycle Proteins/genetics , Chromatin , HIV-1/genetics , Humans , Nuclear Proteins/genetics , Proviruses/physiology , T-Lymphocytes , Transcription Factors/genetics , Virus Latency/genetics
2.
Nat Microbiol ; 5(9): 1144-1157, 2020 09.
Article in English | MEDLINE | ID: mdl-32541947

ABSTRACT

Quiescence is a hallmark of CD4+ T cells latently infected with human immunodeficiency virus 1 (HIV-1). While reversing this quiescence is an effective approach to reactivate latent HIV from T cells in culture, it can cause deleterious cytokine dysregulation in patients. As a key regulator of T-cell quiescence, FOXO1 promotes latency and suppresses productive HIV infection. We report that, in resting T cells, FOXO1 inhibition impaired autophagy and induced endoplasmic reticulum (ER) stress, thereby activating two associated transcription factors: activating transcription factor 4 (ATF4) and nuclear factor of activated T cells (NFAT). Both factors associate with HIV chromatin and are necessary for HIV reactivation. Indeed, inhibition of protein kinase R-like ER kinase, an ER stress sensor that can mediate the induction of ATF4, and calcineurin, a calcium-dependent regulator of NFAT, synergistically suppressed HIV reactivation induced by FOXO1 inhibition. Thus, our studies uncover a link of FOXO1, ER stress and HIV infection that could be therapeutically exploited to selectively reverse T-cell quiescence and reduce the size of the latent viral reservoir.


Subject(s)
Endoplasmic Reticulum Stress/drug effects , Forkhead Box Protein O1/metabolism , Forkhead Box Protein O1/pharmacology , HIV-1/drug effects , Virus Activation/drug effects , Virus Latency/drug effects , Activating Transcription Factor 4/metabolism , CD4-Positive T-Lymphocytes/virology , Forkhead Box Protein O1/genetics , Gene Knockdown Techniques , HIV Infections/virology , Humans , K562 Cells
3.
NPJ Aging Mech Dis ; 5: 7, 2019.
Article in English | MEDLINE | ID: mdl-31602311

ABSTRACT

Aging is characterized by the progressive loss of physiological function in all organisms. Remarkably, the aging process can be modulated by environmental modifications, including diet and small molecules. The natural compound nordihydroguaiaretic acid (NDGA) robustly increases lifespan in flies and mice, but its mechanism of action remains unclear. Here, we report that NDGA is an inhibitor of the epigenetic regulator p300. We find that NDGA inhibits p300 acetyltransferase activity in vitro and suppresses acetylation of a key p300 target in histones (i.e., H3K27) in cells. We use the cellular thermal shift assay to uniquely demonstrate NDGA binding to p300 in cells. Finally, in agreement with recent findings indicating that p300 is a potent blocker of autophagy, we show that NDGA treatment induces autophagy. These findings identify p300 as a target of NDGA and provide mechanistic insight into its role in longevity.

4.
Nucleic Acids Res ; 46(11): 5587-5600, 2018 06 20.
Article in English | MEDLINE | ID: mdl-29733381

ABSTRACT

The HSF and FOXO families of transcription factors play evolutionarily conserved roles in stress resistance and lifespan. In humans, the rs2802292 G-allele at FOXO3 locus has been associated with longevity in all human populations tested; moreover, its copy number correlated with reduced frequency of age-related diseases in centenarians. At the molecular level, the intronic rs2802292 G-allele correlated with increased expression of FOXO3, suggesting that FOXO3 intron 2 may represent a regulatory region. Here we show that the 90-bp sequence around the intronic single nucleotide polymorphism rs2802292 has enhancer functions, and that the rs2802292 G-allele creates a novel HSE binding site for HSF1, which induces FOXO3 expression in response to diverse stress stimuli. At the molecular level, HSF1 mediates the occurrence of a promoter-enhancer interaction at FOXO3 locus involving the 5'UTR and the rs2802292 region. These data were confirmed in various cellular models including human HAP1 isogenic cell lines (G/T). Our functional studies highlighted the importance of the HSF1-FOXO3-SOD2/CAT/GADD45A cascade in cellular stress response and survival by promoting ROS detoxification, redox balance and DNA repair. Our findings suggest the existence of an HSF1-FOXO3 axis in human cells that could be involved in stress response pathways functionally regulating lifespan and disease susceptibility.


Subject(s)
Enhancer Elements, Genetic , Forkhead Box Protein O3/genetics , Heat Shock Transcription Factors/metabolism , Polymorphism, Single Nucleotide , Stress, Physiological/genetics , Transcriptional Activation , 5' Untranslated Regions , Alleles , Binding Sites , Cell Line , Cell Survival , Cells, Cultured , Forkhead Box Protein O3/biosynthesis , Humans , Introns , Longevity/genetics , Promoter Regions, Genetic
5.
Cell Death Dis ; 9(2): 231, 2018 02 14.
Article in English | MEDLINE | ID: mdl-29445193

ABSTRACT

While aberrant cancer cell growth is frequently associated with altered biochemical metabolism, normal mitochondrial functions are usually preserved and necessary for full malignant transformation. The transcription factor FoxO3A is a key determinant of cancer cell homeostasis, playing a dual role in survival/death response to metabolic stress and cancer therapeutics. We recently described a novel mitochondrial arm of the AMPK-FoxO3A axis in normal cells upon nutrient shortage. Here, we show that in metabolically stressed cancer cells, FoxO3A is recruited to the mitochondria through activation of MEK/ERK and AMPK, which phosphorylate serine 12 and 30, respectively, on FoxO3A N-terminal domain. Subsequently, FoxO3A is imported and cleaved to reach mitochondrial DNA, where it activates expression of the mitochondrial genome to support mitochondrial metabolism. Using FoxO3A-/- cancer cells generated with the CRISPR/Cas9 genome editing system and reconstituted with FoxO3A mutants being impaired in their nuclear or mitochondrial subcellular localization, we show that mitochondrial FoxO3A promotes survival in response to metabolic stress. In cancer cells treated with chemotherapeutic agents, accumulation of FoxO3A into the mitochondria promoted survival in a MEK/ERK-dependent manner, while mitochondrial FoxO3A was required for apoptosis induction by metformin. Elucidation of FoxO3A mitochondrial vs. nuclear functions in cancer cell homeostasis might help devise novel therapeutic strategies to selectively disable FoxO3A prosurvival activity.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Nucleus/metabolism , Forkhead Box Protein O3/genetics , Gene Expression Regulation, Neoplastic , Mitochondria/metabolism , AMP-Activated Protein Kinases/genetics , AMP-Activated Protein Kinases/metabolism , Animals , Apoptosis/drug effects , Apoptosis/genetics , CRISPR-Cas Systems , Cell Line, Tumor , Cell Nucleus/drug effects , Cell Survival , Cisplatin/pharmacology , Extracellular Signal-Regulated MAP Kinases/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Fluorouracil/pharmacology , Forkhead Box Protein O3/metabolism , Gene Editing , Genome, Mitochondrial , HEK293 Cells , Humans , Irinotecan/pharmacology , MAP Kinase Kinase Kinases/genetics , MAP Kinase Kinase Kinases/metabolism , Metformin/pharmacology , Mice , Mice, Inbred C57BL , Mitochondria/drug effects , Mitochondria/genetics , NIH 3T3 Cells , Phosphorylation , Signal Transduction , Stress, Physiological/drug effects , Stress, Physiological/genetics
6.
Mol Pharm ; 15(3): 729-736, 2018 03 05.
Article in English | MEDLINE | ID: mdl-29240439

ABSTRACT

Nonsteroidal anti-inflammatory drugs (NSAIDs) are well-known for their effects on inflammatory gene expression. Although NSAIDs are known to impact multiple cellular signaling mechanisms, a recent finding is that the NSAID salicylate can disrupt histone acetylation, in part through direct inhibition of the lysine acetyltransferase (KAT) p300/CBP. While salicylate is a relatively weak KAT inhibitor, its CoA-linked metabolite is more potent; however, the ability of NSAID metabolites to inhibit KAT enzymes biochemically and in cells remains relatively unexplored. Here we define the role of metabolic and nonmetabolic mechanisms in inhibition of KAT activity by NSAID chemotypes. First, we screen a small panel of NSAIDs for biochemical inhibition of the prototypical KAT p300, leading to the finding that many carboxylate-containing NSAIDs, including ibuprofen, are able to function as weak inhibitors. Assessing the inhibition of p300 by ibuprofen-CoA, a known NSAID metabolite, reveals that linkage of ibuprofen to CoA increases its biochemical potency toward p300 and other KAT enzymes. In cellular studies, we find that carboxylate-containing NSAIDs inhibit histone acetylation. Finally, we exploit the stereoselective metabolism of ibuprofen to assess the role of its acyl-CoA metabolite in regulation of histone acetylation. This unique strategy reveals that formation of ibuprofen-CoA and histone acetylation are poorly correlated, suggesting metabolism may not be required for ibuprofen to inhibit histone acetylation. Overall, these studies provide new insights into the ability of NSAIDs to alter histone acetylation, and illustrate how selective metabolism may be leveraged as a tool to explore the influence of metabolic acyl-CoAs on cellular enzyme activity.


Subject(s)
Acetylation/drug effects , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , E1A-Associated p300 Protein/antagonists & inhibitors , Histone Code/drug effects , Acyl Coenzyme A/metabolism , E1A-Associated p300 Protein/metabolism , Enzyme Assays/methods , Gene Expression Regulation/drug effects , HEK293 Cells , Histones/metabolism , Humans , Metabolic Networks and Pathways/drug effects , Protein Processing, Post-Translational/drug effects , Recombinant Proteins/metabolism
7.
Mol Carcinog ; 55(11): 1584-1597, 2016 Nov.
Article in English | MEDLINE | ID: mdl-26373689

ABSTRACT

Most tumors primarily rely on glycolysis rather than mitochondrial respiration for ATP production. This phenomenon, also known as Warburg effect, renders tumors more sensitive to glycolytic disturbances compared to normal cells. 3-bromopyruvate is a potent inhibitor of glycolysis that shows promise as an anticancer drug candidate. Although investigations revealed that 3-BP triggers apoptosis through ATP depletion and subsequent AMPK activation, the underlying molecular mechanisms coupling AMPK to apoptosis are poorly understood. We showed that 3-BP leads to a rapid ATP depletion which was followed by growth inhibition and Bax-dependent apoptosis in HCT116 cells. Apoptosis was accompanied with activation of caspase-9 and -3 while pretreatment with a general caspase inhibitor attenuated cell death. AMPK, p38, JNK, and Akt were phosphorylated immediately upon treatment. Pharmacological inhibition and silencing of AMPK largely inhibited 3-BP-induced apoptosis and reversed phosphorylation of JNK. Transcriptional activity of FoxO3a was dramatically increased subsequent to AMPK-mediated phosphorylation of FoxO3a at Ser413. Cell death analysis of cells transiently transfected with wt or AMPK-phosphorylation-deficient FoxO3 expression plasmids verified the contributory role of AMPK-FoxO3a axis in 3-BP-induced apoptosis. In addition, expression of proapoptotic Bcl-2 proteins Bim and Bax were upregulated in an AMPK-dependent manner. Bim was transcriptionally activated in association with FoxO3a activity, while Bax upregulation was abolished in p53-null cells. Together, these data suggest that AMPK couples 3-BP-induced metabolic disruption to intrinsic apoptosis via modulation of FoxO3a-Bim axis and Bax expression. © 2015 Wiley Periodicals, Inc.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Forkhead Box Protein O3/genetics , Neoplasms/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Pyruvates/pharmacology , Adenosine Triphosphate/metabolism , Apoptosis , Cell Proliferation/drug effects , Cell Survival/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Glycolysis/drug effects , HCT116 Cells , HeLa Cells , Humans , Neoplasms/drug therapy , Neoplasms/genetics , Signal Transduction/drug effects , Transcriptional Activation/drug effects , Up-Regulation
8.
World J Gastroenterol ; 20(29): 9744-58, 2014 Aug 07.
Article in English | MEDLINE | ID: mdl-25110412

ABSTRACT

Colorectal cancer (CRC) remains one of the most common malignancies in the world. Although surgical resection combined with adjuvant therapy is effective at the early stages of the disease, resistance to conventional therapies is frequently observed in advanced stages, where treatments become ineffective. Resistance to cisplatin, irinotecan and 5-fluorouracil chemotherapy has been shown to involve mitogen-activated protein kinase (MAPK) signaling and recent studies identified p38α MAPK as a mediator of resistance to various agents in CRC patients. Studies published in the last decade showed a dual role for the p38α pathway in mammals. Its role as a negative regulator of proliferation has been reported in both normal (including cardiomyocytes, hepatocytes, fibroblasts, hematopoietic and lung cells) and cancer cells (colon, prostate, breast, lung tumor cells). This function is mediated by the negative regulation of cell cycle progression and the transduction of some apoptotic stimuli. However, despite its anti-proliferative and tumor suppressor activity in some tissues, the p38α pathway may also acquire an oncogenic role involving cancer related-processes such as cell metabolism, invasion, inflammation and angiogenesis. In this review, we summarize current knowledge about the predominant role of the p38α MAPK pathway in CRC development and chemoresistance. In our view, this might help establish the therapeutic potential of the targeted manipulation of this pathway in clinical settings.


Subject(s)
Antineoplastic Agents/therapeutic use , Colorectal Neoplasms/drug therapy , Drug Resistance, Neoplasm , Mitogen-Activated Protein Kinase 14/antagonists & inhibitors , Protein Kinase Inhibitors/therapeutic use , Animals , Autophagy/drug effects , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Colorectal Neoplasms/enzymology , Colorectal Neoplasms/pathology , Humans , Mitogen-Activated Protein Kinase 14/metabolism , Molecular Targeted Therapy , Signal Transduction/drug effects , Treatment Outcome
9.
Curr Pharm Des ; 20(18): 3052-67, 2014.
Article in English | MEDLINE | ID: mdl-24079770

ABSTRACT

Aging is a stage of life of all living organisms. According to the free-radical theory, aging cells gradually become unable to maintain cellular homeostasis due to the adverse effects of reactive oxygen species (ROS). ROS can cause irreversible DNA mutations, protein and lipid damage which are increasingly accumulated in the course of time if cells could not overcome these effects by the antioxidant defence system. Accrued damaged molecules in cells may either induce cellular death or contribute to develop various pathologies. Hence, programmed cell death mechanisms, apoptosis and autophagy, play a vital role in the aging process. Although they are strictly controlled by various interconnected signalling pathways, alterations in their regulations may contribute to severe pathologies including cancer, Alzheimer's and Parkinson's diseases. In this review, we summarized our current understanding and hypotheses regarding oxidative stress and age-related dysregulation of cell death signalling pathways.


Subject(s)
Aging/physiology , Cell Death/physiology , Oxidative Stress/physiology , Animals , Antioxidants/metabolism , Apoptosis/physiology , Autophagy/physiology , Free Radicals/metabolism , Homeostasis/physiology , Humans , Reactive Oxygen Species/metabolism , Signal Transduction/physiology
10.
Cancer Lett ; 344(1): 110-118, 2014 Mar 01.
Article in English | MEDLINE | ID: mdl-24215867

ABSTRACT

Chemoresistance is a major obstacle to effective therapy against colorectal cancer (CRC) and may lead to deadly consequences. The metabolism of CRC cells depends highly on the p38 MAPK pathway, whose involvement in maintaining a chemoresistant behavior is currently being investigated. Our previous studies revealed that p38α is the main p38 isoform in CRC cells. Here we show that p38α pharmacological inhibition combined with cisplatin administration decreases colony formation and viability of cancer cells and strongly increases Bax-dependent apoptotic cell death by activating the tumor suppressor protein FoxO3A. Our results indicate that FoxO3A activation up-regulates transcription of its target genes (p21, PTEN, Bim and GADD45), which forces both chemosensitive and chemoresistant CRC cells to undergo apoptosis. Additionally, we found that FoxO3A is required for apoptotic cell death induction, as confirmed by RNA interference experiments. In animal models xenografted with chemoresistant HT29 cells, we further confirmed that the p38-targeted dual therapy strategy produced an increase in apoptosis in cancer tissue leading to tumor regression. Our study uncovers a major role for the p38-FoxO3A axis in chemoresistance, thereby suggesting a new therapeutic approach for CRC treatment; moreover, our results indicate that Bax status may be used as a predictive biomarker.


Subject(s)
Colorectal Neoplasms/metabolism , Drug Resistance, Neoplasm/physiology , Forkhead Transcription Factors/metabolism , Mitogen-Activated Protein Kinase 14/antagonists & inhibitors , Molecular Targeted Therapy/methods , Animals , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Survival , Cisplatin/pharmacology , Female , Flow Cytometry , Fluorescent Antibody Technique , Forkhead Box Protein O3 , HT29 Cells , Humans , Immunoblotting , Mice , Mice, Nude , Real-Time Polymerase Chain Reaction , Xenograft Model Antitumor Assays
11.
Cell Signal ; 24(6): 1361-8, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22313691

ABSTRACT

Chemotherapeutic drugs proved only 50% successful in breast cancer because of cell type-dependent resistance mechanisms. FOXO3 is known to be involved in the regulation of several cell death-related genes; however, the extent of FOXO3 regulation in chemoresistance is still not fully understood. Here, we show that FOXO3 critically mediates cisplatin chemosensitivity of MCF-7 breast cancer cells which express higher levels of FOXO3 compared to resistant MDA-MB-231 cells. Administration of cisplatin induces apoptosis in MCF-7 cells in a FOXO3-dependent manner as indicated by RNA interference. On the other hand, IKK-ß (IκB kinase) appears to inhibit FOXO3 action after cisplatin treatment and promotes chemoresistance in MDA-MB-231 cells. IKK-ß directly interacts and sequesters FOXO3 in the cytosol preventing its nuclear localization. Moreover, cisplatin treatment induces autophagosome formation through LC-3 conversion while inhibiting the cleavage of caspase 9 and caspase 3 in MDA-MB-231 cells manipulated to overexpress FOXO3. In brief, our findings demonstrate that in addition to cellular level of active FOXO3, cisplatin chemoresistance is also regulated by IKK-ß sequestration of FOXO3 in cytosol.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Cisplatin/pharmacology , Drug Resistance, Neoplasm , Forkhead Transcription Factors/metabolism , I-kappa B Kinase/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Death/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Female , Forkhead Box Protein O3 , Forkhead Transcription Factors/genetics , Humans , Up-Regulation
12.
J Cell Physiol ; 227(11): 3556-65, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22262057

ABSTRACT

Apoptosis of macrophage foam cells loaded with modified/oxidized lipids is implicated in destabilization of advanced atherosclerotic plaques in humans. Concentration of HNE, main aldehydic product of plasma LDL peroxidation, elevates in atherosclerotic lesions as well as in cultured cells under oxidative stress. Although this reactive aldehyde has been shown to promote apoptosis with the involvement of p38 MAPK and JNK in various mammalian cell lines, roles of B-cell lymphoma 2 (Bcl-2) family proteins remain to be deciphered. We demonstrated that HNE-induced apoptosis was accompanied by concurrent downregulations of antiapoptotic Bcl-x(L) and Mcl-1 as well as upregulation of proapoptotic Bak. Furthermore, phoshorylation of Bcl-2 at Thr56, Ser70, and probably more phosphorylation sites located on N-terminal loop domain associated with HNE-induced apoptosis in both U937 and HeLa cells while ectopic expression of a phospho-defective Bcl-2 mutant significantly attenuated apoptosis. In parallel to this, HNE treatment caused release of proapoptotic Bax from Bcl-2. Pharmacological inhbition of IKK inhibited HNE-induced Bcl-2 phosphorylation. Similarly, silencing IKKα and -ß both ended up with abrogation of Bcl-2 phosphorylation along with attenuation of apoptosis. Moreover, both IKKα and -ß coimmunoprecipitated with Bcl-2 and in vitro kinase assay proved the ability of IKK to phosphorylate Bcl-2. In view of these findings and considering HNE inhibits DNA-binding activity of nuclear factor-κB (NF-κB) through prevention of IκB phosphorylation/ubiquitination/proteolysis, IKK appears to directly interfere with Bcl-2 activity through phosphorylation in HNE-mediated apoptosis independent of NF-κB signaling.


Subject(s)
Aldehydes , Apoptosis , I-kappa B Kinase/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Aldehydes/administration & dosage , Aldehydes/metabolism , Apoptosis/drug effects , Apoptosis/physiology , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , HeLa Cells , Humans , I-kappa B Kinase/genetics , Macrophages/physiology , Myeloid Cell Leukemia Sequence 1 Protein , NF-kappa B/metabolism , Oxidative Stress , Phosphorylation , Proto-Oncogene Proteins c-bcl-2/genetics , Signal Transduction , U937 Cells , bcl-2-Associated X Protein/metabolism , bcl-X Protein/metabolism
13.
Breast Cancer Res Treat ; 119(2): 271-81, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19238538

ABSTRACT

Inhibition or downregulation of Bcl-2 represents a new therapeutic approach to by-pass chemoresistance in cancer cells. Therefore, we explored the potential of this approach in breast cancer cells. Cisplatin and paclitaxel induced apoptosis in a dose-dependent manner in MCF-7 (drug-sensitive) and MDA-MB-231 (drug-insensitive) cells. Furthermore, when we transiently silenced Bcl-2, both cisplatin and paclitaxel induced apoptosis more than parental cells. Dose dependent induction of apoptosis by drugs was enhanced by the pre-treatment of these cells with HA14-1, a Bcl-2 inhibitor. Although the effect of cisplatin was significant on both cell lines, the effect of paclitaxel was much less potent only in MDA-MB-231 cells. To further understand the distinct role of drugs in MDA-MB-231 cells pretreated with HA14-1, caspases and Bcl-2 family proteins were studied. The apoptotic effect of cisplatin with or without HA14-1 pre-treatment is shown to be caspase-dependent. Among pro-apoptotic Bcl-2 proteins, Bax and Puma were found to be up-regulated whereas Bcl-2 and Bcl-x(L) were down-regulated when cells were pretreated with HA14-1 followed by paclitaxel or cisplatin. Enforced Bcl-2 expression in MDA-MB-231 cells abrogated the sensitizing effect of HA14-1 in cisplatin induced apoptosis. These results suggest that the potentiating effect of HA14-1 is drug and cell type specific and may not only depend on the inhibition of Bcl-2. Importantly, alteration of other pro-apoptotic or anti-apoptotic Bcl-2 family members may dictate the apoptotic response when HA14-1 is combined with chemotherapeutic drugs.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Apoptosis/drug effects , Breast Neoplasms/pathology , Drug Resistance, Neoplasm/drug effects , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Apoptosis Regulatory Proteins/metabolism , Benzopyrans/pharmacology , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Caspases/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Cisplatin/pharmacology , Dose-Response Relationship, Drug , Female , Humans , Nitriles/pharmacology , Paclitaxel/pharmacology , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , RNA Interference , Time Factors , Transfection , bcl-2-Associated X Protein/metabolism , bcl-X Protein/metabolism
14.
Carcinogenesis ; 30(9): 1517-27, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19578044

ABSTRACT

Increased expression of antiapoptotic Bcl-2 proteins confers therapeutic resistance in various cancer types. Targeting Bcl-2 proteins by small molecules or activating alternative pathways to bypass Bcl-2-mediated protection to promote apoptosis are two approaches to overcoming therapeutic resistance. Here, we show that cisplatin triggers a Bak-dependent pathway to induce apoptosis in Bcl-2-overexpressing MCF-7 cells. p53-mediated induction of Noxa expression, generation of lipid peroxidation end products and induction of Noxa-Mcl-1 interaction are necessary for this pathway to function. Although Puma is also induced by cisplatin treatment, it is not required for apoptosis. Similarly, reactive oxygen species production by cisplatin did not have any effect on cisplatin-induced apoptosis in MCF-7 Bcl-2 cells. Furthermore, p53 promotes cisplatin-induced apoptosis by directly binding and counteracting Bcl-x(L) antiapoptotic function. In conclusion, our findings suggest a novel mode of action for cisplatin to overcome Bcl-2-mediated protection against apoptosis, which requires preferential activation of Bak and p53-mediated upregulation of Noxa protein levels and lipid peroxidation.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cisplatin/pharmacology , Lipid Peroxidation/drug effects , Proto-Oncogene Proteins c-bcl-2/physiology , bcl-2 Homologous Antagonist-Killer Protein/physiology , Cell Line, Tumor , Drug Resistance, Neoplasm , Humans , Myeloid Cell Leukemia Sequence 1 Protein , Reactive Oxygen Species/metabolism , Tumor Suppressor Protein p53/physiology , bcl-2-Associated X Protein/physiology , bcl-X Protein/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...