Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Trends Cancer ; 6(8): 688-701, 2020 08.
Article in English | MEDLINE | ID: mdl-32451306

ABSTRACT

Cancer cells survive and adapt to many types of stress including hypoxia, nutrient deprivation, metabolic, and oxidative stress. These stresses are sensed by diverse cellular signaling processes, leading to either degradation of mitochondria or alleviation of mitochondrial stress. This review discusses signaling during sensing and mitigation of stress involving mitochondrial communication with the endoplasmic reticulum, and how retrograde signaling upregulates the mitochondrial stress response to maintain mitochondrial integrity. The importance of the mitochondrial unfolded protein response, an emerging pathway that alleviates cellular stress, will be elaborated with respect to cancer. Detailed understanding of cellular pathways will establish mitochondrial stress response as a key mechanism for cancer cell survival leading to cancer progression and resistance, and provide a potential therapeutic target in cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Mitochondria/pathology , Neoplasms/pathology , Unfolded Protein Response/drug effects , Activating Transcription Factors/metabolism , Antineoplastic Agents/therapeutic use , Cell Survival/drug effects , Chaperonin 60/metabolism , Disease Progression , Drug Resistance, Neoplasm/drug effects , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/pathology , Endoplasmic Reticulum Stress/drug effects , Humans , Mitochondria/drug effects , Mitochondrial Proteins/metabolism , Neoplasms/drug therapy , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects
2.
Biochim Biophys Acta Mol Cell Res ; 1867(1): 118573, 2020 01.
Article in English | MEDLINE | ID: mdl-31678591

ABSTRACT

Cytochrome c (Cyt c) released from mitochondria interacts with Apaf-1 to form the heptameric apoptosome, which initiates the caspase cascade to execute apoptosis. Although lysine residue at 72 (K72) of Cyt c plays an important role in the Cyt c-Apaf-1 interaction, the underlying mechanism of interaction between Cyt c and Apaf-1 is still not clearly defined. Here we identified multiple lysine residues including K72, which are also known to interact with ATP, to play a key role in Cyt c-Apaf-1 interaction. Mutation of these lysine residues abrogates the apoptosome formation causing inhibition of caspase activation. Using in-silico molecular docking, we have identified Cyt c-binding interface on Apaf-1. Although mutant Cyt c shows higher affinity for Apaf-1, the presence of Cyt c-WT restores the apoptosome activity. ATP addition modulates only mutant Cyt c binding to Apaf-1 but not WT Cyt c binding to Apaf-1. Using TCGA and cBioPortal, we identified multiple mutations in both Apaf-1 and Cyt c that are predicted to interfere with apoptosome assembly. We also demonstrate that transcript levels of various enzymes involved with dATP or ATP synthesis are increased in various cancers. Silencing of nucleotide metabolizing enzymes such as ribonucleotide reductase subunit M1 (RRM1) and ATP-producing glycolytic enzymes PKM2 attenuated ATP production and enhanced caspase activation. These findings suggest important role for lysine residues of Cyt c and nucleotides in the regulation of apoptosome-dependent apoptotic cell death as well as demonstrate how these mutations and nucleotides may have a pivotal role in human diseases such as cancer.


Subject(s)
Apoptosomes/physiology , Cytochromes c/chemistry , Molecular Docking Simulation , Neoplasms/pathology , Nucleotides/chemistry , Alanine/chemistry , Alanine/genetics , Amino Acid Substitution , Apoptosomes/chemistry , Apoptotic Protease-Activating Factor 1/chemistry , Apoptotic Protease-Activating Factor 1/metabolism , Case-Control Studies , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cells, Cultured , Cytochromes c/genetics , Cytochromes c/metabolism , Female , Humans , Lysine/chemistry , Lysine/genetics , Male , Models, Molecular , Mutant Proteins/chemistry , Mutant Proteins/metabolism , Mutant Proteins/physiology , Neoplasms/genetics , Neoplasms/metabolism , Nucleotides/metabolism , PC-3 Cells , Protein Binding/genetics , Protein Interaction Mapping , Protein Multimerization/genetics , Signal Transduction/genetics
3.
Br J Cancer ; 121(11): 934-943, 2019 11.
Article in English | MEDLINE | ID: mdl-31673102

ABSTRACT

BACKGROUND: Interleukin-8 (IL-8) and heat shock protein 60 (Hsp60) play crucial roles in cell survival and maintenance of cellular homoeostasis. However, cross talks between these two proteins are not defined. METHODS: IL-8 expression in tumour tissue sections was analysed by immunohistochemistry. IL-8 expression and release in cancer cells was quantified using enzyme-linked immunosorbent assay (ELISA). Apoptosis was quantified using caspase activity and Annexin-V/PI staining. RESULTS: We observed IL-8 release from cancer cells in response to histone deacetylase inhibitor, apicidin (Api), and non-competitive inhibitor of the sarco/endoplasmic reticulum Ca2+ ATPase, thapsigargin (TG). IL-8 release was increased upon TG-treatment. TG-induced IL-8 expression was reduced in the presence of Api in Bax-dependent manner. Increased apoptosis was associated with decreased IL-8 expression in response to combined treatment of TG and Api. TG and Api combination induced caspase-8 and caspase-9 dependent apoptosis. Hsp60 knockdown abrogated IL-8 expression induced by Api, TG, and their combination. The level of TGF-ß, an upstream regulator of IL-8, was decreased upon Hsp60-silencing. Knocking down Hsp60 decreased IL-8 expression and its release in prostate cancer cell xenograft tumours in SCID mice. CONCLUSION: This study describes the underlying mechanism associated with apoptosis resistance mediated via Hsp60-IL-8 axis in cancer.


Subject(s)
Apoptosis/drug effects , Chaperonin 60/metabolism , Interleukin-8/metabolism , Mitochondrial Proteins/metabolism , Neoplasms/metabolism , Animals , Caspase 8/genetics , Caspase 9/genetics , Chaperonin 60/genetics , Gene Knockdown Techniques , HCT116 Cells , Heterografts , Humans , Interleukin-8/genetics , Male , Mice , Mice, SCID , Mitochondrial Proteins/genetics , Neoplasms/pathology , PC-3 Cells , Peptides, Cyclic/pharmacology , Signal Transduction/drug effects , Thapsigargin/pharmacology
4.
Cancer Res ; 79(7): 1353-1368, 2019 04 01.
Article in English | MEDLINE | ID: mdl-30765600

ABSTRACT

Although African-American (AA) patients with prostate cancer tend to develop greater therapeutic resistance and faster prostate cancer recurrence compared with Caucasian-American (CA) men, the molecular mechanisms of this racial prostate cancer disparity remain undefined. In this study, we provide the first comprehensive evidence that cytochrome c deficiency in AA primary tumors and cancer cells abrogates apoptosome-mediated caspase activation and contributes to mitochondrial dysfunction, thereby promoting therapeutic resistance and prostate cancer aggressiveness in AA men. In AA prostate cancer cells, decreased nuclear accumulation of nuclear respiration factor 1 (Nrf1) and its subsequent loss of binding to the cytochrome c promoter mediated cytochrome c deficiency. The activation of cellular Myc (c-Myc) and NF-κB or inhibition of AKT prevented nuclear translocation of Nrf1. Genetic and pharmacologic inhibition of c-Myc and NF-κB or activation of AKT promoted Nrf1 binding to cytochrome c promoter, cytochrome c expression, caspase activation, and cell death. The lack of p-Drp1S616 in AA prostate cancer cells contributed to defective cytochrome c release and increased resistance to apoptosis, indicating that restoration of cytochrome c alone may be insufficient to induce effective apoptosis. Cytochrome c deficiency promoted the acquisition of glycolytic phenotypes and mitochondrial dysfunction, whereas cytochrome c restoration via inhibition of c-Myc and NF-κB or activation of AKT attenuated glycolysis in AA prostate cancer cells. Inhibition of c-Myc and NF-κB enhanced the efficacy of docetaxel in tumor xenografts. Therefore, restoring cytochrome c may overcome therapeutic resistance and prostate cancer aggressiveness in AA men. Overall, this study provides the first comprehensive experimental, mechanistic, and clinical evidence for apoptosome and mitochondrial dysfunction in prostate cancer racial disparity. SIGNIFICANCE: Mechanistic insights on prostate cancer health disparity among American men provide novel approaches to restore mitochondrial function, which can address therapeutic resistance and aggressiveness in African-American men with prostate cancer.


Subject(s)
Apoptosomes/physiology , Black or African American , Cytochromes c/deficiency , Mitochondria/physiology , Prostatic Neoplasms/pathology , Animals , Cell Line, Tumor , Cytochromes c/metabolism , Humans , Male , Mice , Mice, SCID , Mitochondrial Membranes/enzymology , NF-kappa B/metabolism , Nuclear Respiratory Factor 1/metabolism , Oxidative Phosphorylation , Proto-Oncogene Proteins c-myc/metabolism
5.
Cancer Lett ; 413: 82-93, 2018 01 28.
Article in English | MEDLINE | ID: mdl-29107110

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is highly aggressive disease and current treatment regimens fail to effectively cure PDAC. Development of resistance to current therapy is one of the key reasons for this outcome. Nimbolide (NL), a triterpenoid obtained from Azadirachta indica, exhibits anticancer properties in various cancer including PDAC cells. However, the underlying mechanism of this anticancer agent in PDAC cells remains undefined. We show that NL exerts a higher level of apoptotic cell death compared to the first-line agent gemcitabine for PDAC, as well as other anticancer agents including sorafenib and curcumin. The anticancer efficacy of NL was further evidenced by a reduction in the CD44+ as well as cancer stem-like cell (CSC) population, as it causes decreased sphere formation. Mechanistically, the anticancer efficacy of NL associates with reduced mutant p53 as well as increased mitochondrial activity in the form of increased mitochondrial reactive oxygen species and mitochondrial mass. Together, this study highlights the therapeutic potential of NL in mutant p53 expressing pancreatic cancer.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis/drug effects , Carcinoma, Pancreatic Ductal/drug therapy , Caspase Inhibitors/pharmacology , Hyaluronan Receptors/metabolism , Limonins/pharmacology , Mitochondria/drug effects , Neoplastic Stem Cells/drug effects , Pancreatic Neoplasms/drug therapy , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Cell Movement/drug effects , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Dose-Response Relationship, Drug , Humans , Mitochondria/metabolism , Mitochondria/pathology , Mutation , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Reactive Oxygen Species/metabolism , Time Factors , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Gemcitabine
6.
Cancer Lett ; 397: 52-60, 2017 07 01.
Article in English | MEDLINE | ID: mdl-28342983

ABSTRACT

Metastatic castration-resistant prostate cancer (mCRPC) remains incurable and is one of the leading causes of cancer-related death among American men. Therefore, detection of prostate cancer (PCa) at early stages may reduce PCa-related mortality in men. We show that lipid quantification by vibrational Raman Microspectroscopy and Biomolecular Component Analysis may serve as a potential biomarker in PCa. Transcript levels of lipogenic genes including sterol regulatory element-binding protein-1 (SREBP-1) and its downstream effector fatty acid synthase (FASN), and rate-limiting enzyme acetyl CoA carboxylase (ACACA) were upregulated corresponding to both Gleason score and pathologic T stage in the PRAD TCGA cohort. Increased lipid accumulation in late-stage transgenic adenocarcinoma of mouse prostate (TRAMP) tumors compared to early-stage TRAMP and normal prostate tissues were observed. FASN along with other lipogenesis enzymes, and SREBP-1 proteins were upregulated in TRAMP tumors compared to wild-type prostatic tissues. Genetic alterations of key lipogenic genes predicted the overall patient survival using TCGA PRAD cohort. Correlation between lipid accumulation and tumor stage provides quantitative marker for PCa diagnosis. Thus, Raman spectroscopy-based lipid quantification could be a sensitive and reliable tool for PCa diagnosis and staging.


Subject(s)
Adenocarcinoma/metabolism , Biomarkers, Tumor/analysis , Lipogenesis , Prostatic Neoplasms, Castration-Resistant/metabolism , Spectrum Analysis, Raman , Adenocarcinoma/enzymology , Adenocarcinoma/genetics , Adenocarcinoma/secondary , Animals , Biomarkers, Tumor/genetics , Computational Biology , Databases, Genetic , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Genetic Predisposition to Disease , Humans , Lipogenesis/genetics , Male , Mice , Mice, Transgenic , Neoplasm Grading , Neoplasm Staging , Phenotype , Predictive Value of Tests , Prostatic Neoplasms, Castration-Resistant/enzymology , Prostatic Neoplasms, Castration-Resistant/genetics , Prostatic Neoplasms, Castration-Resistant/pathology , RNA, Messenger/genetics
7.
Biochim Biophys Acta Rev Cancer ; 1867(1): 58-66, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27988298

ABSTRACT

Abrogation of endoplasmic reticulum (ER) protein folding triggered by exogenous or endogenous factors, stimulates a cellular stress response, termed ER stress. ER stress re-establishes ER homeostasis through integrated signaling termed the ER-unfolded protein response (UPRER). In the presence of severe toxic or prolonged ER stress, the pro-survival function of UPRER is transformed into a lethal signal transmitted to and executed through mitochondria. Mitochondria are key for both apoptotic and autophagic cell death. Thus ER is vital in sensing and coordinating stress pathways to maintain overall physiological homeostasis. However, this function is deregulated in cancer, resulting in resistance to apoptosis induction in response to various stressors including therapeutic agents. Here we review the connections between ER stress and mitochondrial apoptosis, describing potential cancer therapeutic targets.


Subject(s)
Apoptosis/physiology , Endoplasmic Reticulum Stress/physiology , Endoplasmic Reticulum/pathology , Mitochondria/pathology , Neoplasms/pathology , Unfolded Protein Response/physiology , Animals , Humans , Protein Folding , Signal Transduction/physiology
8.
Front Biosci (Schol Ed) ; 9(1): 154-164, 2017 01 01.
Article in English | MEDLINE | ID: mdl-27814581

ABSTRACT

The gap between prostate cancer disparities among American men is narrowing, which is mostly due to increased screening of African American (AA) men. However, the biological reasons for prostate cancer disparities among American men still remain undefined. Mitochondrion, an organelle within cells, regulates both cell survival and cell death mechanisms. These cellular signaling pathways require various proteins localized to mitochondria, which are encoded by both nuclear DNA (nDNA) and mitochondrial DNA (mtDNA). Interestingly, prostate tissues from AA men harbor reduced mtDNA content compared to Caucasian American (CA) men. Therefore, changes in mitochondrial genes may have detrimental consequences in terms of cellular signaling regulated by mitochondria in AA men. This review describes the plausible underlying mechanism of mtDNA depletion and its impact in driving resistance to therapy leading to faster progression and poor prognosis in African American men with prostate cancer. Since defective cellular signaling is critical for prostate cancer cell survival, restoring mitochondrial function may provide strategies to alleviate prostate cancer disparities among American men.


Subject(s)
Black or African American , Health Status Disparities , Mitochondria/genetics , Mitochondria/metabolism , Prostatic Neoplasms/ethnology , Prostatic Neoplasms/physiopathology , White People , DNA, Mitochondrial/genetics , DNA, Mitochondrial/metabolism , Humans , Male , Oxidative Phosphorylation , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , United States
9.
Mol Oncol ; 10(7): 949-65, 2016 08.
Article in English | MEDLINE | ID: mdl-27106131

ABSTRACT

Development of therapeutic resistance is responsible for most prostate cancer (PCa) related mortality. Resistance has been attributed to an acquired or selected cancer stem cell phenotype. Here we report the histone deacetylase inhibitor apicidin (APC) or ER stressor thapsigargin (TG) potentiate paclitaxel (TXL)-induced apoptosis in PCa cells and limit accumulation of cancer stem cells. TXL-induced responses were modulated in the presence of TG with increased accumulation of cells at G1-phase, rearrangement of the cytoskeleton, and changes in cytokine release. Cytoskeletal rearrangement was associated with modulation of the cytoplasmic and mitochondrial unfolded protein response leading to mitochondrial dysfunction and release of proapoptotic proteins from mitochondria. TXL in combination with APC or TG enhanced caspase activation. Importantly, TXL in combination with TG induced caspase activation and apoptosis in X-ray resistant LNCaP cells. Increased release of transforming growth factor-beta (TGF-ß) was observed while phosphorylated ß-catenin level was suppressed with TXL combination treatments. This was accompanied by a decrease in the CD44(+)CD133(+) cancer stem cell-like population, suggesting treatment affects cancer stem cell properties. Taken together, combination treatment with TXL and either APC or TG induces efficient apoptosis in both proliferating and cancer stem cells, suggesting this therapeutic combination may overcome drug resistance and recurrence in PCa.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Apoptosis , Cytoskeleton/metabolism , Mitochondria/metabolism , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/metabolism , Unfolded Protein Response , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Apoptosis/drug effects , Apoptosis/radiation effects , Caspases/metabolism , Cell Cycle Checkpoints/drug effects , Cell Cycle Checkpoints/radiation effects , Cell Death/drug effects , Cell Death/radiation effects , Cell Line, Tumor , Cytoskeleton/drug effects , Cytoskeleton/radiation effects , Enzyme Activation/drug effects , G1 Phase/drug effects , G1 Phase/radiation effects , G2 Phase/drug effects , G2 Phase/radiation effects , HSP70 Heat-Shock Proteins/metabolism , Humans , Interferon-gamma/metabolism , Interleukin-8/metabolism , Male , Matrix Metalloproteinases/metabolism , Membrane Potential, Mitochondrial/drug effects , Membrane Potential, Mitochondrial/radiation effects , Mitochondria/drug effects , Mitochondria/radiation effects , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Neoplastic Stem Cells/radiation effects , Paclitaxel , Peptides, Cyclic/pharmacology , Peptides, Cyclic/therapeutic use , Phosphorylation/drug effects , Prostatic Neoplasms/pathology , Prostatic Neoplasms/radiotherapy , Reactive Oxygen Species/metabolism , Thapsigargin/pharmacology , Thapsigargin/therapeutic use , Transforming Growth Factor beta/metabolism , Unfolded Protein Response/drug effects , Unfolded Protein Response/radiation effects , X-Rays , beta Catenin/metabolism
10.
Free Radic Biol Med ; 90: 261-71, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26627937

ABSTRACT

We have previously reported that neem limonoids (neem) induce multiple cancer cell death pathways. Here we dissect the underlying mechanisms of neem-induced apoptotic cell death in cancer. We observed that neem-induced caspase activation does not require Bax/Bak channel-mediated mitochondrial outer membrane permeabilization, permeability transition pore, and mitochondrial fragmentation. Neem enhanced mitochondrial DNA and mitochondrial biomass. While oxidative phosphorylation (OXPHOS) Complex-I activity was decreased, the activities of other OXPHOS complexes including Complex-II and -IV were unaltered. Increased reactive oxygen species (ROS) levels were associated with an increase in mitochondrial biomass and apoptosis upon neem exposure. Complex-I deficiency due to the loss of Ndufa1-encoded MWFE protein inhibited neem-induced caspase activation and apoptosis, but cell death induction was enhanced. Complex II-deficiency due to the loss of succinate dehydrogenase complex subunit C (SDHC) robustly decreased caspase activation, apoptosis, and cell death. Additionally, the ablation of Complexes-I, -III, -IV, and -V together did not inhibit caspase activation. Together, we demonstrate that neem limonoids target OXPHOS system to induce cancer cell death, which does not require upregulation or activation of proapoptotic Bcl-2 family proteins.


Subject(s)
Apoptosis/drug effects , Azadirachta/chemistry , Limonins/pharmacology , Neoplasms/pathology , Oxidative Phosphorylation , Caspases/metabolism , Cyclin-Dependent Kinase Inhibitor p21/physiology , DNA, Mitochondrial/analysis , Dynamins , Electron Transport Complex I/physiology , GTP Phosphohydrolases/analysis , HCT116 Cells , Humans , Microtubule-Associated Proteins/analysis , Mitochondrial Membrane Transport Proteins/physiology , Mitochondrial Permeability Transition Pore , Mitochondrial Proteins/analysis , Neoplasms/drug therapy , Tumor Suppressor Protein p53/physiology
11.
Drug Discov Today ; 21(1): 38-47, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26232549

ABSTRACT

X-chromosome-linked inhibitor of apoptosis protein (XIAP) has an important regulatory role in programmed cell death by inhibiting the caspase cascade. Activation of XIAP-dependent signaling culminates into regulation of multiple cellular processes including apoptosis, innate immunity, epithelial-to-mesenchymal transition, cell migration, invasion, metastasis and differentiation. Although XIAP localizes to the cytosolic compartment, XIAP-mediated cellular signaling encompasses mitochondrial and post-mitochondrial levels. Recent findings demonstrate that XIAP also localizes to mitochondria and regulates mitochondria functions. XIAP acts upstream of mitochondrial cytochrome c release and modulates caspase-dependent apoptosis. The new function of XIAP has potential to enhance mitochondrial membrane permeabilization and other cellular functions controlling cytochrome c release. These findings could exploit the overexpression of XIAP in human tumors for therapeutic benefits.


Subject(s)
Mitochondria/metabolism , Mitochondrial Membranes/metabolism , Neoplasms/metabolism , X-Linked Inhibitor of Apoptosis Protein/metabolism , Animals , Apoptosis/physiology , Cell Membrane Permeability/physiology , Cytochromes c/metabolism , Humans , Signal Transduction/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...