Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Cell Death Dis ; 15(4): 276, 2024 Apr 18.
Article in English | MEDLINE | ID: mdl-38637497

ABSTRACT

The Unfolded Protein Response (UPR) is an essential cellular process activated by the accumulation of unfolded proteins within the Endoplasmic Reticulum (ER), a condition referred to as ER stress. Three ER anchored receptors, IRE1, PERK and ATF6 act as ER stress sensors monitoring the health of the ER. Upon detection of ER stress, IRE1, PERK and ATF6 initiate downstream signaling pathways collectively referred to as the UPR. The overarching aim of the UPR is to restore ER homeostasis by reducing ER stress, however if that is not possible, the UPR transitions from a pro-survival to a pro-death response. While our understanding of the key signaling pathways central to the UPR is well defined, the same is not true of the subtle signaling events that help fine tune the UPR, supporting its ability to adapt to varying amplitudes or durations of ER stress. In this study, we demonstrate cross talk between the IRE1 and PERK branches of the UPR, wherein IRE1 via XBP1s signaling helps to sustain PERK expression during prolonged ER stress. Our findings suggest cross talk between UPR branches aids adaptiveness thereby helping to support the plasticity of UPR signaling responses.


Subject(s)
Protein Serine-Threonine Kinases , eIF-2 Kinase , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , eIF-2 Kinase/genetics , eIF-2 Kinase/metabolism , Endoplasmic Reticulum Stress/physiology , Signal Transduction , Unfolded Protein Response
2.
Mol Neurobiol ; 60(3): 1476-1485, 2023 Mar.
Article in English | MEDLINE | ID: mdl-36478320

ABSTRACT

Examination of post-mortem brain tissues has previously revealed a strong association between Parkinson's disease (PD) pathophysiology and endoplasmic reticulum (ER) stress. Evidence in the literature regarding the circulation of ER stress-regulated factors released from neurons provides a rationale for investigating ER stress biomarkers in the blood to aid diagnosis of PD. The levels of ER stress-regulated proteins in serum collected from 29 PD patients and 24 non-PD controls were measured using enzyme-linked immunosorbent assays. A panel of four biomarkers, protein disulfide-isomerase A1, protein disulfide-isomerase A3, mesencephalic astrocyte-derived neurotrophic factor, and clusterin, together with age and gender had higher ability (area under the curve 0.64, sensitivity 66%, specificity 57%) and net benefit to discriminate PD patients from the non-PD group compared with other analyzed models. Addition of oligomeric and total α-synuclein to the model did not improve the diagnostic power of the biomarker panel. We provide evidence that ER stress-regulated proteins merit further investigation for their potential as diagnostic biomarkers of PD.


Subject(s)
Parkinson Disease , Humans , Parkinson Disease/diagnosis , Parkinson Disease/metabolism , Protein Disulfide-Isomerases/metabolism , alpha-Synuclein/metabolism , Endoplasmic Reticulum Stress/physiology , Molecular Chaperones , Neurons/metabolism
4.
Nat Commun ; 13(1): 2493, 2022 05 06.
Article in English | MEDLINE | ID: mdl-35524156

ABSTRACT

IRE1α is constitutively active in several cancers and can contribute to cancer progression. Activated IRE1α cleaves XBP1 mRNA, a key step in production of the transcription factor XBP1s. In addition, IRE1α cleaves select mRNAs through regulated IRE1α-dependent decay (RIDD). Accumulating evidence implicates IRE1α in the regulation of lipid metabolism. However, the roles of XBP1s and RIDD in this process remain ill-defined. In this study, transcriptome and lipidome profiling of triple negative breast cancer cells subjected to pharmacological inhibition of IRE1α reveals changes in lipid metabolism genes associated with accumulation of triacylglycerols (TAGs). We identify DGAT2 mRNA, encoding the rate-limiting enzyme in TAG biosynthesis, as a RIDD target. Inhibition of IRE1α, leads to DGAT2-dependent accumulation of TAGs in lipid droplets and sensitizes cells to nutritional stress, which is rescued by treatment with the DGAT2 inhibitor PF-06424439. Our results highlight the importance of IRE1α RIDD activity in reprograming cellular lipid metabolism.


Subject(s)
Endoribonucleases , Lipid Metabolism , Neoplasms , Protein Serine-Threonine Kinases , Endoplasmic Reticulum Stress/genetics , Endoribonucleases/genetics , Endoribonucleases/metabolism , Humans , Lipid Metabolism/genetics , Protein Serine-Threonine Kinases/genetics , RNA, Messenger/metabolism , Transcription Factors/metabolism , X-Box Binding Protein 1/genetics , X-Box Binding Protein 1/metabolism
5.
J Cell Mol Med ; 25(18): 8809-8820, 2021 09.
Article in English | MEDLINE | ID: mdl-34363313

ABSTRACT

Stress-induced apoptosis is mediated primarily through the intrinsic pathway that involves caspase-9. We previously reported that in caspase-9-deficient cells, a protein complex containing ATG5 and Fas-associated death domain (FADD) facilitated caspase-8 activation and cell death in response to endoplasmic reticulum (ER) stress. Here, we investigated whether this complex could be activated by other forms of cell stress. We show that diverse stress stimuli, including etoposide, brefeldin A and paclitaxel, as well as heat stress and gamma-irradiation, caused formation of a complex containing ATG5-ATG12, FADD and caspase-8 leading to activation of downstream caspases in caspase-9-deficient cells. We termed this complex the 'stressosome'. However, in these cells, only ER stress and heat shock led to stressosome-dependent cell death. Using in silico molecular modelling, we propose the structure of the stressosome complex, with FADD acting as an adaptor protein, interacting with pro-caspase-8 through their respective death effector domains (DEDs) and interacting with ATG5-ATG12 through its death domain (DD). This suggests that the complex could be regulated by cellular FADD-like interleukin-1ß-converting enzyme-inhibitory protein (cFLIPL ), which was confirmed experimentally. This study provides strong evidence for an alternative mechanism of caspase-8 activation involving the stressosome complex.


Subject(s)
Autophagy-Related Protein 5/metabolism , Caspase 8/metabolism , Caspase 9/metabolism , Endoplasmic Reticulum Stress , Animals , Fibroblasts , HEK293 Cells , Humans , Mice , Mouse Embryonic Stem Cells
6.
J Cell Mol Med ; 25(3): 1359-1370, 2021 02.
Article in English | MEDLINE | ID: mdl-33398919

ABSTRACT

The endoplasmic reticulum (ER) is the site of protein folding and secretion, Ca2+ storage and lipid synthesis in eukaryotic cells. Disruption to protein folding or Ca2+ homeostasis in the ER leads to the accumulation of unfolded proteins, a condition known as ER stress. This leads to activation of the unfolded protein response (UPR) pathway in order to restore protein homeostasis. Three ER membrane proteins, namely inositol-requiring enzyme 1 (IRE1), protein kinase RNA-like ER kinase (PERK) and activating transcription factor 6 (ATF6), sense the accumulation of unfolded/misfolded proteins and are activated, initiating an integrated transcriptional programme. Recent literature demonstrates that activation of these sensors can alter lipid enzymes, thus implicating the UPR in the regulation of lipid metabolism. Given the presence of ER stress and UPR activation in several diseases including cancer and neurodegenerative diseases, as well as the growing recognition of altered lipid metabolism in disease, it is timely to consider the role of the UPR in the regulation of lipid metabolism. This review provides an overview of the current knowledge on the impact of the three arms of the UPR on the synthesis, function and regulation of fatty acids, triglycerides, phospholipids and cholesterol.


Subject(s)
Gene Expression Regulation , Lipid Metabolism , Unfolded Protein Response , Animals , Biomarkers , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum Stress , Humans , Metabolic Networks and Pathways
7.
Cancers (Basel) ; 13(2)2021 Jan 12.
Article in English | MEDLINE | ID: mdl-33445669

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is the most common form of pancreatic cancer and one of the leading causes of cancer-associated deaths in the world. It is characterised by dismal response rates to conventional therapies. A major challenge in treatment strategies for PDAC is the presence of a dense stroma that surrounds the tumour cells, shielding them from treatment. This unique tumour microenvironment is fuelled by paracrine signalling between pancreatic cancer cells and supporting stromal cell types including the pancreatic stellate cells (PSC). While our molecular understanding of PDAC is improving, there remains a vital need to develop effective, targeted treatments. The unfolded protein response (UPR) is an elaborate signalling network that governs the cellular response to perturbed protein homeostasis in the endoplasmic reticulum (ER) lumen. There is growing evidence that the UPR is constitutively active in PDAC and may contribute to the disease progression and the acquisition of resistance to therapy. Given the importance of the tumour microenvironment and cytokine signalling in PDAC, and an emerging role for the UPR in shaping the tumour microenvironment and in the regulation of cytokines in other cancer types, this review explores the importance of the UPR in PDAC biology and its potential as a therapeutic target in this disease.

8.
Mol Pharm ; 17(8): 3009-3023, 2020 08 03.
Article in English | MEDLINE | ID: mdl-32628022

ABSTRACT

The design, synthesis, characterization, and biological activity of a series of platinum(IV) prodrugs containing the axial ligand 3-(4-phenylquinazoline-2-carboxamido)propanoate (L3) are reported. L3 is a derivative of the quinazolinecarboxamide class of ligands that binds to the translocator protein (TSPO) at the outer mitochondrial membrane. The cytotoxicities of cis,cis,trans-[Pt(NH3)2Cl2(L3)(OH)] (C-Pt1), cis,cis,trans-[Pt(NH3)2Cl2(L3)(BZ)] (C-Pt2), trans-[Pt(DACH)(OX)(L3)(OH)] (C-Pt3), and trans-[Pt(DACH)(OX)(L3)(BZ)] (C-Pt4) (DACH: R,R-diaminocyclohexane, BZ: benzoate, OX: oxalate) in MCF-7 breast cancer and noncancerous MCF-10A epithelial cells were assessed and compared with those of cisplatin, oxaliplatin, and the free ligand L3. Moreover, the cellular uptake, ROS generation, DNA damage, and the effect on the mitochondrial function, mitochondrial membrane potential, and morphology were investigated. Molecular interactions of L3 in the TSPO binding site were studied using molecular docking. The results showed that complex C-Pt1 is the most effective Pt(IV) complex and exerts a multimodal mechanism involving DNA damage, potent ROS production, loss of the mitochondrial membrane potential, and mitochondrial damage.


Subject(s)
Antineoplastic Agents/pharmacology , Mitochondria/drug effects , Organoplatinum Compounds/pharmacology , Prodrugs/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , Cisplatin/pharmacology , DNA Damage/drug effects , Epithelial Cells/drug effects , Humans , Ligands , MCF-7 Cells , Mitochondrial Membranes/drug effects , Oxaliplatin/pharmacology , Reactive Oxygen Species/metabolism
9.
Cell Death Dis ; 11(1): 12, 2020 Jan 06.
Article in English | MEDLINE | ID: mdl-31907350

ABSTRACT

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

10.
Cell Death Dis ; 10(9): 622, 2019 08 16.
Article in English | MEDLINE | ID: mdl-31417078

ABSTRACT

The inflammasome is a multiprotein complex assembled in response to Pathogen Associated Molecular Patterns (PAMPs) and Danger Associated Molecular Patterns (DAMPs). Inflammasome activation occurs through a two-step mechanism, with the first signal facilitating priming of inflammasome components while the second signal triggers complex assembly. Once assembled, the inflammasome recruits and activates pro-caspase-1, which in turn processes pro-interleukin (IL)-18 and pro-IL-1ß into their bio-active forms. Owing to its key role in the regulation of innate immune responses, the inflammasome has emerged as a therapeutic target for the treatment of inflammatory conditions. In this study we demonstrate that IRE1α, a key component of the Unfolded Protein Response, contributes to assembly of the NLRP3 inflammasome. Blockade of IRE1α RNase signaling lowered NLRP3 inflammasome assembly, caspase-1 activation and pro-IL-1ß processing. These results underscore both the importance and potential therapeutic relevance of targeting IRE1α signaling in conditions of excessive inflammasome formation.


Subject(s)
Endoribonucleases/antagonists & inhibitors , Inflammasomes/metabolism , Interleukin-1/metabolism , Interleukin-1beta/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Protein Precursors/metabolism , Protein Serine-Threonine Kinases/antagonists & inhibitors , Humans , Inflammasomes/drug effects , Lipopolysaccharides/pharmacology , Nigericin/pharmacology , Signal Transduction , THP-1 Cells , Transfection
11.
Cancers (Basel) ; 10(10)2018 Sep 21.
Article in English | MEDLINE | ID: mdl-30248920

ABSTRACT

In 2018, in the US alone, it is estimated that 268,670 people will be diagnosed with breast cancer, and that 41,400 will die from it. Since breast cancers often become resistant to therapies, and certain breast cancers lack therapeutic targets, new approaches are urgently required. A cell-stress response pathway, the unfolded protein response (UPR), has emerged as a promising target for the development of novel breast cancer treatments. This pathway is activated in response to a disturbance in endoplasmic reticulum (ER) homeostasis but has diverse physiological and disease-specific functions. In breast cancer, UPR signalling promotes a malignant phenotype and can confer tumours with resistance to widely used therapies. Here, we review several roles for UPR signalling in breast cancer, highlighting UPR-mediated therapy resistance and the potential for targeting the UPR alone or in combination with existing therapies.

12.
Nat Commun ; 9(1): 3267, 2018 08 15.
Article in English | MEDLINE | ID: mdl-30111846

ABSTRACT

Triple-negative breast cancer (TNBC) lacks targeted therapies and has a worse prognosis than other breast cancer subtypes, underscoring an urgent need for new therapeutic targets and strategies. IRE1 is an endoplasmic reticulum (ER) stress sensor, whose activation is predominantly linked to the resolution of ER stress and, in the case of severe stress, to cell death. Here we demonstrate that constitutive IRE1 RNase activity contributes to basal production of pro-tumorigenic factors IL-6, IL-8, CXCL1, GM-CSF, and TGFß2 in TNBC cells. We further show that the chemotherapeutic drug, paclitaxel, enhances IRE1 RNase activity and this contributes to paclitaxel-mediated expansion of tumor-initiating cells. In a xenograft mouse model of TNBC, inhibition of IRE1 RNase activity increases paclitaxel-mediated tumor suppression and delays tumor relapse post therapy. We therefore conclude that inclusion of IRE1 RNase inhibition in therapeutic strategies can enhance the effectiveness of current chemotherapeutics.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Endoribonucleases/metabolism , Protein Serine-Threonine Kinases/metabolism , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/metabolism , Xenograft Model Antitumor Assays , Animals , Cell Line , Cell Line, Tumor , Endoribonucleases/antagonists & inhibitors , Endoribonucleases/genetics , Enzyme Inhibitors/administration & dosage , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , HEK293 Cells , Humans , Mice, Nude , Paclitaxel/administration & dosage , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics , RNA Interference , Triple Negative Breast Neoplasms/genetics
13.
Biochem Biophys Res Commun ; 497(1): 115-121, 2018 02 26.
Article in English | MEDLINE | ID: mdl-29421659

ABSTRACT

Receptor-interacting protein 2 (RIP2) is an essential mediator of inflammation and innate immunity, but little is known about its role outside the immune system. Recently, RIP2 has been linked to chemoresistance of triple negative breast cancer (TNBC), the most aggressive breast cancer subtype for which there is an urgent need for targeted therapies. In this study we show that high expression of RIP2 in breast tumors correlates with a worse prognosis and a higher risk of recurrence. We also demonstrate that RIP2 confers TNBC cell resistance against paclitaxel and ceramide-induced apoptosis. Overexpression of RIP2 lead to NF-κB activation, which contributed to higher expression of pro-survival proteins and cell survival. Conversely, RIP2 knockdown inhibited NF-κB signaling, reduced levels of anti-apoptotic proteins and sensitized cells to drug treatment. Together, these data show that RIP2 promotes survival of breast cancer cells through NF-κB activation and that targeting RIP2 may be therapeutically beneficial for treatment of TNBC.


Subject(s)
Cell Survival , NF-kappa B/metabolism , Receptor-Interacting Protein Serine-Threonine Kinase 2/metabolism , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/pathology , Antineoplastic Agents/therapeutic use , Ceramides/therapeutic use , Female , Humans , MCF-7 Cells , Paclitaxel/therapeutic use , Treatment Outcome , Triple Negative Breast Neoplasms/drug therapy , Tumor Cells, Cultured
14.
Biochem Biophys Res Commun ; 495(1): 700-705, 2018 01 01.
Article in English | MEDLINE | ID: mdl-29108999

ABSTRACT

Nerve growth factor (NGF) is the prototypic member of the neurotrophin family and binds two receptors, TrkA and the 75 kDa neurotrophin receptor (p75NTR), through which diverse and sometimes opposing effects are mediated. Using the FoldX protein design algorithm, we generated eight NGF variants with different point mutations predicted to have altered binding to TrkA or p75NTR. Of these, the I31R NGF variant exhibited specific binding to p75NTR. The generation of this NGF variant with selective affinity for p75NTR can be used to enhance understanding of neurotrophin receptor imbalance in diseases and identifies a key targetable residue for the development of small molecules to disrupt binding of NGF to TrkA with potential uses in chronic pain.


Subject(s)
Drug Design , Mutagenesis, Site-Directed/methods , Nerve Growth Factor/biosynthesis , Nerve Growth Factor/chemistry , Protein Engineering/methods , Receptors, Nerve Growth Factor/chemistry , Receptors, Nerve Growth Factor/metabolism , Animals , Binding Sites , Nerve Growth Factor/genetics , PC12 Cells , Protein Binding , Rats , Structure-Activity Relationship
15.
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
16.
EMBO Rep ; 17(10): 1374-1395, 2016 10.
Article in English | MEDLINE | ID: mdl-27629041

ABSTRACT

In response to diverse stress stimuli, eukaryotic cells activate a common adaptive pathway, termed the integrated stress response (ISR), to restore cellular homeostasis. The core event in this pathway is the phosphorylation of eukaryotic translation initiation factor 2 alpha (eIF2α) by one of four members of the eIF2α kinase family, which leads to a decrease in global protein synthesis and the induction of selected genes, including the transcription factor ATF4, that together promote cellular recovery. The gene expression program activated by the ISR optimizes the cellular response to stress and is dependent on the cellular context, as well as on the nature and intensity of the stress stimuli. Although the ISR is primarily a pro-survival, homeostatic program, exposure to severe stress can drive signaling toward cell death. Here, we review current understanding of the ISR signaling and how it regulates cell fate under diverse types of stress.


Subject(s)
Gene Expression Regulation , Signal Transduction , Stress, Physiological , Animals , Gene Expression Regulation/drug effects , Homeostasis , Host-Pathogen Interactions , Humans , Protein Binding , Protein Processing, Post-Translational/drug effects , Signal Transduction/drug effects , Transcription Factors/metabolism
17.
Biochem Biophys Res Commun ; 478(4): 1541-7, 2016 09 30.
Article in English | MEDLINE | ID: mdl-27577679

ABSTRACT

Triple negative breast cancer [TNBC] cells are reported to secrete the neurotrophin nerve growth factor [NGF] and express its receptors, p75 neurotrophin receptor [p75(NTR)] and TrkA, leading to NGF-activated pro-survival autocrine signaling. This provides a rationale for NGF as a potential therapeutic target for TNBC. Here we show that exposure of TNBC cells to NGF leads to increased levels of p75(NTR), which was diminished by NGF-neutralizing antibody or NGF inhibitors [Ro 08-2750 and Y1086]. NGF-mediated increase in p75(NTR) levels were partly due to increased transcription and partly due to inhibition of proteolytic processing of p75(NTR). In contrast, proNGF caused a decrease in p75(NTR) levels. Functionally, NGF-induced increase in p75(NTR) caused a decrease in the sensitivity of TNBC cells to apoptosis induction. In contrast, knock-down of p75(NTR) using shRNA or small molecule inhibition of NGF-p75(NTR) interaction [using Ro 08-2750] sensitized TNBC cells to drug-induced apoptosis. In patient samples, the expression of NGF and NGFR [the p75(NTR) gene] mRNA are positively correlated in several subtypes of breast cancer, including basal-like breast cancer. Together these data suggest a positive feedback loop through which NGF-mediated upregulation of p75(NTR) can contribute to the chemo-resistance of TNBC cells.


Subject(s)
Nerve Growth Factor/pharmacology , Nerve Tissue Proteins/metabolism , Receptors, Nerve Growth Factor/metabolism , Triple Negative Breast Neoplasms/metabolism , Animals , Apoptosis/drug effects , Cell Line, Tumor , Drug Resistance, Neoplasm/drug effects , Humans , Mice , Triple Negative Breast Neoplasms/pathology
18.
Oncotarget ; 7(11): 12254-66, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26930721

ABSTRACT

Upregulation of SESTRIN 2 (SESN2) has been reported in response to diverse cellular stresses. In this study we demonstrate SESTRIN 2 induction following endoplasmic reticulum (ER) stress. ER stress-induced increases in SESTRIN 2 expression were dependent on both PERK and IRE1/XBP1 arms of the unfolded protein response (UPR). SESTRIN 2 induction, post ER stress, was responsible for mTORC1 inactivation and contributed to autophagy induction. Conversely, knockdown of SESTRIN 2 prolonged mTORC1 signaling, repressed autophagy and increased ER stress-induced cell death. Unexpectedly, the increase in ER stress-induced cell death was not linked to autophagy inhibition. Analysis of UPR pathways identified prolonged eIF2α, ATF4 and CHOP signaling in SESTRIN 2 knockdown cells following ER stress. SESTRIN 2 regulation enables UPR derived signals to indirectly control mTORC1 activity shutting down protein translation thus preventing further exacerbation of ER stress.


Subject(s)
Endoplasmic Reticulum Stress/physiology , Nuclear Proteins/biosynthesis , Cell Line, Tumor , Cell Survival/physiology , Endoplasmic Reticulum Stress/genetics , Endoribonucleases/metabolism , HCT116 Cells , Humans , MCF-7 Cells , Mechanistic Target of Rapamycin Complex 1/metabolism , Nuclear Proteins/genetics , Protein Serine-Threonine Kinases/metabolism , Up-Regulation , X-Box Binding Protein 1/metabolism , eIF-2 Kinase/metabolism
19.
Semin Cancer Biol ; 33: 57-66, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25814342

ABSTRACT

Cancer cells are exposed to intrinsic (oncogene) or extrinsic (microenvironmental) challenges, leading to activation of stress response pathways. The unfolded protein response (UPR) is the cellular response to endoplasmic reticulum (ER) stress and plays a pivotal role in tumor development. Depending on ER stress intensity and duration, the UPR is either pro-survival to preserve ER homeostasis or pro-death if the stress cannot be resolved. On one hand, the adaptive arm of the UPR is essential for cancer cells to survive the harsh conditions they are facing, and on the other hand, cancer cells have evolved mechanisms to bypass ER stress-induced cell death, thereby conferring them with a selective advantage for malignant transformation. Therefore, the mechanisms involved in the balance between survival and death outcomes of the UPR may be exploited as therapeutic tools to treat cancer.


Subject(s)
Apoptosis , Neoplasms/metabolism , Neoplasms/pathology , Unfolded Protein Response , Activating Transcription Factor 6/metabolism , Adenosine Triphosphate/chemistry , Animals , Cell Lineage , Cell Survival , Cell Transformation, Neoplastic , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum Stress , Endoribonucleases/metabolism , Homeostasis , Humans , Protein Serine-Threonine Kinases/metabolism , Signal Transduction
20.
FEBS Open Bio ; 4: 813-21, 2014.
Article in English | MEDLINE | ID: mdl-25349785

ABSTRACT

A mild heat shock (HS) preconditioning and acquisition of thermotolerance protects cells against a variety of cytotoxic agents that otherwise induce apoptosis. Here we tested whether there is a molecular link between HS preconditioning and endoplasmic reticulum (ER) stress-induced apoptosis. ER stress results from a loss of ER lumen homeostasis, culminating in an accumulation of unfolded/misfolded proteins in the ER and activation of unfolded protein response (UPR). Unresolved, ER stress leads to activation of BH3-only proteins, mitochondrial membrane permeabilization, caspase activation and apoptotic cell death. HS preconditioning (1 h at 42 °C) induced a rapid increase in HSPA1 (HSP70) levels which remained elevated for at least 48 h post-HS. HS preconditioning significantly reduced BAX, caspase activation and apoptosis in cell cultures treated with the ER stress-inducing agents thapsigargin (TG) and tunicamycin (TM). HS-mediated protection was found to be due to regulation of the BH3-only protein BIM. Further, overexpression of HSPA1 could not mimic the effect of HS on BIM expression, suggesting that other HS factors may play a role in inhibiting ER stress-induced apoptosis by regulating BIM.

SELECTION OF CITATIONS
SEARCH DETAIL
...