Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
2.
J Cell Mol Med ; 27(21): 3363-3377, 2023 11.
Article in English | MEDLINE | ID: mdl-37753803

ABSTRACT

Activating point mutations of the RAS gene act as driver mutations for a subset of precursor-B cell acute lymphoblastic leukaemias (pre-B ALL) and represent an ambitious target for therapeutic approaches. The X box-binding protein 1 (XBP1), a key regulator of the unfolded protein response (UPR), is critical for pre-B ALL cell survival, and high expression of XBP1 confers poor prognosis in ALL patients. However, the mechanism of XBP1 activation has not yet been elucidated in RAS mutated pre-B ALL. Here, we demonstrate that XBP1 acts as a downstream linchpin of the IL-7 receptor signalling pathway and that pharmacological inhibition or genetic ablation of XBP1 selectively abrogates IL-7 receptor signalling via inhibition of its downstream effectors, JAK1 and STAT5. We show that XBP1 supports malignant cell growth of pre-B NRASG12D ALL cells and that genetic loss of XBP1 consequently leads to cell cycle arrest and apoptosis. Our findings reveal that active XBP1 prevents the cytotoxic effects of a dual PI3K/mTOR pathway inhibitor (BEZ235) in pre-B NRASG12D ALL cells. This implies targeting XBP1 in combination with BEZ235 as a promising new targeted strategy against the oncogenic RAS in NRASG12D -mutated pre-B ALL.


Subject(s)
Precursor B-Cell Lymphoblastic Leukemia-Lymphoma , Precursor Cell Lymphoblastic Leukemia-Lymphoma , Humans , Genes, ras , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Signal Transduction , Unfolded Protein Response/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Membrane Proteins/genetics , GTP Phosphohydrolases/genetics , X-Box Binding Protein 1/genetics
6.
Carcinogenesis ; 42(2): 272-284, 2021 02 25.
Article in English | MEDLINE | ID: mdl-32915195

ABSTRACT

BCR-ABL1-positive acute lymphoblastic leukemia (ALL) cell survival is dependent on the inositol-requiring enzyme 1 alpha (IRE1α) branch of the unfolded protein response. In the current study, we have focused on exploring the efficacy of a simultaneous pharmacological inhibition of BCR-ABL1 and IRE1α in Philadelphia-positive (Ph+) ALL using tyrosine kinase inhibitor (TKI) nilotinib and the IRE1α inhibitor MKC-8866. The combination of 0.5 µM nilotinib and 30 µM MKC-8866 in Ph+ ALL cell lines led to a synergistic effect on cell viability. To mimic this dual inhibition on a genetic level, pre-B-cells from conditional Xbp1+/fl mice were transduced with a BCR-ABL1 construct and with either tamoxifen-inducible cre or empty vector. Cells showed a significant sensitization to the effect of TKIs after the induction of the heterozygous deletion. Finally, we performed a phosphoproteomic analysis on Ph+ ALL cell lines treated with the combination of nilotinib and MKC-8866 to identify potential targets involved in their synergistic effect. An enhanced activation of p38 mitogen-activated protein kinase α (p38α MAPK) was identified. In line with this findings, p38 MAPK and, another important endoplasmic reticulum-stress-related kinase, c-Jun N-terminal kinase (JNK) were found to mediate the potentiated cytotoxic effect induced by the combination of MKC-8866 and nilotinib since the targeting of p38 MAPK with its specific inhibitor BIRB-796 or JNK with JNK-in-8 hindered the synergistic effect observed upon treatment with nilotinib and MKC-8866. In conclusion, the identified combined action of nilotinib and MKC-8866 might represent a successful therapeutic strategy in high-risk Ph+ ALL.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Endoribonucleases/antagonists & inhibitors , Fusion Proteins, bcr-abl/antagonists & inhibitors , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Protein Serine-Threonine Kinases/antagonists & inhibitors , Synthetic Lethal Mutations/drug effects , Animals , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Benzamides/pharmacology , Benzopyrans/pharmacology , Benzopyrans/therapeutic use , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/genetics , Disease Models, Animal , Drug Synergism , Fusion Proteins, bcr-abl/genetics , Humans , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , JNK Mitogen-Activated Protein Kinases/metabolism , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/genetics , Mice, Transgenic , Morpholines/pharmacology , Morpholines/therapeutic use , Naphthalenes/pharmacology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Primary Cell Culture , Pyrazoles/pharmacology , Pyridines/pharmacology , Pyrimidines/pharmacology , Pyrimidines/therapeutic use , X-Box Binding Protein 1/genetics , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/metabolism
7.
Cells ; 9(11)2020 10 22.
Article in English | MEDLINE | ID: mdl-33105603

ABSTRACT

Glioblastoma (GBM) is the most prevalent malignant primary brain tumor with a very poor survival rate. Temozolomide (TMZ) is the common chemotherapeutic agent used for GBM treatment. We recently demonstrated that simvastatin (Simva) increases TMZ-induced apoptosis via the inhibition of autophagic flux in GBM cells. Considering the role of the unfolded protein response (UPR) pathway in the regulation of autophagy, we investigated the involvement of UPR in Simva-TMZ-induced cell death by utilizing highly selective IRE1 RNase activity inhibitor MKC8866, PERK inhibitor GSK-2606414 (PERKi), and eIF2α inhibitor salubrinal. Simva-TMZ treatment decreased the viability of GBM cells and significantly increased apoptotic cell death when compared to TMZ or Simva alone. Simva-TMZ induced both UPR, as determined by an increase in GRP78, XBP splicing, eukaryote initiation factor 2α (eIF2α) phosphorylation, and inhibited autophagic flux (accumulation of LC3ß-II and inhibition of p62 degradation). IRE1 RNase inhibition did not affect Simva-TMZ-induced cell death, but it significantly induced p62 degradation and increased the microtubule-associated proteins light chain 3 (LC3)ß-II/LC3ß-I ratio in U87 cells, while salubrinal did not affect the Simva-TMZ induced cytotoxicity of GBM cells. In contrast, protein kinase RNA-like endoplasmic reticulum kinase (PERK) inhibition significantly increased Simva-TMZ-induced cell death in U87 cells. Interestingly, whereas PERK inhibition induced p62 accumulation in both GBM cell lines, it differentially affected the LC3ß-II/LC3ß-I ratio in U87 (decrease) and U251 (increase) cells. Simvastatin sensitizes GBM cells to TMZ-induced cell death via a mechanism that involves autophagy and UPR pathways. More specifically, our results imply that the IRE1 and PERK signaling arms of the UPR regulate Simva-TMZ-mediated autophagy flux inhibition in U251 and U87 GBM cells.


Subject(s)
Antineoplastic Agents/pharmacology , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Simvastatin/pharmacology , Temozolomide/pharmacology , Unfolded Protein Response/drug effects , Apoptosis/drug effects , Brain Neoplasms , Caspases/metabolism , Cell Death/drug effects , Cell Line, Tumor , Cell Survival , Drug Synergism , Endoplasmic Reticulum Chaperone BiP , Endoplasmic Reticulum Stress/drug effects , Eukaryotic Initiation Factor-2/metabolism , Glioblastoma , Humans , Phosphorylation , Signal Transduction/drug effects , Temozolomide/therapeutic use
8.
Cancer Lett ; 494: 73-83, 2020 12 01.
Article in English | MEDLINE | ID: mdl-32882336

ABSTRACT

Glioblastoma multiforme (GBM) is the most severe primary brain cancer. Despite an aggressive treatment comprising surgical resection and radio/chemotherapy, patient's survival post diagnosis remains short. A limitation for success in finding novel improved therapeutic options for such dismal disease partly lies in the lack of a relevant animal model that accurately recapitulates patient disease and standard of care. In the present study, we have developed an immunocompetent GBM model that includes tumor surgery and a radio/chemotherapy regimen resembling the Stupp protocol and we have used this model to test the impact of the pharmacological inhibition of the endoplasmic reticulum (ER) stress sensor IRE1, on treatment efficacy.


Subject(s)
Benzopyrans/administration & dosage , Brain Neoplasms/therapy , Combined Modality Therapy/methods , Glioblastoma/therapy , Morpholines/administration & dosage , Animals , Benzopyrans/pharmacology , Brain Neoplasms/genetics , Brain Neoplasms/immunology , Cell Line, Tumor , Craniotomy , Drug Therapy , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Glioblastoma/genetics , Glioblastoma/immunology , Humans , Immunocompetence , Injections, Intralesional , Mice , Morpholines/pharmacology , Neoadjuvant Therapy , Radiotherapy , Treatment Outcome , Tumor Microenvironment/drug effects , Xenograft Model Antitumor Assays
9.
Cancer Lett ; 490: 76-88, 2020 10 10.
Article in English | MEDLINE | ID: mdl-32679165

ABSTRACT

Rhabdomyosarcoma (RMS), the most common soft-tissue sarcoma, is associated with a low 5-year survival and harsh treatment side effects, underscoring an urgent need for therapy. The unfolded protein response (UPR) is activated in response to endoplasmic reticulum (ER) stress, where three ER stress receptors, IRE1, PERK and ATF6, aim to restore cellular homeostasis. The UPR is pro-tumourigenic in many cancers. In this study, we investigate basal UPR activity in RMS. Basal activation of IRE1 and PERK was observed in RMS cell lines, which was diminished upon addition of the IRE1 RNase inhibitor, MKC8866, or PERK inhibitor, AMGEN44. UPR inhibition caused a reduction in cell viability, cell proliferation and inhibition of long-term colony formation in both subtypes of RMS. Alveolar RMS (ARMS) subtype was highly sensitive to IRE1 inhibition, whereas embryonal RMS (ERMS) subtypes responded more markedly to PERK inhibition. Further investigation revealed a robust activation of senescence upon UPR inhibition. For the first time, the UPR is implicated in RMS biology and phenotype, and inhibition of UPR signalling reduces cell growth, suggesting that the UPR may be a promising target in RMS.


Subject(s)
Endoribonucleases/metabolism , Protein Serine-Threonine Kinases/metabolism , Rhabdomyosarcoma/pathology , Unfolded Protein Response/physiology , eIF-2 Kinase/metabolism , Cell Line, Tumor , Cell Survival , Humans , Rhabdomyosarcoma/metabolism
10.
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.

11.
Nat Chem Biol ; 15(11): 1129, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31439938

ABSTRACT

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

12.
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
13.
Nat Chem Biol ; 15(8): 764-775, 2019 08.
Article in English | MEDLINE | ID: mdl-31320759

ABSTRACT

Accumulation of unfolded proteins at the endoplasmic reticulum (ER) is a salient attribute of many human diseases including obesity, liver disorders, cancer, diabetes and neurodegeneration. To restore ER proteostasis, cells activate the unfolded protein response (UPR), a signaling pathway that imposes adaptive programs or triggers apoptosis of damaged cells. The UPR is critical to sustain the normal function of specialized secretory cells (i.e., pancreatic ß cells and B lymphocytes) and to control the production of lipids and cholesterol in the liver. In the context of disease, adaptive UPR responses have been linked to the growth of solid tumors, whereas chronic ER stress contributes to cell dysfunction in brain diseases, metabolic syndromes, among other conditions. Here we discuss recent developments in the design and optimization of novel compounds to manipulate UPR signaling and their efficacy in various disease models.


Subject(s)
Drug Delivery Systems , Signal Transduction , Unfolded Protein Response , Animals , Endoplasmic Reticulum Stress/drug effects , Endoplasmic Reticulum Stress/physiology , Humans , Neoplasms/drug therapy , Neoplasms/metabolism
14.
Nat Commun ; 10(1): 323, 2019 01 24.
Article in English | MEDLINE | ID: mdl-30679434

ABSTRACT

Activation of endoplasmic reticulum (ER) stress/the unfolded protein response (UPR) has been linked to cancer, but the molecular mechanisms are poorly understood and there is a paucity of reagents to translate this for cancer therapy. Here, we report that an IRE1α RNase-specific inhibitor, MKC8866, strongly inhibits prostate cancer (PCa) tumor growth as monotherapy in multiple preclinical models in mice and shows synergistic antitumor effects with current PCa drugs. Interestingly, global transcriptomic analysis reveal that IRE1α-XBP1s pathway activity is required for c-MYC signaling, one of the most highly activated oncogenic pathways in PCa. XBP1s is necessary for optimal c-MYC mRNA and protein expression, establishing, for the first time, a direct link between UPR and oncogene activation. In addition, an XBP1-specific gene expression signature is strongly associated with PCa prognosis. Our data establish IRE1α-XBP1s signaling as a central pathway in PCa and indicate that its targeting may offer novel treatment strategies.


Subject(s)
Antineoplastic Agents/pharmacology , Benzopyrans/pharmacology , Endoribonucleases/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Morpholines/pharmacology , Prostatic Neoplasms/metabolism , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Signal Transduction/physiology , X-Box Binding Protein 1/metabolism , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/therapeutic use , Benzopyrans/chemistry , Benzopyrans/therapeutic use , Cell Line, Tumor , Cell Survival , Endoribonucleases/antagonists & inhibitors , Endoribonucleases/genetics , Humans , Male , Mice , Mice, Nude , Morpholines/chemistry , Morpholines/therapeutic use , Prostatic Neoplasms/drug therapy , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/therapeutic use , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins c-myc/genetics , Random Allocation
15.
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
16.
Cell Metab ; 27(4): 828-842.e7, 2018 04 03.
Article in English | MEDLINE | ID: mdl-29551590

ABSTRACT

Dietary restriction (DR) was shown to impact on tumor growth with very variable effects depending on the cancer type. However, how DR limits cancer progression remains largely unknown. Here, we demonstrate that feeding mice a low-protein (Low PROT) isocaloric diet but not a low-carbohydrate (Low CHO) diet reduced tumor growth in three independent mouse cancer models. Surprisingly, this effect relies on anticancer immunosurveillance, as depleting CD8+ T cells, antigen-presenting cells (APCs), or using immunodeficient mice prevented the beneficial effect of the diet. Mechanistically, we established that a Low PROT diet induces the unfolded protein response (UPR) in tumor cells through the activation of IRE1α and RIG1 signaling, thereby resulting in cytokine production and mounting an efficient anticancer immune response. Collectively, our data suggest that a Low PROT diet induces an IRE1α-dependent UPR in cancer cells, enhancing a CD8-mediated T cell response against tumors.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Diet, Protein-Restricted , Endoribonucleases/metabolism , Immunologic Surveillance , Neoplasms, Experimental/diet therapy , Neoplasms, Experimental/immunology , Protein Serine-Threonine Kinases/metabolism , Unfolded Protein Response/immunology , Animals , Antigen-Presenting Cells/immunology , Cell Line, Tumor , Colorectal Neoplasms/diet therapy , Colorectal Neoplasms/immunology , Endoribonucleases/genetics , Female , Lymphocyte Depletion , Lymphoma/diet therapy , Lymphoma/immunology , Melanoma, Experimental/diet therapy , Melanoma, Experimental/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Protein Serine-Threonine Kinases/genetics , RNA Helicases/metabolism , Signal Transduction
17.
Oncotarget ; 9(3): 2984-3000, 2018 Jan 09.
Article in English | MEDLINE | ID: mdl-29423023

ABSTRACT

The intensity and duration of endoplasmic reticulum (ER) stress converts the unfolded protein response (UPR) from an adaptive into a terminal response. The first regulates homeostasis, the latter triggers apoptosis. Cells that rapidly proliferate and possess developed secretory capabilities, such as leukemia cells, depend on an efficiently operating UPR to maintain proteostasis. Activation of terminal UPR by either blockade of adaptive UPR or exaggeration of ER stress has been explored as a novel approach in cancer therapy. For mast cell leukemia (MCL) the efficacy of both approaches, by utilizing the KITV560G,D816V-positive MCL cell line HMC-1.2, was investigated. We show that HMC-1.2 cells display a tonic activation of the IRE1α arm of the UPR, which constitutively generates spliced XBP1. Inhibition of IRE1α by different types of inhibitors (MKC-8866, STF-083010, and KIRA6) suppressed proliferation at concentrations needed for blockade of IRE1α-mediated XBP1 splicing. At higher concentrations, these inhibitors triggered an apoptotic response. Blocking the proteasome by bortezomib, which confers an exaggerated UPR, resulted in a marked cytotoxic response. Bortezomib treatment also caused activation of the kinase JNK, which played a pro-proliferative and anti-apoptotic role. Hence, the combination of bortezomib with a JNK inhibitor synergized to induce cell death. In summary, the UPR can be addressed as an effective therapeutic target against KITD816V-positive MCL.

18.
J Clin Invest ; 128(4): 1283-1299, 2018 04 02.
Article in English | MEDLINE | ID: mdl-29480818

ABSTRACT

The unfolded protein response (UPR) is a cellular homeostatic mechanism that is activated in many human cancers and plays pivotal roles in tumor progression and therapy resistance. However, the molecular mechanisms for UPR activation and regulation in cancer cells remain elusive. Here, we show that oncogenic MYC regulates the inositol-requiring enzyme 1 (IRE1)/X-box binding protein 1 (XBP1) branch of the UPR in breast cancer via multiple mechanisms. We found that MYC directly controls IRE1 transcription by binding to its promoter and enhancer. Furthermore, MYC forms a transcriptional complex with XBP1, a target of IRE1, and enhances its transcriptional activity. Importantly, we demonstrate that XBP1 is a synthetic lethal partner of MYC. Silencing of XBP1 selectively blocked the growth of MYC-hyperactivated cells. Pharmacological inhibition of IRE1 RNase activity with small molecule inhibitor 8866 selectively restrained the MYC-overexpressing tumor growth in vivo in a cohort of preclinical patient-derived xenograft models and genetically engineered mouse models. Strikingly, 8866 substantially enhanced the efficacy of docetaxel chemotherapy, resulting in rapid regression of MYC-overexpressing tumors. Collectively, these data establish the synthetic lethal interaction of the IRE1/XBP1 pathway with MYC hyperactivation and provide a potential therapy for MYC-driven human breast cancers.


Subject(s)
Breast Neoplasms/drug therapy , Docetaxel/pharmacology , Drug Delivery Systems , Endoribonucleases/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Response Elements , Signal Transduction/drug effects , X-Box Binding Protein 1/metabolism , Animals , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Endoribonucleases/genetics , Female , Humans , Mice , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins c-myc/genetics , Saccharomyces cerevisiae , Signal Transduction/genetics , Unfolded Protein Response/drug effects , Unfolded Protein Response/genetics , X-Box Binding Protein 1/genetics , Xenograft Model Antitumor Assays
19.
EMBO Mol Med ; 10(3)2018 03.
Article in English | MEDLINE | ID: mdl-29311133

ABSTRACT

Proteostasis imbalance is emerging as a major hallmark of cancer, driving tumor aggressiveness. Evidence suggests that the endoplasmic reticulum (ER), a major site for protein folding and quality control, plays a critical role in cancer development. This concept is valid in glioblastoma multiform (GBM), the most lethal primary brain cancer with no effective treatment. We previously demonstrated that the ER stress sensor IRE1α (referred to as IRE1) contributes to GBM progression, through XBP1 mRNA splicing and regulated IRE1-dependent decay (RIDD) of RNA Here, we first demonstrated IRE1 signaling significance to human GBM and defined specific IRE1-dependent gene expression signatures that were confronted to human GBM transcriptomes. This approach allowed us to demonstrate the antagonistic roles of XBP1 mRNA splicing and RIDD on tumor outcomes, mainly through selective remodeling of the tumor stroma. This study provides the first demonstration of a dual role of IRE1 downstream signaling in cancer and opens a new therapeutic window to abrogate tumor progression.


Subject(s)
Brain Neoplasms/enzymology , Brain Neoplasms/pathology , Carcinogenesis/pathology , Endoribonucleases/metabolism , Glioblastoma/enzymology , Glioblastoma/pathology , Protein Serine-Threonine Kinases/metabolism , Brain Neoplasms/genetics , Carcinogenesis/genetics , Cell Line, Tumor , Endoribonucleases/genetics , Gene Expression Regulation, Neoplastic , Glioblastoma/genetics , Humans , Models, Biological , Mutation/genetics , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Phenotype , Protein Serine-Threonine Kinases/genetics , RNA Splicing/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction , Tumor Microenvironment/genetics
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...