Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
1.
iScience ; 26(6): 106899, 2023 Jun 16.
Article in English | MEDLINE | ID: mdl-37305702

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) remains one of the human cancers with the poorest prognosis. Interestingly, we found that mitochondrial respiration in primary human PDAC cells depends mainly on the fatty acid oxidation (FAO) to meet basic energy requirements. Therefore, we treated PDAC cells with perhexiline, a well-recognized FAO inhibitor used in cardiac diseases. Some PDAC cells respond efficiently to perhexiline, which acts synergistically with chemotherapy (gemcitabine) in vitro and in two xenografts in vivo. Importantly, perhexiline in combination with gemcitabine induces complete tumor regression in one PDAC xenograft. Mechanistically, this co-treatment causes energy and oxidative stress promoting apoptosis but does not exert inhibition of FAO. Yet, our molecular analysis indicates that the carnitine palmitoyltransferase 1C (CPT1C) isoform is a key player in the response to perhexiline and that patients with high CPT1C expression have better prognosis. Our study reveals that repurposing perhexiline in combination with chemotherapy is a promising approach to treat PDAC.

2.
Trends Pharmacol Sci ; 43(12): 1001-1003, 2022 12.
Article in English | MEDLINE | ID: mdl-36049970

ABSTRACT

The antitumor activity of exercise by means of enhanced immune activation is documented, but better identification of the underlying mechanisms is required to develop new therapeutic strategies. Recent work from the Dr Bar-Sagi group reveals that exercise engages IL-15 signaling and pharmacological activation of the IL-15/IL-15R axis mimics the exercise-driven immune cell-mediated cytotoxicity in pancreatic cancer.


Subject(s)
Interleukin-15 , Signal Transduction , Humans
3.
Dig Liver Dis ; 54(12): 1605-1613, 2022 12.
Article in English | MEDLINE | ID: mdl-36089524

ABSTRACT

Pancreatic adenocarcinoma (PA) incidence is rising worldwide, especially in France. The evolution of known risk factors such as tobacco smoking, obesity, type 2 diabetes, chronic pancreatitis, or constitutional mutations is not sufficient to explain this trend. Pesticides are known risk factors in other malignancies. Previous studies have outlined pesticides' influence in PA, such as dichlorodiphenyltrichloroethane as plausible risk factors. The general population is directly or indirectly exposed to pesticides through air, food or water. Some of these chemicals may accumulate in the body all along lifetime and may harm carriers. The toxic mixing effects of these chemicals are not well documented. Several hypotheses have been put forward to explain how pesticides can induce indirect (fatty pancreas, induced diabetes) or direct (oxidative stress, cell damage) carcinogenesis in pancreatic cells through inflammation. A strong corpus exists acknowledging pesticides as a PA risk factor. However, published studies do not provide a sufficient level of evidence to prove causality and current prospective case-control studies are still ongoing.


Subject(s)
Adenocarcinoma , Diabetes Mellitus, Type 2 , Pancreatic Neoplasms , Pesticides , Humans , Pancreatic Neoplasms/chemically induced , Pancreatic Neoplasms/epidemiology , Pesticides/toxicity , Adenocarcinoma/chemically induced , Adenocarcinoma/epidemiology , Diabetes Mellitus, Type 2/etiology , Diabetes Mellitus, Type 2/complications , Pancreatic Neoplasms
4.
Trends Pharmacol Sci ; 43(9): 706-708, 2022 09.
Article in English | MEDLINE | ID: mdl-35382944

ABSTRACT

Targeting metabolic reprogramming has proven successful in oncology, but this field requires better identification of drugs that inhibit mitochondrial metabolism in cancer cells. Recent work from Dr Wolf's group reveals that the primary target of the antitumor compound SMIP004-7 is mitochondrial complex I (NDUFS2 subunit), inhibition of which promotes anticancer immune surveillance.


Subject(s)
Antineoplastic Agents , Neoplasms , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Electron Transport Complex I/metabolism , Humans , Mitochondria/metabolism , Neoplasms/pathology
5.
Cells ; 11(3)2022 01 26.
Article in English | MEDLINE | ID: mdl-35159234

ABSTRACT

Metabolic reprogramming is a feature of cancers for which recent research has been particularly active, providing numerous insights into the mechanisms involved. It occurs across the entire cancer process, from development to resistance to therapies. Established tumors exhibit dependencies for metabolic pathways, constituting vulnerabilities that can be targeted in the clinic. This knowledge is of particular importance for cancers that are refractory to any therapeutic approach, such as Pancreatic Ductal Adenocarcinoma (PDAC). One of the metabolic pathways dysregulated in PDAC is autophagy, a survival process that feeds the tumor with recycled intracellular components, through both cell-autonomous (in tumor cells) and nonautonomous (from the local and distant environment) mechanisms. Autophagy is elevated in established PDAC tumors, contributing to aberrant proliferation and growth even in a nutrient-poor context. Critical elements link autophagy to PDAC including genetic alterations, mitochondrial metabolism, the tumor microenvironment (TME), and the immune system. Moreover, high autophagic activity in PDAC is markedly related to resistance to current therapies. In this context, combining autophagy inhibition with standard chemotherapy, and/or drugs targeting other vulnerabilities such as metabolic pathways or the immune response, is an ongoing clinical strategy for which there is still much to do through translational and multidisciplinary research.


Subject(s)
Carcinoma, Pancreatic Ductal , Pancreatic Neoplasms , Autophagy/genetics , Carcinoma, Pancreatic Ductal/metabolism , Drug Resistance, Neoplasm , Humans , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Tumor Microenvironment , Pancreatic Neoplasms
6.
FEBS J ; 289(18): 5516-5526, 2022 09.
Article in English | MEDLINE | ID: mdl-34817127

ABSTRACT

The 4th International meeting Metabolism and Cancer initially programed to take place in Bordeaux (France) was held virtually on May 27-29, 2021. The three-day event was followed by around 600 participants daily from 47 countries around the world. The meeting hosted 21 speakers including selected talks and a keynote lecture from the Nobel Prize winner Sir Peter J. Ratcliffe (Oxford, UK). Presentations and discussions were divided in four scientific sessions: (a) Redox and energy metabolism (b) Redox and hypoxia (c) Metabolic profiling and epigenetic control and (d) Signalling, fuelling and metabolism in cancer and a general public session on cancer and nutrition. This report summarises the presentations and outcomes of the 4th annual Metabolism and Cancer symposium. We provide here a summary of the scientific highlights of this exciting meeting.


Subject(s)
Metabolism , Neoplasms , Humans , Neoplasms/metabolism , Societies, Medical
7.
EMBO Mol Med ; 13(7): e13502, 2021 07 07.
Article in English | MEDLINE | ID: mdl-34033220

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) patients frequently suffer from undetected micro-metastatic disease. This clinical situation would greatly benefit from additional investigation. Therefore, we set out to identify key signalling events that drive metastatic evolution from the pancreas. We searched for a gene signature that discriminate localised PDAC from confirmed metastatic PDAC and devised a preclinical protocol using circulating cell-free DNA (cfDNA) as an early biomarker of micro-metastatic disease to validate the identification of key signalling events. An unbiased approach identified, amongst actionable markers of disease progression, the PI3K pathway and a distinctive PI3Kα activation signature as predictive of PDAC aggressiveness and prognosis. Pharmacological or tumour-restricted genetic PI3Kα-selective inhibition prevented macro-metastatic evolution by hindering tumoural cell migratory behaviour independently of genetic alterations. We found that PI3Kα inhibition altered the quantity and the species composition of the produced lipid second messenger PIP3 , with a selective decrease of C36:2 PI-3,4,5-P3 . Tumoural PI3Kα inactivation prevented the accumulation of pro-tumoural CD206-positive macrophages in the tumour-adjacent tissue. Tumour cell-intrinsic PI3Kα promotes pro-metastatic features that could be pharmacologically targeted to delay macro-metastatic evolution.


Subject(s)
Adenocarcinoma , Carcinoma, Pancreatic Ductal , Pancreatic Neoplasms , Carcinoma, Pancreatic Ductal/genetics , Humans , Macrophages , Pancreatic Neoplasms/genetics , Phosphatidylinositol 3-Kinases/genetics
8.
Cell Death Dis ; 12(5): 460, 2021 05 08.
Article in English | MEDLINE | ID: mdl-33966044

ABSTRACT

TP53INP1 is a stress-induced protein, which acts as a dual positive regulator of transcription and of autophagy and whose deficiency has been linked with cancer and metabolic syndrome. Here, we addressed the unexplored role of TP53INP1 and of its Drosophila homolog dDOR in the maintenance of neuronal homeostasis under chronic stress, focusing on dopamine (DA) neurons under normal ageing- and Parkinson's disease (PD)-related context. Trp53inp1-/- mice displayed additional loss of DA neurons in the substantia nigra compared to wild-type (WT) mice, both with ageing and in a PD model based on targeted overexpression of α-synuclein. Nigral Trp53inp1 expression of WT mice was not significantly modified with ageing but was markedly increased in the PD model. Trp53inp2 expression showed similar evolution and did not differ between WT and Trp53inp1-/- mice. In Drosophila, pan-neuronal dDOR overexpression improved survival under paraquat exposure and mitigated the progressive locomotor decline and the loss of DA neurons caused by the human α-synuclein A30P variant. dDOR overexpression in DA neurons also rescued the locomotor deficit in flies with RNAi-induced downregulation of dPINK1 or dParkin. Live imaging, confocal and electron microscopy in fat bodies, neurons, and indirect flight muscles showed that dDOR acts as a positive regulator of basal autophagy and mitophagy independently of the PINK1-mediated pathway. Analyses in a mammalian cell model confirmed that modulating TP53INP1 levels does not impact mitochondrial stress-induced PINK1/Parkin-dependent mitophagy. These data provide the first evidence for a neuroprotective role of TP53INP1/dDOR and highlight its involvement in the regulation of autophagy and mitophagy in neurons.


Subject(s)
Carrier Proteins/metabolism , Heat-Shock Proteins/metabolism , Neuroprotection/genetics , Parkinson Disease/genetics , Stress, Physiological/genetics , Age Factors , Animals , Humans , Mice
9.
Int J Mol Sci ; 22(4)2021 Feb 03.
Article in English | MEDLINE | ID: mdl-33546421

ABSTRACT

Cell metabolism is reprogrammed in cancer cells to meet their high bioenergetics and biosynthetic demands. This metabolic reprogramming is accompanied by alterations in redox metabolism, characterized by accumulation of reactive oxygen species (ROS). Elevated production of ROS, mostly by mitochondrial respiration, is counteracted by higher production of antioxidant defenses (mainly glutathione and antioxidant enzymes). Cancer cells are adapted to a high concentration of ROS, which contributes to tumorigenesis, metastasis formation, resistance to therapy and relapse. Frequent genetic alterations observed in pancreatic ductal adenocarcinoma (PDAC) affect KRAS and p53 proteins, which have a role in ROS production and control, respectively. These observations led to the proposal of the use of antioxidants to prevent PDAC development and relapse. In this review, we focus on the therapeutic strategies to further increase ROS level to induce PDAC cell death. Combining the promotion of ROS production and inhibition of antioxidant capacity is a promising avenue for pancreatic cancer therapy in the clinic.


Subject(s)
Energy Metabolism , Oxidation-Reduction , Pancreatic Neoplasms/metabolism , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Antioxidants/metabolism , Biomarkers, Tumor , Clinical Studies as Topic , Disease Management , Disease Susceptibility , Drug Evaluation, Preclinical , Energy Metabolism/drug effects , Humans , Mitochondria/drug effects , Mitochondria/metabolism , Molecular Targeted Therapy , Oxidation-Reduction/drug effects , Oxidative Stress/drug effects , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/etiology , Pancreatic Neoplasms/pathology , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Treatment Outcome
10.
Cell Rep Med ; 1(8): 100143, 2020 11 17.
Article in English | MEDLINE | ID: mdl-33294863

ABSTRACT

Mitochondrial respiration (oxidative phosphorylation, OXPHOS) is an emerging target in currently refractory cancers such as pancreatic ductal adenocarcinoma (PDAC). However, the variability of energetic metabolic adaptations between PDAC patients has not been assessed in functional investigations. In this work, we demonstrate that OXPHOS rates are highly heterogeneous between patient tumors, and that high OXPHOS tumors are enriched in mitochondrial respiratory complex I at protein and mRNA levels. Therefore, we treated PDAC cells with phenformin (complex I inhibitor) in combination with standard chemotherapy (gemcitabine), showing that this treatment is synergistic specifically in high OXPHOS cells. Furthermore, phenformin cooperates with gemcitabine in high OXPHOS tumors in two orthotopic mouse models (xenografts and syngeneic allografts). In conclusion, this work proposes a strategy to identify PDAC patients likely to respond to the targeting of mitochondrial energetic metabolism in combination with chemotherapy, and that phenformin should be clinically tested in appropriate PDAC patient subpopulations.


Subject(s)
Cell Respiration/genetics , Drug Resistance, Neoplasm/genetics , Electron Transport Complex I/genetics , Pancreatic Neoplasms/genetics , Animals , Carcinoma, Pancreatic Ductal/drug therapy , Carcinoma, Pancreatic Ductal/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/genetics , Cell Respiration/drug effects , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Drug Resistance, Neoplasm/drug effects , Female , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Nude , Mitochondria/drug effects , Mitochondria/genetics , Oxidative Phosphorylation/drug effects , PC-3 Cells , Pancreatic Neoplasms/drug therapy , Phenformin/pharmacology , Xenograft Model Antitumor Assays/methods , Gemcitabine , Pancreatic Neoplasms
11.
Nat Commun ; 11(1): 4056, 2020 08 13.
Article in English | MEDLINE | ID: mdl-32792483

ABSTRACT

Autophagy has been associated with oncogenesis with one of its emerging key functions being its contribution to the metabolism of tumors. Therefore, deciphering the mechanisms of how autophagy supports tumor cell metabolism is essential. Here, we demonstrate that the inhibition of autophagy induces an accumulation of lipid droplets (LD) due to a decrease in fatty acid ß-oxidation, that leads to a reduction of oxidative phosphorylation (OxPHOS) in acute myeloid leukemia (AML), but not in normal cells. Thus, the autophagic process participates in lipid catabolism that supports OxPHOS in AML cells. Interestingly, the inhibition of OxPHOS leads to LD accumulation with the concomitant inhibition of autophagy. Mechanistically, we show that the disruption of mitochondria-endoplasmic reticulum (ER) contact sites (MERCs) phenocopies OxPHOS inhibition. Altogether, our data establish that mitochondria, through the regulation of MERCs, controls autophagy that, in turn finely tunes lipid degradation to fuel OxPHOS supporting proliferation and growth in leukemia.


Subject(s)
Autophagy/physiology , Endoplasmic Reticulum/genetics , Endoplasmic Reticulum/metabolism , Leukemia, Myeloid, Acute/metabolism , Leukemia/metabolism , Mitochondria/metabolism , Animals , Autophagy/genetics , Cell Line, Tumor , Cell Proliferation/genetics , Cell Proliferation/physiology , Flow Cytometry , Humans , Leukemia/genetics , Leukemia, Myeloid, Acute/pathology , Lipid Metabolism/genetics , Lipid Metabolism/physiology , Lipogenesis/genetics , Lipogenesis/physiology , Mice , Mitochondria/genetics , Oxidation-Reduction , Oxidative Phosphorylation
12.
Biomedicines ; 8(8)2020 Aug 03.
Article in English | MEDLINE | ID: mdl-32756381

ABSTRACT

Cancer cells reprogram their metabolism to meet bioenergetics and biosynthetic demands. The first observation of metabolic reprogramming in cancer cells was made a century ago ("Warburg effect" or aerobic glycolysis), leading to the classical view that cancer metabolism relies on a glycolytic phenotype. There is now accumulating evidence that most cancers also rely on mitochondria to satisfy their metabolic needs. Indeed, the current view of cancer metabolism places mitochondria as key actors in all facets of cancer progression. Importantly, mitochondrial metabolism has become a very promising target in cancer therapy, including for refractory cancers such as Pancreatic Ductal AdenoCarcinoma (PDAC). In particular, mitochondrial oxidative phosphorylation (OXPHOS) is an important target in cancer therapy. Other therapeutic strategies include the targeting of glutamine and fatty acids metabolism, as well as the inhibition of the TriCarboxylic Acid (TCA) cycle intermediates. A better knowledge of how pancreatic cancer cells regulate mitochondrial metabolism will allow the identification of metabolic vulnerabilities and thus novel and more efficient therapeutic options for the benefit of each patient.

14.
FASEB J ; 33(11): 12447-12463, 2019 11.
Article in English | MEDLINE | ID: mdl-31557059

ABSTRACT

The dismal prognosis of pancreatic ductal adenocarcinoma (PDAC) is mainly due to its rapidly acquired resistance to all conventional treatments. Despite drug-specific mechanisms of resistance, none explains how these cells resist the stress induced by any kind of anticancer treatment. Activation of stress-response pathways relies on the post-translational modifications (PTMs) of involved proteins. Among all PTMs, those mediated by the ubiquitin family of proteins play a central role. Our aim was to identify alterations of ubiquitination, neddylation, and sumoylation associated with the multiresistant phenotype and demonstrate their implications in the survival of PDAC cells undergoing treatment. This approach pointed at an alteration of promyelocytic leukemia (PML) protein sumoylation associated with both gemcitabine and oxaliplatin resistance. We could show that this alteration of PML sumoylation is part of a general mechanism of drug resistance, which in addition involves the abnormal activation of NF-κB and cAMP response element binding pathways. Importantly, using patient-derived tumors and cell lines, we identified a correlation between the levels of PML expression and sumoylation and the sensitivity of tumors to anticancer treatments.-Swayden, M., Alzeeb, G., Masoud, R., Berthois, Y., Audebert, S., Camoin, L., Hannouche, L., Vachon, H., Gayet, O., Bigonnet, M., Roques, J., Silvy, F., Carrier, A., Dusetti, N., Iovanna, J. L., Soubeyran, P. PML hyposumoylation is responsible for the resistance of pancreatic cancer.


Subject(s)
Drug Resistance, Neoplasm , Promyelocytic Leukemia Protein/metabolism , Second Messenger Systems , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cyclic AMP/genetics , Cyclic AMP/metabolism , HEK293 Cells , Humans , NF-kappa B/genetics , NF-kappa B/metabolism , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Promyelocytic Leukemia Protein/genetics , Sumoylation
15.
Physiol Rep ; 7(10): e14055, 2019 05.
Article in English | MEDLINE | ID: mdl-31124296

ABSTRACT

Tumor protein 53-induced nuclear protein 1 (TP53INP1) deficiency leads to oxidative stress-associated obesity and insulin resistance. Although skeletal muscle has a predominant role in the development of metabolic syndrome, the bioenergetics and functional consequences of TP53INP1 deficiency upon this tissue remain undocumented. To clarify this issue, gastrocnemius muscle mechanical performance, energy metabolism, and anatomy were investigated in TP53INP1-deficient and wild-type mice using a multidisciplinary approach implementing noninvasive multimodal-NMR techniques. TP53INP1 deficiency increased body adiposity but did not affect cytosolic oxidative stress, lipid content, and mitochondrial pool and capacity in myocyte. During a fatiguing bout of exercise, the in vivo oxidative ATP synthesis capacity was dramatically reduced in TP53INP1-deficient mice despite ADP level (the main in vivo stimulator of mitochondrial respiration) did not differ between both genotypes. Moreover, TP53INP1 deficiency did not alter fatigue resistance but paradoxically increased the contractile force, whereas there were no differences for muscle fiber-type distribution and calcium homeostasis between both genotypes. In addition, muscle proton efflux was decreased in TP53INP1-deficient mice, thereby indicating a reduced blood supply. In conclusion, TP53INP1 plays a role in muscle function and bioenergetics through oxidative capacity impairment possibly as the consequence of abnormal mitochondrial respiration regulation and/or defective blood supply.


Subject(s)
Energy Metabolism , Mitochondria, Muscle/metabolism , Muscle Contraction , Muscle, Skeletal/blood supply , Muscle, Skeletal/metabolism , Nuclear Proteins/deficiency , Oxidative Stress , Adenosine Triphosphate/metabolism , Adiposity , Animals , Genotype , Magnetic Resonance Imaging , Magnetic Resonance Spectroscopy , Male , Mice, Inbred C57BL , Mice, Knockout , Muscle Fatigue , Muscle Strength , Muscle, Skeletal/diagnostic imaging , Nuclear Proteins/genetics , Oxidation-Reduction , Phenotype , Regional Blood Flow
16.
Immunogenetics ; 71(3): 223-232, 2019 03.
Article in English | MEDLINE | ID: mdl-30225612

ABSTRACT

The lifespan of T cells is determined by continuous interactions of their T cell receptors (TCR) with self-peptide-MHC (self-pMHC) complexes presented by different subsets of antigen-presenting cells (APC). In the thymus, developing thymocytes are positively selected through recognition of self-pMHC presented by cortical thymic epithelial cells (cTEC). They are subsequently negatively selected by medullary thymic epithelial cells (mTEC) or thymic dendritic cells (DC) presenting self-pMHC complexes. In the periphery, the homeostasis of mature T cells is likewise controlled by the interaction of their TCR with self-pMHC complexes presented by lymph node stromal cells while they may be tolerized by DC presenting tissue-derived self-antigens. To perform these tasks, the different subsets of APC are equipped with distinct combination of antigen processing enzymes and consequently present specific repertoire of self-peptides. Here, we discuss one such antigen processing enzyme, the thymus-specific serine protease (TSSP), which is predominantly expressed by thymic stromal cells. In thymic DC and TEC, TSSP edits the repertoire of peptide presented by class II molecules and thus shapes the CD4 T cell repertoire.


Subject(s)
Antigen Presentation/immunology , CD4-Positive T-Lymphocytes/immunology , Epithelial Cells/immunology , Serine Proteases/immunology , Thymus Gland/immunology , Animals , CD4-Positive T-Lymphocytes/metabolism , Epithelial Cells/metabolism , Humans , Serine Proteases/metabolism , Thymus Gland/enzymology
17.
Proc Natl Acad Sci U S A ; 116(1): 211-216, 2019 01 02.
Article in English | MEDLINE | ID: mdl-30559202

ABSTRACT

Bone marrow (BM) produces all blood and immune cells deriving from hematopoietic stem cells (HSCs). The decrease of immune cell production during aging is one of the features of immunosenescence. The impact of redox dysregulation in BM aging is still poorly understood. Here we use TP53INP1-deficient (KO) mice endowed with chronic oxidative stress to assess the influence of aging-associated redox alterations in BM homeostasis. We show that TP53INP1 deletion has no impact on aging-related accumulation of HSCs. In contrast, the aging-related contraction of the lymphoid compartment is mitigated in TP53INP1 KO mice. B cells that accumulate in old KO BM are differentiating cells that can mature into functional B cells. Importantly, this phenotype results from B cell-intrinsic events associated with defective redox control. Finally, we show that oxidative stress in aged TP53INP1-deficient mice maintains STAT5 expression and activation in early B cells, driving high Pax5 expression, which provides a molecular mechanism for maintenance of B cell development upon aging.


Subject(s)
B-Lymphocytes/physiology , Bone Marrow/physiology , Lymphopoiesis , Nuclear Proteins/deficiency , Receptors, Interleukin-7/metabolism , STAT5 Transcription Factor/metabolism , Signal Transduction , Aging/physiology , Animals , B-Lymphocytes/metabolism , Bone Marrow/metabolism , Lymphopoiesis/physiology , Male , Mice , Mice, Inbred C57BL , Oxidation-Reduction , Oxidative Stress
18.
Sci Rep ; 8(1): 16999, 2018 11 19.
Article in English | MEDLINE | ID: mdl-30451898

ABSTRACT

It was already described that genetic inhibition of NUPR1 induces tumor growth arrest. In this paper we studied the metabolism changes after NUPR1 downregulation in pancreatic cancer cells, which results in a significant decrease of OXPHOS activity with a concomitant lower ATP production which precedes the necrotic cell death. We demonstrated that NUPR1 downregulation induces a mitochondrial failure with a loss of the mitochondrial membrane potential, a strong increase in ROS production and a concomitant relocalization of mitochondria to the vicinity of the endoplasmic reticulum (ER). In addition, the transcriptomic analysis of NUPR1-deficient cells shows a decrease in the expression of some ER stress response-associated genes. Indeed, in ER stressors-treated cells with thapsigargin, brefeldin A or tunicamycin, a greater increase in necrosis and decrease of ATP content was observed in NUPR1-defficent cells. Finally, in vivo experiments, using acute pancreatitis which induces ER stress as well as NUPR1 activation, we observed that NUPR1 expression protects acinar cells from necrosis in mice. Importantly, we also report that the cell death observed after knocking-down NUPR1 expression is completely reversed by incubation with Necrostatin-1, but not by inhibiting caspase activity with Z-VAD-FMK. Altogether, these data enable us to describe a model in which inactivation of NUPR1 in pancreatic cancer cells results in an ER stress that induces a mitochondrial malfunction, a deficient ATP production and, as consequence, the cell death mediated by a programmed necrosis.


Subject(s)
Cell Death , DNA-Binding Proteins/physiology , Endoplasmic Reticulum Stress , Mitochondria/pathology , Necrosis , Neoplasm Proteins/physiology , Pancreatic Neoplasms/pathology , Pancreatitis/pathology , Acinar Cells , Adenosine Triphosphate/metabolism , Animals , Apoptosis , Basic Helix-Loop-Helix Transcription Factors/antagonists & inhibitors , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , CRISPR-Cas Systems , Cells, Cultured , Humans , Mice , Mice, Knockout , Mitochondria/metabolism , Mitophagy , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Oxidative Phosphorylation , Pancreas/metabolism , Pancreas/pathology , Pancreatic Neoplasms/metabolism , Pancreatitis/metabolism
19.
Cancer Res ; 77(17): 4602-4612, 2017 09 01.
Article in English | MEDLINE | ID: mdl-28674078

ABSTRACT

Identifying critical factors involved in the metastatic progression of hepatocellular carcinoma (HCC) may offer important therapeutic opportunities. Here, we report that the proapoptotic stress response factor TP53INP1 is often selectively downregulated in advanced stage IV and metastatic human HCC tumors. Mechanistic investigations revealed that TP53INP1 downregulation in early-stage HCC cells promoted metastasis via DUSP10 phosphatase-mediated activation of the ERK pathway. The DUSP10 promoter included putative binding sites for p73 directly implicated in modulation by TP53INP1. Overall, our findings show how TP53INP1 plays a critical role in limiting the progression of early-stage HCC, with implications for developing new therapeutic strategies to attack metastatic HCC. Cancer Res; 77(17); 4602-12. ©2017 AACR.


Subject(s)
Carcinoma, Hepatocellular/pathology , Carrier Proteins/metabolism , Dual-Specificity Phosphatases/metabolism , Heat-Shock Proteins/metabolism , Lung Neoplasms/secondary , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Mitogen-Activated Protein Kinase Phosphatases/metabolism , Tumor Protein p73/metabolism , Animals , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Carrier Proteins/genetics , Dual-Specificity Phosphatases/genetics , Gene Expression Profiling , Heat-Shock Proteins/genetics , Humans , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Mice , Mice, Inbred BALB C , Mice, Nude , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase Phosphatases/genetics , Neoplasm Metastasis , Prognosis , Protein Array Analysis , Signal Transduction , Tumor Cells, Cultured , Tumor Protein p73/genetics , Xenograft Model Antitumor Assays
20.
Sci Rep ; 7: 45136, 2017 03 23.
Article in English | MEDLINE | ID: mdl-28332584

ABSTRACT

Metabolic reprogramming is a hallmark of cancer development, mediated by genetic and epigenetic alterations that may be pharmacologically targeted. Among oncogenes, the kinase Akt is commonly overexpressed in tumors and favors glycolysis, providing a rationale for using Akt inhibitors. Here, we addressed the question of whether and how inhibiting Akt activity could improve therapy of non-small cell lung cancer (NSCLC) that represents more than 80% of all lung cancer cases. First, we demonstrated that Akt inhibitors interacted synergistically with Microtubule-Targeting Agents (MTAs) and specifically in cancer cell lines, including those resistant to chemotherapy agents and anti-EGFR targeted therapies. In vivo, we further revealed that the chronic administration of low-doses of paclitaxel - i.e. metronomic scheduling - and the anti-Akt perifosine was the most efficient and the best tolerated treatment against NSCLC. Regarding drug mechanism of action, perifosine potentiated the pro-apoptotic effects of paclitaxel, independently of cell cycle arrest, and combining paclitaxel/perifosine resulted in a sustained suppression of glycolytic and mitochondrial metabolism. This study points out that targeting cancer cell bioenergetics may represent a novel therapeutic avenue in NSCLC, and provides a strong foundation for future clinical trials of metronomic MTAs combined with Akt inhibitors.


Subject(s)
Carcinoma, Non-Small-Cell Lung/metabolism , Energy Metabolism/drug effects , Lung Neoplasms/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/mortality , Carcinoma, Non-Small-Cell Lung/pathology , Cell Culture Techniques , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Disease Models, Animal , Glycolysis , Humans , Kaplan-Meier Estimate , Lung Neoplasms/drug therapy , Lung Neoplasms/mortality , Lung Neoplasms/pathology , Mice , Mitochondria/metabolism , Paclitaxel/pharmacology , Phosphorylcholine/analogs & derivatives , Phosphorylcholine/pharmacology , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...