Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
1.
Cancer Res ; 83(15): 2461-2470, 2023 08 01.
Article in English | MEDLINE | ID: mdl-37272750

ABSTRACT

Dependency on mitochondrial oxidative phosphorylation (OxPhos) is a potential weakness for leukemic stem cells (LSC) that can be exploited for therapeutic purposes. Fatty acid oxidation (FAO) is a crucial OxPhos-fueling catabolic pathway for some acute myeloid leukemia (AML) cells, particularly chemotherapy-resistant AML cells. Here, we identified cold sensitivity at 4°C (cold killing challenge; CKC4), commonly used for sample storage, as a novel vulnerability that selectively kills AML LSCs with active FAO-supported OxPhos while sparing normal hematopoietic stem cells. Cell death of OxPhos-positive leukemic cells was induced by membrane permeabilization at 4°C; by sharp contrast, leukemic cells relying on glycolysis were resistant. Forcing glycolytic cells to activate OxPhos metabolism sensitized them to CKC4. Lipidomic and proteomic analyses showed that OxPhos shapes the composition of the plasma membrane and introduces variation of 22 lipid subfamilies between cold-sensitive and cold-resistant cells. Together, these findings indicate that steady-state energy metabolism at body temperature predetermines the sensitivity of AML LSCs to cold temperature, suggesting that cold sensitivity could be a potential OxPhos biomarker. These results could have important implications for designing experiments for AML research to avoid cell storage at 4°C. SIGNIFICANCE: Mitochondrial metabolism fueled by FAO alters the membrane composition and introduces membrane fragility upon cold exposure in OxPhos-driven AML and in LSCs. See related commentary by Jones, p. 2441.


Subject(s)
Leukemia, Myeloid, Acute , Oxidative Phosphorylation , Humans , Cold Temperature , Proteomics , Leukemia, Myeloid, Acute/drug therapy , Hematopoietic Stem Cells/metabolism , Fatty Acids/metabolism , Neoplastic Stem Cells/metabolism
2.
Oncoimmunology ; 11(1): 2116844, 2022.
Article in English | MEDLINE | ID: mdl-36046811

ABSTRACT

IRE1α is one of the three ER transmembrane transducers of the Unfolded Protein Response (UPR) activated under endoplasmic reticulum (ER) stress. IRE1α activation has a dual role in cancer as it may be either pro- or anti-tumoral depending on the studied models. Here, we describe the discovery that exogenous expression of IRE1α, resulting in IRE1α auto-activation, did not affect cancer cell proliferation in vitro but resulted in a tumor-suppressive phenotype in syngeneic immunocompetent mice. We found that exogenous expression of IRE1α in murine colorectal and Lewis lung carcinoma cells impaired tumor growth when syngeneic tumor cells were subcutaneously implanted in immunocompetent mice but not in immunodeficient mice. Mechanistically, the in vivo tumor-suppressive effect of overexpressing IRE1α in tumor cells was associated with IRE1α RNAse activity driving both XBP1 mRNA splicing and regulated IRE1-dependent decay of RNA (RIDD). We showed that the tumor-suppressive phenotype upon IRE1α overexpression was characterized by the induction of apoptosis in tumor cells along with an enhanced adaptive anti-cancer immunosurveillance. Hence, our work indicates that IRE1α overexpression and/or activation in tumor cells can limit tumor growth in immunocompetent mice. This finding might point toward the need of adjusting the use of IRE1α inhibitors in cancer treatments based on the predominant outcome of the RNAse activity of IRE1α.


Subject(s)
Endoribonucleases , Neoplasms , Animals , Endoribonucleases/genetics , Endoribonucleases/metabolism , Immunity , Mice , Neoplastic Processes , Protein Serine-Threonine Kinases/genetics , Signal Transduction , X-Box Binding Protein 1/genetics , X-Box Binding Protein 1/metabolism
3.
Sci Adv ; 8(27): eabn6491, 2022 07 08.
Article in English | MEDLINE | ID: mdl-35857457

ABSTRACT

Depletion of circulating asparagine with l-asparaginase (ASNase) is a mainstay of leukemia treatment and is under investigation in many cancers. Expression levels of asparagine synthetase (ASNS), which catalyzes asparagine synthesis, were considered predictive of cancer cell sensitivity to ASNase treatment, a notion recently challenged. Using [U-13C5]-l-glutamine in vitro and in vivo in a mouse model of B cell lymphomas (BCLs), we demonstrated that supraphysiological or physiological concentrations of asparagine prevent de novo asparagine biosynthesis, regardless of ASNS expression levels. Overexpressing ASNS in ASNase-sensitive BCL was insufficient to confer resistance to ASNase treatment in vivo. Moreover, we showed that ASNase's glutaminase activity enables its maximal anticancer effect. Together, our results indicate that baseline ASNS expression (low or high) cannot dictate BCL dependence on de novo asparagine biosynthesis and predict BCL sensitivity to dual ASNase activity. Thus, except for ASNS-deficient cancer cells, ASNase's glutaminase activity should be considered in the clinic.


Subject(s)
Antineoplastic Agents , Aspartate-Ammonia Ligase , Lymphoma, B-Cell , Precursor Cell Lymphoblastic Leukemia-Lymphoma , Animals , Antineoplastic Agents/therapeutic use , Asparaginase/therapeutic use , Asparagine/metabolism , Aspartate-Ammonia Ligase/genetics , Aspartate-Ammonia Ligase/metabolism , Cell Line, Tumor , Glutaminase/therapeutic use , Lymphoma, B-Cell/drug therapy , Mice , Tumor Microenvironment
4.
Pharmacol Res Perspect ; 9(5): e00854, 2021 10.
Article in English | MEDLINE | ID: mdl-34478236

ABSTRACT

Targeting the first protein complex of the mitochondrial electron transport chain (MC1) in cancer has become an attractive therapeutic approach in the recent years, given the metabolic vulnerabilities of cancer cells. The anticancer effect exerted by the pleiotropic drug metformin and the associated reduction in hypoxia-inducible factor 1α (HIF-1α) levels putatively mediated by MC1 inhibition led to the development of HIF-1α inhibitors, such as BAY87-2243, with a more specific MC1 targeting. However, the development of BAY87-2243 was stopped early in phase 1 due to dose-independent emesis and thus there is still no clinical proof of concept for the approach. Given the importance of mitochondrial metabolism during cancer progression, there is still a strong therapeutic need to develop specific and safe MC1 inhibitors. We recently reported the synthesis of compounds with a novel chemotype and potent action on HIF-1α degradation and MC1 inhibition. We describe here the selectivity, safety profile and anti-cancer activity in solid tumors of lead compound EVT-701. In addition, using murine models of lung cancer and of Non-Hodgkin's B cell lymphoma we demonstrated that EVT-701 reduced tumor growth and lymph node invasion when used as a single agent therapy. LKB1 deficiency in lung cancer was identified as a potential indicator of accrued sensitivity to EVT-701, allowing stratification and selection of patients in clinical trials. Altogether these results support further evaluation of EVT-701 alone or in combination in preclinical models and eventually in patients.


Subject(s)
Apoptosis/drug effects , Carcinoma, Lewis Lung/metabolism , Cell Proliferation/drug effects , Electron Transport Complex I/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Lung Neoplasms/metabolism , Lymph Nodes/drug effects , Lymphoma, B-Cell/metabolism , Mitochondria/drug effects , AMP-Activated Protein Kinases/genetics , Animals , Carcinoma, Lewis Lung/pathology , Cell Line, Tumor , Cell Respiration , In Vitro Techniques , Lung Neoplasms/pathology , Lymph Nodes/pathology , Lymphoma, B-Cell/pathology , Mice , Mitochondria/metabolism , Neoplasm Invasiveness , Neoplasm Transplantation
5.
Mol Metab ; 53: 101294, 2021 11.
Article in English | MEDLINE | ID: mdl-34256164

ABSTRACT

BACKGROUND: There is growing interest in the analysis of tumor metabolism to identify cancer-specific metabolic vulnerabilities and therapeutic targets. Finding of such candidate metabolic pathways mainly relies on the highly sensitive identification and quantitation of numerous metabolites and metabolic fluxes using metabolomics and isotope tracing analyses. However, nutritional requirements and metabolic routes used by cancer cells cultivated in vitro do not always reflect the metabolic demands of malignant cells within the tumor milieu. Therefore, to understand how the metabolism of tumor cells in its physiological environment differs from that of normal cells, these analyses must be performed in vivo. SCOPE OF REVIEW: This review covers the physiological impact of the exogenous administration of a stable isotope tracer into cancer animal models. We discuss specific aspects of in vivo isotope tracing protocols based on discrete bolus injections of a labeled metabolite: the tracer administration per se and the fasting period prior to it. In addition, we illustrate the complex physiological scenarios that arise when studying tumor metabolism - by isotopic labeling in animal models fed with a specific amino acid restricted diet. Finally, we provide strategies to minimize these limitations. MAJOR CONCLUSIONS: There is growing evidence that metabolic dependencies in cancers are influenced by tissue environment, cancer lineage, and genetic events. An increasing number of studies describe discrepancies in tumor metabolic dependencies when studied in in vitro settings or in vivo models, including cancer patients. Therefore, in-depth in vivo profiling of tumor metabolic routes within the appropriate pathophysiological environment will be key to identify relevant alterations that contribute to cancer onset and progression.


Subject(s)
Isotope Labeling , Neoplasms/metabolism , Animals , Humans
6.
FEBS J ; 288(11): 3547-3569, 2021 06.
Article in English | MEDLINE | ID: mdl-33340237

ABSTRACT

Myocardial ischemia/reperfusion (I/R) injury is a frequent perioperative threat, with numerous strategies developed to limit and/or prevent it. One interesting axis of research is the anesthetic preconditioning (APc) agent's hypothesis (such as sevoflurane, SEV). However, APc's mode of action is still poorly understood and volatile anesthetics used as preconditioning agents are often not well suited in clinical practice. Here, in vitro using H9C2 cells lines (in myeloblast state or differentiated toward cardiomyocytes) and in vivo in mice, we identified that SEV-induced APc is mediated by a mild induction of reactive oxygen species (ROS) that activates Akt and induces the expression of the anti-apoptotic protein B-cell lymphoma-extra large (Bcl-xL), therefore protecting cardiomyocytes from I/R-induced death. Furthermore, we extended these results to human cardiomyocytes (derived from induced pluripotent stem - IPS - cells). Importantly, we demonstrated that this protective signaling pathway induced by SEV could be stimulated using the antidiabetic agent metformin (MET), suggesting the preconditioning properties of MET. Altogether, our study identified a signaling pathway allowing APc of cardiac injuries as well as a rational for the use of MET as a pharmacological preconditioning agent to prevent I/R injuries.


Subject(s)
Apoptosis/drug effects , Myocardial Reperfusion Injury/drug therapy , Reperfusion Injury/drug therapy , bcl-X Protein/genetics , Animals , Cell Survival/drug effects , Humans , Hypoglycemic Agents/pharmacology , Metformin/pharmacology , Myocardial Reperfusion Injury/genetics , Myocardial Reperfusion Injury/pathology , Myocytes, Cardiac/drug effects , Rats , Reactive Oxygen Species/metabolism , Reperfusion Injury/genetics , Reperfusion Injury/pathology , Sevoflurane/pharmacology , Signal Transduction/drug effects
7.
Cancer Cell ; 36(3): 268-287.e10, 2019 09 16.
Article in English | MEDLINE | ID: mdl-31447347

ABSTRACT

GAPDH is emerging as a key player in T cell development and function. To investigate the role of GAPDH in T cells, we generated a transgenic mouse model overexpressing GAPDH in the T cell lineage. Aged mice developed a peripheral Tfh-like lymphoma that recapitulated key molecular, pathological, and immunophenotypic features of human angioimmunoblastic T cell lymphoma (AITL). GAPDH induced non-canonical NF-κB pathway activation in mouse T cells, which was strongly activated in human AITL. We developed a NIK inhibitor to reveal that targeting the NF-κB pathway prolonged AITL-bearing mouse survival alone and in combination with anti-PD-1. These findings suggest the therapeutic potential of targeting NF-κB signaling in AITL and provide a model for future AITL therapeutic investigations.


Subject(s)
Glyceraldehyde-3-Phosphate Dehydrogenase (Phosphorylating)/metabolism , Immunoblastic Lymphadenopathy/pathology , Lymphoma, T-Cell/pathology , NF-kappa B/metabolism , T-Lymphocytes/immunology , Aged , Animals , Cell Line, Tumor , Cell Lineage/immunology , Datasets as Topic , Disease Models, Animal , Female , Gene Knockdown Techniques , Glyceraldehyde-3-Phosphate Dehydrogenase (Phosphorylating)/genetics , HEK293 Cells , Humans , Immunoblastic Lymphadenopathy/genetics , Lymphoma, T-Cell/drug therapy , Lymphoma, T-Cell/genetics , Lymphoma, T-Cell/immunology , Male , Mice, Transgenic , Middle Aged , NF-kappa B/genetics , Protein Kinase Inhibitors/administration & dosage , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Signal Transduction/immunology , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism , NF-kappaB-Inducing Kinase
8.
Cell Metab ; 29(6): 1243-1257.e10, 2019 06 04.
Article in English | MEDLINE | ID: mdl-30827861

ABSTRACT

Diffuse large B cell lymphoma (DLBCL) is a heterogeneous disease treated with anti-CD20-based immuno-chemotherapy (R-CHOP). We identified that low levels of GAPDH predict a poor response to R-CHOP treatment. Importantly, we demonstrated that GAPDHlow lymphomas use OxPhos metabolism and rely on mTORC1 signaling and glutaminolysis. Consistently, disruptors of OxPhos metabolism (phenformin) or glutaminolysis (L-asparaginase) induce cytotoxic responses in GAPDHlow B cells and improve GAPDHlow B cell-lymphoma-bearing mice survival, while they are low or not efficient on GAPDHhigh B cell lymphomas. Ultimately, we selected four GAPDHlow DLBCL patients, who were refractory to all anti-CD20-based therapies, and targeted DLBCL metabolism using L-asparaginase (K), mTOR inhibitor (T), and metformin (M) (called KTM therapy). Three out of the four patients presented a complete response upon one cycle of KTM. These findings establish that the GAPDH expression level predicts DLBCL patients' response to R-CHOP treatment and their sensitivity to specific metabolic inhibitors.


Subject(s)
Antimetabolites, Antineoplastic/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Glyceraldehyde-3-Phosphate Dehydrogenases/genetics , Lymphoma, Large B-Cell, Diffuse/drug therapy , Lymphoma, Large B-Cell, Diffuse/metabolism , Adult , Aged , Aged, 80 and over , Animals , Antimetabolites, Antineoplastic/administration & dosage , Cells, Cultured , Cohort Studies , Cyclophosphamide/therapeutic use , Doxorubicin/therapeutic use , Female , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Glyceraldehyde-3-Phosphate Dehydrogenases/metabolism , HEK293 Cells , Humans , Lymphoma, Large B-Cell, Diffuse/diagnosis , Lymphoma, Large B-Cell, Diffuse/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Middle Aged , Prednisone/therapeutic use , Prognosis , Retrospective Studies , Rituximab/therapeutic use , Treatment Outcome , Vincristine/therapeutic use , Young Adult
9.
J Invest Dermatol ; 139(6): 1306-1317, 2019 06.
Article in English | MEDLINE | ID: mdl-30571969

ABSTRACT

Inflammatory caspases, activated within the inflammasome, are responsible for the maturation and secretion of IL-1ß/IL-18. Although their expression in psoriasis was shown several years ago, little is known about the role of inflammatory caspases in the context of psoriasis. Here, we confirmed that caspases 1, 4, and 5 are activated in lesional skin from psoriasis patients. We showed in three psoriasis-like models that inflammatory caspases are activated, and accordingly, caspase 1/11 invalidation or pharmacological inhibition by Ac-YVAD-CMK (i.e., Ac-Tyr-Val-Ala-Asp-chloromethylketone) injection induced a decrease in ear thickness, erythema, scaling, inflammatory cytokine expression, and immune cell infiltration in mice. We observed that keratinocytes were primed to secrete IL-1ß when cultured in conditions mimicking psoriasis. Generation of chimeric mice by bone marrow transplantation was carried out to decipher the respective contribution of keratinocytes and/or immune cells in the activation of inflammatory caspases during psoriasis-like inflammatory response. Our data showed that the presence of caspase 1/11 in the immune system is sufficient for a fully inflammatory response, whereas the absence of caspase 1/11 in keratinocytes/fibroblasts had no impact. In summary, our study indicates that inflammatory caspases activated in immune cells are implicated in psoriasis pathogenesis.


Subject(s)
Caspase 1/deficiency , Caspase Inhibitors/administration & dosage , Caspases, Initiator/deficiency , Psoriasis/drug therapy , Amino Acid Chloromethyl Ketones/administration & dosage , Animals , Biopsy , Bone Marrow Transplantation , Caspase 1/genetics , Caspase 1/immunology , Caspases, Initiator/genetics , Caspases, Initiator/immunology , Caspases, Initiator/metabolism , Cells, Cultured , Clinical Trials as Topic , Female , Humans , Injections, Intraperitoneal , Interleukin-1beta/immunology , Interleukin-1beta/metabolism , Keratinocytes , Male , Mice , Mice, Knockout , Primary Cell Culture , Psoriasis/immunology , Psoriasis/pathology , Signal Transduction/drug effects , Signal Transduction/immunology , Skin/immunology , Skin/pathology , Transplantation Chimera
10.
Front Oncol ; 8: 556, 2018.
Article in English | MEDLINE | ID: mdl-30564554

ABSTRACT

Metabolism is a wide and general term that refers to any intracellular pathways the cell utilizes in order to satisfy its energetic demand and to support cell viability and/or division. Along with phenotypic changes, all mammalian cells including immune cells modulate their metabolic program in order to reach their effector functions. Exacerbated metabolism and metabolic flexibility are also hallmarks of tumor initiation and of tumor cell progression in a complex tumor microenvironment. Metabolic reprogramming is mainly directed by the serine/threonine kinase mTOR (mammalian target of rapamycin). mTOR exists in two structurally and functionally distinct complexes, mTORC1 and mTORC2 that coordinate environmental signals and metabolic/anabolic pathways to provide macromolecules and energy needed for survival and growth. Activation of mTORC1 is required during development, differentiation and activation of immune cells. Aberrant and persistent activation of mTORC1 is often observed in malignant B cells such as Non-Hodgkin's (NH) B-cell lymphomas. Here, we review recent insights on cell metabolism and on basic mechanisms of mTORC1 regulation and metabolic functions. We highlight the distinct mechanisms driving mTORC1 activation in the three most-common types of NH B-cell lymphomas (Diffuse Large B Cell Lymphomas, Follicular Lymphomas, and Mantle Cell Lymphomas), for which the first generation of mTORC1 inhibitors (rapalogs) have been extensively evaluated in preclinical and clinical settings. Finally, we discuss the reasons for limited clinical success of this therapy and focus on potential therapeutic strategies targeting metabolic pathways, upstream and downstream of mTORC1, that can be combined to rapalogs in order to improve patient's outcome.

11.
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
12.
Nat Biomed Eng ; 2(2): 85-94, 2018 02.
Article in English | MEDLINE | ID: mdl-31015628

ABSTRACT

Positron emission tomography-computed tomography (PET-CT) is the most sensitive molecular imaging modality, but it does not easily allow for rapid temporal acquisition. Ultrafast ultrasound imaging (UUI)-a recently introduced technology based on ultrasonic holography-leverages frame rates of up to several thousand images per second to quantitatively map, at high resolution, haemodynamic, biomechanical, electrophysiological and structural parameters. Here, we describe a pre-clinical scanner that registers PET-CT and UUI volumes acquired simultaneously and offers multiple combinations for imaging. We demonstrate that PET-CT-UUI allows for simultaneous images of the vasculature and metabolism during tumour growth in mice and rats, as well as for synchronized multi-modal cardiac cine-loops. Combined anatomical, functional and molecular imaging with PET-CT-UUI represents a high-performance and clinically translatable technology for biomedical research.


Subject(s)
Neoplasms/pathology , Positron Emission Tomography Computed Tomography , Ultrasonography , Animals , Cell Line, Tumor , Cricetinae , Female , Glucose/metabolism , Heart/anatomy & histology , Heart/diagnostic imaging , Mice , Myocardium/metabolism , Neoplasms/diagnostic imaging , Phenotype , Rats , Rats, Wistar
13.
Cell Rep ; 20(12): 2846-2859, 2017 Sep 19.
Article in English | MEDLINE | ID: mdl-28930681

ABSTRACT

Mitophagy is an evolutionarily conserved process that selectively targets impaired mitochondria for degradation. Defects in mitophagy are often associated with diverse pathologies, including cancer. Because the main known regulators of mitophagy are frequently inactivated in cancer cells, the mechanisms that regulate mitophagy in cancer cells are not fully understood. Here, we identified an E3 ubiquitin ligase (ARIH1/HHARI) that triggers mitophagy in cancer cells in a PINK1-dependent manner. We found that ARIH1/HHARI polyubiquitinates damaged mitochondria, leading to their removal via autophagy. Importantly, ARIH1 is widely expressed in cancer cells, notably in breast and lung adenocarcinomas; ARIH1 expression protects against chemotherapy-induced death. These data challenge the view that the main regulators of mitophagy are tumor suppressors, arguing instead that ARIH1-mediated mitophagy promotes therapeutic resistance.


Subject(s)
Antineoplastic Agents/pharmacology , Carrier Proteins/metabolism , Mitophagy , Neoplasms/metabolism , Ubiquitin-Protein Ligases/metabolism , Autophagy/drug effects , Cell Death/drug effects , Cell Line, Tumor , Cytoprotection/drug effects , HeLa Cells , Humans , Mitochondria/drug effects , Mitochondria/metabolism , Mitophagy/drug effects , Neoplasms/pathology , Protein Kinases/metabolism , Protein Stability/drug effects
14.
J Biol Chem ; 292(6): 2159-2173, 2017 02 10.
Article in English | MEDLINE | ID: mdl-28028180

ABSTRACT

Zinc-requiring ectoenzymes, including both secreted and membrane-bound enzymes, are considered to capture zinc in their active site for their activation in the early secretory pathway. This idea has been confirmed by our studies conducted using tissue-nonspecific alkaline phosphatase (TNAP), which is elaborately activated by means of a two-step mechanism by zinc transporter 5 (ZNT5)-ZNT6 heterodimers and ZNT7 homodimers, through protein stabilization followed by enzyme activation with zinc in the early secretory pathway. However, the molecular basis of the activation process in other zinc-requiring ectoenzymes remains largely unknown. In this study, we investigated this activation process by using three cancer-promoting zinc-requiring ectoenzymes, autotaxin (ATX), matrix metalloproteinase 9 (MMP9), and carbonic anhydrase IX (CAIX), and the chicken DT40 cell mutants that we generated; we specifically focused on clarifying whether the same or a similar activation mechanism operates in these ectoenzymes. ATX activation required ZNT5-ZNT6 heterodimers and ZNT7 homodimers in a manner similar to TNAP activation, although the protein stability of ATX was differently regulated from that of TNAP. MMP9 required ZNT5-ZNT6 heterodimers and ZNT7 homodimers for its activation as well as secretion; MMP9 was not secreted into the spent medium unless both zinc-transport complexes were present. Finally, CAIX activation by zinc was mediated not only by ZNT5-ZNT6 heterodimers and ZNT7 homodimers but also by ZNT4 homodimers; thus, these three zinc-transport complexes redundantly contribute to CAIX activation. Our results provide pivotal insights into the activation processes of zinc-requiring ectoenzymes, and furthermore, they offer novel insights for potential cancer therapy applications given the cancer-promoting potencies of ATX, MMP9, and CAIX.


Subject(s)
Carbonic Anhydrase IX/metabolism , Cation Transport Proteins/metabolism , Matrix Metalloproteinase 9/metabolism , Neoplasms/enzymology , Phosphoric Diester Hydrolases/metabolism , Zinc/metabolism , Animals , Cation Transport Proteins/chemistry , Cell Line , Chickens , Dimerization , Enzyme Activation
15.
Oncotarget ; 7(45): 73270-73279, 2016 11 08.
Article in English | MEDLINE | ID: mdl-27689327

ABSTRACT

Overexpression of Mcl-1 is implicated in resistance of several cancers to chemotherapeutic treatment, therefore identifying a safe way to decrease its expression in tumor cells represents a central goal. We investigated if a modulation of the diet could impact on Mcl-1 expression using a Myc-driven lymphoma model. We established that a partial reduction of caloric intake by 25% represents an efficient way to decrease Mcl-1 expression in tumor cells. Furthermore, using isocaloric custom diets, we observed that carbohydrates (CHO) are the main regulators of Mcl-1 expression within the food. Indeed, feeding lymphoma-bearing mice with a diet having 25% less carbohydrates was sufficient to decrease Mcl-1 expression by 50% in lymphoma cells. We showed that a low CHO diet resulted in AMPK activation and mTOR inhibition leading to eukaryotic elongation factor 2 (eEF2) inhibition, blocking protein translation elongation. Strikingly, a low CHO diet was sufficient to sensitize Myc-driven lymphoma-bearing mice to ABT-737-induced cell death in vivo. Thus reducing carbohydrate intake may represent a safe way to decrease Mcl-1 expression and to sensitize tumor cells to anti-cancer therapeutics.


Subject(s)
Biological Mimicry , Diet, Carbohydrate-Restricted , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Peptide Fragments/pharmacology , Proto-Oncogene Proteins/pharmacology , AMP-Activated Protein Kinases/metabolism , Animals , Biphenyl Compounds/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , Disease Models, Animal , Drug Resistance, Neoplasm/genetics , Heterografts , Humans , Lymphoma/drug therapy , Lymphoma/genetics , Lymphoma/metabolism , Lymphoma/pathology , Mice , Myeloid Cell Leukemia Sequence 1 Protein/chemistry , Myeloid Cell Leukemia Sequence 1 Protein/genetics , Nitrophenols/pharmacology , Piperazines/pharmacology , Protein Biosynthesis/drug effects , Signal Transduction , Sulfonamides/pharmacology , TOR Serine-Threonine Kinases/metabolism
16.
Blood ; 128(2): 253-64, 2016 07 14.
Article in English | MEDLINE | ID: mdl-27257182

ABSTRACT

Here we demonstrate that in a niche-like coculture system, cells from both primary and cultured acute myeloid leukemia (AML) sources take up functional mitochondria from murine or human bone marrow stromal cells. Using different molecular and imaging approaches, we show that AML cells can increase their mitochondrial mass up to 14%. After coculture, recipient AML cells showed a 1.5-fold increase in mitochondrial adenosine triphosphate production and were less prone to mitochondrial depolarization after chemotherapy, displaying a higher survival. This unidirectional transfer enhanced by some chemotherapeutic agents required cell-cell contacts and proceeded through an endocytic pathway. Transfer was greater in AML blasts compared with normal cord blood CD34(+) cells. Finally, we demonstrate that mitochondrial transfer was observed in vivo in an NSG immunodeficient mouse xenograft model and also occurred in human leukemia initiating cells and progenitors. As mitochondrial transfer provides a clear survival advantage following chemotherapy and a higher leukemic long-term culture initiating cell potential, targeting mitochondrial transfer could represent a future therapeutic target for AML treatment.


Subject(s)
Bone Marrow Cells/metabolism , Leukemia, Myeloid, Acute/metabolism , Mitochondria/metabolism , Animals , Bone Marrow Cells/pathology , Coculture Techniques , HL-60 Cells , Heterografts , Humans , Leukemia, Myeloid, Acute/pathology , Mice , Mice, Nude , Mitochondria/pathology , Neoplasm Transplantation , Stromal Cells/metabolism , Stromal Cells/pathology , U937 Cells
17.
J Enzyme Inhib Med Chem ; 30(5): 689-721, 2015.
Article in English | MEDLINE | ID: mdl-25347767

ABSTRACT

The hypoxic areas of solid cancers represent a negative prognostic factor irrespective of which treatment modality is chosen for the patient. Still, after almost 80 years of focus on the problems created by hypoxia in solid tumours, we still largely lack methods to deal efficiently with these treatment-resistant cells. The consequences of this lack may be serious for many patients: Not only is there a negative correlation between the hypoxic fraction in tumours and the outcome of radiotherapy as well as many types of chemotherapy, a correlation has been shown between the hypoxic fraction in tumours and cancer metastasis. Thus, on a fundamental basis the great variety of problems related to hypoxia in cancer treatment has to do with the broad range of functions oxygen (and lack of oxygen) have in cells and tissues. Therefore, activation-deactivation of oxygen-regulated cascades related to metabolism or external signalling are important areas for the identification of mechanisms as potential targets for hypoxia-specific treatment. Also the chemistry related to reactive oxygen radicals (ROS) and the biological handling of ROS are part of the problem complex. The problem is further complicated by the great variety in oxygen concentrations found in tissues. For tumour hypoxia to be used as a marker for individualisation of treatment there is a need for non-invasive methods to measure oxygen routinely in patient tumours. A large-scale collaborative EU-financed project 2009-2014 denoted METOXIA has studied all the mentioned aspects of hypoxia with the aim of selecting potential targets for new hypoxia-specific therapy and develop the first stage of tests for this therapy. A new non-invasive PET-imaging method based on the 2-nitroimidazole [(18)F]-HX4 was found to be promising in a clinical trial on NSCLC patients. New preclinical models for testing of the metastatic potential of cells were developed, both in vitro (2D as well as 3D models) and in mice (orthotopic grafting). Low density quantitative real-time polymerase chain reaction (qPCR)-based assays were developed measuring multiple hypoxia-responsive markers in parallel to identify tumour hypoxia-related patterns of gene expression. As possible targets for new therapy two main regulatory cascades were prioritised: The hypoxia-inducible-factor (HIF)-regulated cascades operating at moderate to weak hypoxia (<1% O(2)), and the unfolded protein response (UPR) activated by endoplasmatic reticulum (ER) stress and operating at more severe hypoxia (<0.2%). The prioritised targets were the HIF-regulated proteins carbonic anhydrase IX (CAIX), the lactate transporter MCT4 and the PERK/eIF2α/ATF4-arm of the UPR. The METOXIA project has developed patented compounds targeting CAIX with a preclinical documented effect. Since hypoxia-specific treatments alone are not curative they will have to be combined with traditional anti-cancer therapy to eradicate the aerobic cancer cell population as well.


Subject(s)
Drug Discovery , Neoplasms/drug therapy , Animals , Cell Hypoxia/drug effects , Dose-Response Relationship, Drug , Humans , Molecular Structure , Neoplasm Metastasis/drug therapy , Neoplasm Metastasis/pathology , Neoplasms/pathology , Structure-Activity Relationship
18.
Blood ; 122(14): 2402-11, 2013 Oct 03.
Article in English | MEDLINE | ID: mdl-23966420

ABSTRACT

Caloric restriction (CR) is proposed to decrease tumorigenesis through a variety of mechanisms including effects on glycolysis. However, the understanding of how CR affects the response to cancer therapy is still rudimentary. Here, using the Eµ-Myc transgenic mouse model of B-cell lymphoma, we report that by reducing protein translation, CR can reduce expression of the prosurvival Bcl-2 family member Mcl-1 and sensitize lymphomas to ABT-737-induced death in vivo. By using Eµ-Myc lymphoma cells lacking p53, we showed that CR mimetics such as 2-deoxyglucose led to a decrease in Mcl-1 expression and sensitized lymphoma cells to ABT-737-induced death independently of p53. In keeping with this, Eµ-Myc lymphoma cells lacking the BH3-only proapoptotic members Noxa, Puma, or Bim were also sensitized by CR mimetics to ABT-737-induced death. Remarkably, neither the loss of both Puma and Noxa, the loss of both Puma and Bim, nor the loss of all three BH3-only proteins prevented sensitization to ABT-737 induced by CR mimetics. Thus, CR can influence Mcl-1 expression and sensitize cells to BH3 mimetic-induced apoptosis, independently of the main BH3-only proteins and of p53. Exploiting this may improve the efficiency of, or prevent resistance to, cancer therapy.


Subject(s)
Caloric Restriction , Drug Resistance, Neoplasm/physiology , Lymphoma, B-Cell/metabolism , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Animals , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , BH3 Interacting Domain Death Agonist Protein/metabolism , Biphenyl Compounds/pharmacology , Disease Models, Animal , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Myeloid Cell Leukemia Sequence 1 Protein , Nitrophenols/pharmacology , Piperazines/pharmacology , Sulfonamides/pharmacology
19.
Lung Cancer ; 82(1): 16-23, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23910904

ABSTRACT

The disorganized neo-vasculature in tumours causes fluctuations in the concentration of oxygen, which contributes to tumour development and metastatic potential. Although hypoxic regulation of the expression of the carbonic anhydrases CAIX and CAXII is well established, the effect of re-oxygenation on these proteins remains to be elucidated. A549 and H1975 human lung cancer cell lines were exposed to hypoxia for 24 h and then re-oxygenated. CAIX or CAXII expression and cell cycle progression at different time-points were monitored. A549-shCA9 cells were analyzed for cell cycle progression in the same conditions. We demonstrate for the first time an association between the stability of CAIX and restoration of the S/G2 phase of hypoxia-arrested cells subjected to re-oxygenation. In exchange, we have found that the loss of CA9 did not cause a decreased progression into S/G2 phase during re-oxygenation, but rather affected the hypoxic growth arrest. We previously demonstrated that CAIX expression is a poor prognostic factor and that CAXII expression is a good prognostic factor in non-small cell lung cancer (NSCLC) patients. We further detail the relevance of the combined expression of these proteins for predicting outcome in a large population of NSCLC patients after long-term follow-up. The high CAIX/low CAXII expression sub-group was associated with a high cumulative incidence of relapse and with poor overall survival of NSCLC patients (P < 0.0001). Our results demonstrate a critical role for re-oxygenation on CAIX and CAXII levels that may select for an aggressive lung cancer phenotype. These findings suggest that CAIX and CAXII play dual roles in tumour progression and emphasize their significant prognostic and potential therapeutic value.


Subject(s)
Antigens, Neoplasm/metabolism , Carbonic Anhydrases/metabolism , Carcinoma, Non-Small-Cell Lung/enzymology , Lung Neoplasms/enzymology , Aged , Carbonic Anhydrase IX , Carcinoma, Non-Small-Cell Lung/mortality , Carcinoma, Non-Small-Cell Lung/pathology , Cell Hypoxia , Cell Line, Tumor , Cell Proliferation , Female , Humans , Kaplan-Meier Estimate , Lung Neoplasms/mortality , Lung Neoplasms/pathology , Male , Middle Aged , Oxygen/metabolism , Prognosis , Proportional Hazards Models
20.
Nat Rev Cancer ; 13(9): 611-23, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23969692

ABSTRACT

Intense interest in the 'Warburg effect' has been revived by the discovery that hypoxia-inducible factor 1 (HIF1) reprogrammes pyruvate oxidation to lactic acid conversion; lactic acid is the end product of fermentative glycolysis. The most aggressive and invasive cancers, which are often hypoxic, rely on exacerbated glycolysis to meet the increased demand for ATP and biosynthetic precursors and also rely on robust pH-regulating systems to combat the excessive generation of lactic and carbonic acids. In this Review, we present the key pH-regulating systems and synthesize recent advances in strategies that combine the disruption of pH control with bioenergetic mechanisms. We discuss the possibility of exploiting, in rapidly growing tumours, acute cell death by 'metabolic catastrophe'.


Subject(s)
Energy Metabolism , Neoplasms/drug therapy , Neoplasms/metabolism , Protons , Autophagy , Bicarbonates/metabolism , Carbonic Acid/metabolism , Carbonic Anhydrases/metabolism , Cation Transport Proteins/metabolism , Humans , Hydrogen-Ion Concentration , Hypoxia-Inducible Factor 1/metabolism , Lactic Acid/metabolism , Monocarboxylic Acid Transporters/metabolism , Muscle Proteins/metabolism , Sodium-Hydrogen Exchanger 1 , Sodium-Hydrogen Exchangers/metabolism , Symporters/metabolism , Tumor Microenvironment
SELECTION OF CITATIONS
SEARCH DETAIL
...