88 results on '"Porporato PE"'
Search Results
2. Targeting mitochondrial reactive oxygen species (mtROS) impairs breast cancer cell migration without affecting the cytotoxic effects of standard therapies
- Author
-
UCL - SSS/IREC/FATH - Pôle de Pharmacologie et thérapeutique, De Miranda Capeloa, Tania Isabel, Krzystyniak, Joanna, Porporato, PE, Sonveaux, Pierre, 6th meeting of the International Society of Cancer Metabolism (ISCaM), UCL - SSS/IREC/FATH - Pôle de Pharmacologie et thérapeutique, De Miranda Capeloa, Tania Isabel, Krzystyniak, Joanna, Porporato, PE, Sonveaux, Pierre, and 6th meeting of the International Society of Cancer Metabolism (ISCaM)
- Published
- 2019
3. Ghrelin and unacylated ghrelin inhibit muscle atrophy in C2C12 myotubes through p38 AND mTORC2 pathways without stimulating mTORC1 and protein synthesis
- Author
-
Ferrara, Michele, Reano, Simone, Porporato, Pe, Nicoletta Filigheddu, Graziani, Andrea, Ferrara, M, Reano, S, Porporato, Pe, Filigheddu, N, and Graziani, Andrea
- Published
- 2011
4. Diacylglycerol kinase alpha mediates 17-beta-estradiol-induced proliferation, motility and anchorage-independent growth of Hec-1A endometrial cancer cell line
- Author
-
Filigheddu N, Sampietro S, Porporato PE, Gaggianesi M, Chianale F, Ferrara M, Gregnanin I, Baldanzi G, Surico N, GRAZIANI , ANDREA, Filigheddu, N, Sampietro, S, Porporato, Pe, Gaggianesi, M, Chianale, F, Ferrara, M, Gregnanin, I, Baldanzi, G, Surico, N, and Graziani, Andrea
- Published
- 2011
5. Lactate stimulates angiogenesis, prevents skeletal muscle atrophy and accelerates wound healing
- Author
-
Payen VL, Porporato PE, De Saedeleer CJ, Prxe9at V, Feron O, and Sonveaux P
- Published
- 2014
6. Intradermale in vivo Elektroporation von Host Defense Peptid hCAP-18/LL-37 fördert die Wundheilung von akuten, diabetischen und ischämischen Wunden
- Author
-
Lam, MC, Jacobsen, F, Becerikli, M, Merwart, B, Rittig, A, Vandermeulen, G, Porporato, PE, Sonveaux, P, Préat, V, and Steinsträßer, L
- Subjects
ddc: 610 ,610 Medical sciences ,Medicine - Abstract
Einleitung: Gentherapeutische Ansätze der Applikation von Host Defense Peptiden zeigen vielversprechende Ergebnisse zur Förderung der Wundheilung und Möglichkeiten des Einsatzes als Antibiotikum der Zukunft. Effiziente nicht-virale in-vivo Techniken zum Gentransfer von DNA, die therapeutische[for full text, please go to the a.m. URL], 44. Jahrestagung der Deutschen Gesellschaft der Plastischen, Rekonstruktiven und Ästhetischen Chirurgen (DGPRÄC), 17. Jahrestagung der Vereinigung der Deutschen Ästhetisch-Plastischen Chirurgen (VDÄPC)
- Published
- 2013
- Full Text
- View/download PDF
7. Lactate activates HIF-1 in oxidative but not in Warburg-phenotype tumor cells
- Author
-
De Saedeleer CJ, Copetti T, Porporato PE, Verrax J, Feron O, and Sonveaux P
- Published
- 2012
8. DIACYLGLYCEROL KINASE ALPHA IS REQUIRED FOR PROLIFERATION AND INVASION INDUCED BY GROWTH FACTORS AND CHEMOKINES
- Author
-
Porporato, Pe, Rainero, E., Chianale, F., Ranaldo, G., Arlandis, Gt, Locatelli, I., Gianluca Baldanzi, Nicoletta Filigheddu, Traini, S., Gaggianesi, M., Graziani, Andrea, Porporato, Pe, Rainero, E, Chianale, F, Ranaldo, G, Arlandis, Gt, Locatelli, I, Baldanzi, G, Filigheddu, N, Traini, S, Gaggianesi, M, and Graziani, Andrea
9. Differential Expression of MicroRNAs between Eutopic and Ectopic Endometrium in Ovarian Endometriosis
- Author
-
Paolo E. Porporato, Nicola Surico, Nicoletta Filigheddu, Andrea Graziani, Ilaria Gregnanin, Claudia Patrignani, Daniela Surico, Licia Galli, Beatrice Perego, UCL - SSS/IREC - Institut de recherche expérimentale et clinique, UCL - SSS/IREC/FATH - Pôle de Pharmacologie et thérapeutique, Filigheddu, N, Gregnanin, I, Porporato, Pe, Surico, D, Perego, B, Galli, L, Patrignani, C, Graziani, Andrea, and Surico, N.
- Subjects
Adult ,Pathology ,medicine.medical_specialty ,Article Subject ,Health, Toxicology and Mutagenesis ,lcsh:Biotechnology ,Endometriosis ,Gene Expression ,lcsh:Medicine ,Biology ,lcsh:Chemical technology ,Endometrium ,lcsh:Technology ,lcsh:TP248.13-248.65 ,microRNA ,Gene expression ,Genetics ,medicine ,Humans ,lcsh:TP1-1185 ,Toxicology and Mutagenesis ,Ovarian Diseases ,Molecular Biology ,Regulation of gene expression ,lcsh:T ,Microarray analysis techniques ,Medicine (all) ,lcsh:R ,General Medicine ,Middle Aged ,medicine.disease ,Non-coding RNA ,MicroRNAs ,medicine.anatomical_structure ,Health ,Ovarian Endometriosis ,Cancer research ,Molecular Medicine ,Female ,Biotechnology ,Research Article - Abstract
Endometriosis, defined as the presence of endometrial tissue outside the uterus, is a common gynecological disease with poorly understood pathogenesis. MicroRNAs are members of a class of small noncoding RNA molecules that have a critical role in posttranscriptional regulation of gene expression by repression of target mRNAs translation. We assessed differentially expressed microRNAs in ectopic endometrium compared with eutopic endometrium in 3 patients through microarray analysis. We identified 50 microRNAs differentially expressed and the differential expression of five microRNAs was validated by real-time RT-PCR in other 13 patients. We identifiedin silicotheir predicted targets, several of which match the genes that have been identified to be differentially expressed in ectopicversuseutopic endometrium in studies of gene expression. A functional analysis of the predicted targets indicates that several of these are involved in molecular pathways implicated in endometriosis, thus strengthening the hypothesis of the role of microRNAs in this pathology.
- Published
- 2010
- Full Text
- View/download PDF
10. Diacylglycerol kinase is required for HGF-induced invasiveness and anchorage-independent growth of MDA-MB-231 breast cancer cells
- Author
-
Filigheddu, N., Cutrupi, S., Paolo Ettore Porporato, Riboni, F., Baldanzi, G., Chianale, F., Fortina, E., Piantanida, P., Bortoli, M., Vacca, G., Graziani, A., Surico, N., Filigheddu, N, Cutrupi, S, Porporato, Pe, Riboni, F, Baldanzi, G, Chianale, F, Fortina, E, Piantanida, P, DE BORTOLI, M, Vacca, G, Graziani, Andrea, and Surico, N.
- Subjects
cMet ,Hepatocyte Growth Factor ,invasiveness ,Cell Culture Techniques ,diacylglycerol kinase ,MDA-MB-231 cell line ,Breast Neoplasms ,Breast cancer ,HGF ,Receptors, Estrogen ,Cell Movement ,Cell Line, Tumor ,Humans ,Neoplasm Invasiveness - Abstract
Estrogen receptor (ER)-negative breast cancers have a worse prognosis than ER-positive cancers, being more aggressive and overexposed to stimuli leading to their progression. Hepatocyte growth factor (HGF) has been associated with proliferation, migration and invasion of tumor cells, and several tumors, including those of breast cancer, produce HGF and overexpress its receptor. Diacylglycerol kinases (Dgks), which phosphorylate diacylglycerol to phosphatidic acid, are key regulators of cell signaling. Our research was focused on their role in HGF-induced invasion of MDA-MB-231 cells, a model of ER-negative breast cancer.Dgk activity was evaluated with a kinase assay, MDA-MB-231 cell invasion via culturing of cells in matrigel-coated transwells, and anchorage-independent growth was assessed using a soft agar assay.HGF induces Dgk activation in MDA-MB-231 cells that is required for cell invasiveness. Moreover, Dgks are involved in MDA-MB-231 anchorage-independent growth.Dgks could be a target for ER-negative breast cancer therapy.
11. RICTOR/mTORC2 downregulation in BRAF V600E melanoma cells promotes resistance to BRAF/MEK inhibition.
- Author
-
Ponzone L, Audrito V, Landi C, Moiso E, Levra Levron C, Ferrua S, Savino A, Vitale N, Gasparrini M, Avalle L, Vantaggiato L, Shaba E, Tassone B, Saoncella S, Orso F, Viavattene D, Marina E, Fiorilla I, Burrone G, Abili Y, Altruda F, Bini L, Deaglio S, Defilippi P, Menga A, Poli V, Porporato PE, Provero P, Raffaelli N, Riganti C, Taverna D, Cavallo F, and Calautti E
- Subjects
- Animals, Humans, Mice, Cell Line, Tumor, Down-Regulation, Gene Expression Regulation, Neoplastic, Mutation, Proteomics methods, Xenograft Model Antitumor Assays, MAP Kinase Kinase Kinases antagonists & inhibitors, Drug Resistance, Neoplasm genetics, Mechanistic Target of Rapamycin Complex 2 metabolism, Mechanistic Target of Rapamycin Complex 2 genetics, Melanoma genetics, Melanoma drug therapy, Melanoma metabolism, Melanoma pathology, Protein Kinase Inhibitors pharmacology, Protein Kinase Inhibitors therapeutic use, Proto-Oncogene Proteins B-raf antagonists & inhibitors, Proto-Oncogene Proteins B-raf genetics, Rapamycin-Insensitive Companion of mTOR Protein metabolism, Rapamycin-Insensitive Companion of mTOR Protein genetics
- Abstract
Background: The main drawback of BRAF/MEK inhibitors (BRAF/MEKi)-based targeted therapy in the management of BRAF-mutated cutaneous metastatic melanoma (MM) is the development of therapeutic resistance. We aimed to assess in this context the role of mTORC2, a signaling complex defined by the presence of the essential RICTOR subunit, regarded as an oncogenic driver in several tumor types, including MM., Methods: After analyzing The Cancer Genome Atlas MM patients' database to explore both overall survival and molecular signatures as a function of intra-tumor RICTOR levels, we investigated the effects of RICTOR downregulation in BRAF
V600E MM cell lines on their response to BRAF/MEKi. We performed proteomic screening to identify proteins modulated by changes in RICTOR expression, and Seahorse analysis to evaluate the effects of RICTOR depletion on mitochondrial respiration. The combination of BRAFi with drugs targeting proteins and processes emerged in the proteomic screening was carried out on RICTOR-deficient cells in vitro and in a xenograft setting in vivo., Results: Low RICTOR levels in BRAF-mutated MM correlate with a worse clinical outcome. Gene Set Enrichment Analysis of low-RICTOR tumors display gene signatures suggestive of activation of the mitochondrial Electron Transport Chain (ETC) energy production. RICTOR-deficient BRAFV600E cells are intrinsically tolerant to BRAF/MEKi and anticipate the onset of resistance to BRAFi upon prolonged drug exposure. Moreover, in drug-naïve cells we observed a decline in RICTOR expression shortly after BRAFi exposure. In RICTOR-depleted cells, both mitochondrial respiration and expression of nicotinamide phosphoribosyltransferase (NAMPT) are enhanced, and their pharmacological inhibition restores sensitivity to BRAFi., Conclusions: Our work unveils an unforeseen tumor-suppressing role for mTORC2 in the early adaptation phase of BRAFV600E melanoma cells to targeted therapy and identifies the NAMPT-ETC axis as a potential therapeutic vulnerability of low RICTOR tumors. Importantly, our findings indicate that the evaluation of intra-tumor RICTOR levels has a prognostic value in metastatic melanoma and may help to guide therapeutic strategies in a personalized manner., (© 2024. The Author(s).)- Published
- 2024
- Full Text
- View/download PDF
12. Flvcr1a deficiency promotes heme-based energy metabolism dysfunction in skeletal muscle.
- Author
-
Mistretta M, Fiorito V, Allocco AL, Ammirata G, Hsu MY, Digiovanni S, Belicchi M, Napoli L, Ripolone M, Trombetta E, Mauri P, Farini A, Meregalli M, Villa C, Porporato PE, Miniscalco B, Crich SG, Riganti C, Torrente Y, and Tolosano E
- Subjects
- Mice, Animals, Heme metabolism, Mice, Knockout, Muscle, Skeletal metabolism, Energy Metabolism, Cell Differentiation physiology, Membrane Transport Proteins metabolism, Satellite Cells, Skeletal Muscle metabolism
- Abstract
The definition of cell metabolic profile is essential to ensure skeletal muscle fiber heterogeneity and to achieve a proper equilibrium between the self-renewal and commitment of satellite stem cells. Heme sustains several biological functions, including processes profoundly implicated with cell metabolism. The skeletal muscle is a significant heme-producing body compartment, but the consequences of impaired heme homeostasis on this tissue have been poorly investigated. Here, we generate a skeletal-muscle-specific feline leukemia virus subgroup C receptor 1a (FLVCR1a) knockout mouse model and show that, by sustaining heme synthesis, FLVCR1a contributes to determine the energy phenotype in skeletal muscle cells and to modulate satellite cell differentiation and muscle regeneration., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2024 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
13. FK506 bypasses the effect of erythroferrone in cancer cachexia skeletal muscle atrophy.
- Author
-
Mina E, Wyart E, Sartori R, Angelino E, Zaggia I, Rausch V, Maldotti M, Pagani A, Hsu MY, Friziero A, Sperti C, Menga A, Graziani A, Hirsch E, Oliviero S, Sandri M, Conti L, Kautz L, Silvestri L, and Porporato PE
- Subjects
- Humans, Mice, Animals, Tacrolimus metabolism, Tacrolimus pharmacology, Muscle, Skeletal metabolism, Tacrolimus Binding Protein 1A genetics, Tacrolimus Binding Protein 1A metabolism, Tacrolimus Binding Protein 1A pharmacology, Muscular Atrophy drug therapy, Muscular Atrophy metabolism, Muscular Atrophy pathology, Cachexia drug therapy, Cachexia etiology, Cachexia metabolism, Neoplasms pathology
- Abstract
Skeletal muscle atrophy is a hallmark of cachexia, a wasting condition typical of chronic pathologies, that still represents an unmet medical need. Bone morphogenetic protein (BMP)-Smad1/5/8 signaling alterations are emerging drivers of muscle catabolism, hence, characterizing these perturbations is pivotal to develop therapeutic approaches. We identified two promoters of "BMP resistance" in cancer cachexia, specifically the BMP scavenger erythroferrone (ERFE) and the intracellular inhibitor FKBP12. ERFE is upregulated in cachectic cancer patients' muscle biopsies and in murine cachexia models, where its expression is driven by STAT3. Moreover, the knock down of Erfe or Fkbp12 reduces muscle wasting in cachectic mice. To bypass the BMP resistance mediated by ERFE and release the brake on the signaling, we targeted FKBP12 with low-dose FK506. FK506 restores BMP-Smad1/5/8 signaling, rescuing myotube atrophy by inducing protein synthesis. In cachectic tumor-bearing mice, FK506 prevents muscle and body weight loss and protects from neuromuscular junction alteration, suggesting therapeutic potential for targeting the ERFE-FKBP12 axis., Competing Interests: Declaration of interests E.M., V.R., L.S., and P.E.P. declare to be inventors of the patent PCTIB2022050175 (WO2022/149113)., (Copyright © 2023 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
14. Mitochondria Transplantation Mitigates Damage in an In Vitro Model of Renal Tubular Injury and in an Ex Vivo Model of DCD Renal Transplantation.
- Author
-
Rossi A, Asthana A, Riganti C, Sedrakyan S, Byers LN, Robertson J, Senger RS, Montali F, Grange C, Dalmasso A, Porporato PE, Palles C, Thornton ME, Da Sacco S, Perin L, Ahn B, McCully J, Orlando G, and Bussolati B
- Subjects
- Humans, Swine, Animals, Kidney metabolism, Mitochondria metabolism, Mitochondria pathology, Kidney Transplantation, Reperfusion Injury, Acute Kidney Injury prevention & control, Acute Kidney Injury metabolism
- Abstract
Objectives: To test whether mitochondrial transplantation (MITO) mitigates damage in 2 models of acute kidney injury (AKI)., Background: MITO is a process where exogenous isolated mitochondria are taken up by cells. As virtually any morbid clinical condition is characterized by mitochondrial distress, MITO may find a role as a treatment modality in numerous clinical scenarios including AKI., Methods: For the in vitro experiments, human proximal tubular cells were damaged and then treated with mitochondria or placebo. For the ex vivo experiments, we developed a non-survival ex vivo porcine model mimicking the donation after cardiac death renal transplantation scenario. One kidney was treated with mitochondria, although the mate organ received placebo, before being perfused at room temperature for 24 hours. Perfusate samples were collected at different time points and analyzed with Raman spectroscopy. Biopsies taken at baseline and 24 hours were analyzed with standard pathology, immunohistochemistry, and RNA sequencing analysis., Results: In vitro, cells treated with MITO showed higher proliferative capacity and adenosine 5'-triphosphate production, preservation of physiological polarization of the organelles and lower toxicity and reactive oxygen species production. Ex vivo, kidneys treated with MITO shed fewer molecular species, indicating stability. In these kidneys, pathology showed less damage whereas RNAseq analysis showed modulation of genes and pathways most consistent with mitochondrial biogenesis and energy metabolism and downregulation of genes involved in neutrophil recruitment, including IL1A, CXCL8, and PIK3R1., Conclusions: MITO mitigates AKI both in vitro and ex vivo., Competing Interests: J.R. and R.S.S. are co-founders of Rametrix Technologies, on the board of which J.R. serves as President and CEO, whereas R.S.S. as Chief Technology Officer. The remaining authors report no conflicts of interest., (Copyright © 2023 The Author(s). Published by Wolters Kluwer Health, Inc.)
- Published
- 2023
- Full Text
- View/download PDF
15. NAD + repletion with niacin counteracts cancer cachexia.
- Author
-
Beltrà M, Pöllänen N, Fornelli C, Tonttila K, Hsu MY, Zampieri S, Moletta L, Corrà S, Porporato PE, Kivelä R, Viscomi C, Sandri M, Hulmi JJ, Sartori R, Pirinen E, and Penna F
- Subjects
- Humans, Mice, Animals, NAD metabolism, Cachexia drug therapy, Cachexia etiology, Cachexia metabolism, Niacinamide metabolism, Muscle, Skeletal metabolism, Niacin pharmacology, Niacin therapeutic use, Niacin metabolism, Neoplasms complications, Neoplasms drug therapy, Neoplasms metabolism
- Abstract
Cachexia is a debilitating wasting syndrome and highly prevalent comorbidity in cancer patients. It manifests especially with energy and mitochondrial metabolism aberrations that promote tissue wasting. We recently identified nicotinamide adenine dinucleotide (NAD
+ ) loss to associate with muscle mitochondrial dysfunction in cancer hosts. In this study we confirm that depletion of NAD+ and downregulation of Nrk2, an NAD+ biosynthetic enzyme, are common features of severe cachexia in different mouse models. Testing NAD+ repletion therapy in cachectic mice reveals that NAD+ precursor, vitamin B3 niacin, efficiently corrects tissue NAD+ levels, improves mitochondrial metabolism and ameliorates cancer- and chemotherapy-induced cachexia. In a clinical setting, we show that muscle NRK2 is downregulated in cancer patients. The low expression of NRK2 correlates with metabolic abnormalities underscoring the significance of NAD+ in the pathophysiology of human cancer cachexia. Overall, our results propose NAD+ metabolism as a therapy target for cachectic cancer patients., (© 2023. The Author(s).)- Published
- 2023
- Full Text
- View/download PDF
16. Lysosomal lipid switch sensitises to nutrient deprivation and mTOR targeting in pancreatic cancer.
- Author
-
De Santis MC, Gozzelino L, Margaria JP, Costamagna A, Ratto E, Gulluni F, Di Gregorio E, Mina E, Lorito N, Bacci M, Lattanzio R, Sala G, Cappello P, Novelli F, Giovannetti E, Vicentini C, Andreani S, Delfino P, Corbo V, Scarpa A, Porporato PE, Morandi A, Hirsch E, and Martini M
- Subjects
- Animals, Mice, Cell Line, Tumor, Cell Proliferation, Glutamine metabolism, Mechanistic Target of Rapamycin Complex 1 metabolism, Nutrients, Signal Transduction, TOR Serine-Threonine Kinases metabolism, Glutaminase, Pancreatic Neoplasms, Carcinoma, Pancreatic Ductal drug therapy, Carcinoma, Pancreatic Ductal genetics, Carcinoma, Pancreatic Ductal metabolism, Lipids biosynthesis, Lysosomes metabolism, Pancreatic Neoplasms drug therapy, Pancreatic Neoplasms genetics, Pancreatic Neoplasms metabolism, Phosphatidylinositol 3-Kinases genetics, Phosphatidylinositol 3-Kinases metabolism, Everolimus therapeutic use, MTOR Inhibitors therapeutic use
- Abstract
Objective: Pancreatic ductal adenocarcinoma (PDAC) is an aggressive disease with limited therapeutic options. However, metabolic adaptation to the harsh PDAC environment can expose liabilities useful for therapy. Targeting the key metabolic regulator mechanistic target of rapamycin complex 1 (mTORC1) and its downstream pathway shows efficacy only in subsets of patients but gene modifiers maximising response remain to be identified., Design: Three independent cohorts of PDAC patients were studied to correlate PI3K-C2γ protein abundance with disease outcome. Mechanisms were then studied in mouse (KPC mice) and cellular models of PDAC, in presence or absence of PI3K-C2γ (WT or KO). PI3K-C2γ-dependent metabolic rewiring and its impact on mTORC1 regulation were assessed in conditions of limiting glutamine availability. Finally, effects of a combination therapy targeting mTORC1 and glutamine metabolism were studied in WT and KO PDAC cells and preclinical models., Results: PI3K-C2γ expression was reduced in about 30% of PDAC cases and was associated with an aggressive phenotype. Similarly, loss of PI3K-C2γ in KPC mice enhanced tumour development and progression. The increased aggressiveness of tumours lacking PI3K-C2γ correlated with hyperactivation of mTORC1 pathway and glutamine metabolism rewiring to support lipid synthesis. PI3K-C2γ-KO tumours failed to adapt to metabolic stress induced by glutamine depletion, resulting in cell death., Conclusion: Loss of PI3K-C2γ prevents mTOR inactivation and triggers tumour vulnerability to RAD001 (mTOR inhibitor) and BPTES/CB-839 (glutaminase inhibitors). Therefore, these results might open the way to personalised treatments in PDAC with PI3K-C2γ loss., Competing Interests: Competing interests: EH is a founder of Kither Biotech, a company involved in the development of PI3K inhibitors. The authors declare no potential conflicts of interest., (© Author(s) (or their employer(s)) 2023. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.)
- Published
- 2023
- Full Text
- View/download PDF
17. Inhibition of Mitochondrial Redox Signaling with MitoQ Prevents Metastasis of Human Pancreatic Cancer in Mice.
- Author
-
Capeloa T, Van de Velde JA, d'Hose D, Lipari SG, Derouane F, Hamelin L, Bedin M, Vazeille T, Duhoux FP, Murphy MP, Porporato PE, Gallez B, and Sonveaux P
- Abstract
At diagnosis, about 35% of pancreatic cancers are at the locally invasive yet premetastatic stage. Surgical resection is not a treatment option, leaving patients with a largely incurable disease that often evolves to the polymetastatic stage despite chemotherapeutic interventions. In this preclinical study, we hypothesized that pancreatic cancer metastasis can be prevented by inhibiting mitochondrial redox signaling with MitoQ, a mitochondria-targeted antioxidant. Using four different cancer cell lines, we report that, at clinically relevant concentrations (100-500 nM), MitoQ selectively repressed mesenchymal pancreatic cancer cell respiration, which involved the inhibition of the expression of PGC-1α, NRF1 and a reduced expression of electron-transfer-chain complexes I to III. MitoQ consequently decreased the mitochondrial membrane potential and mitochondrial superoxide production by these cells. Phenotypically, MitoQ further inhibited pancreatic cancer cell migration, invasion, clonogenicity and the expression of stem cell markers. It reduced by ~50% the metastatic homing of human MIA PaCa-2 cells in the lungs of mice. We further show that combination treatments with chemotherapy are conceivable. Collectively, this study indicates that the inhibition of mitochondrial redox signaling is a possible therapeutic option to inhibit the metastatic progression of pancreatic cancer.
- Published
- 2022
- Full Text
- View/download PDF
18. IFI16 Impacts Metabolic Reprogramming during Human Cytomegalovirus Infection.
- Author
-
Griffante G, Hewelt-Belka W, Albano C, Gugliesi F, Pasquero S, Castillo Pacheco SF, Bajetto G, Porporato PE, Mina E, Vallino M, Krapp C, Jakobsen MR, Purdy J, von Einem J, Landolfo S, Dell'Oste V, and Biolatti M
- Subjects
- DNA, Viral genetics, Fibroblasts, Humans, Virus Replication, Cellular Reprogramming, Cytomegalovirus physiology, Cytomegalovirus Infections, Nuclear Proteins genetics, Nuclear Proteins metabolism, Phosphoproteins genetics, Phosphoproteins metabolism
- Abstract
Cellular lipid metabolism plays a pivotal role in human cytomegalovirus (HCMV) infection, as increased lipogenesis in HCMV-infected cells favors the envelopment of newly synthesized viral particles. As all cells are equipped with restriction factors (RFs) able to exert a protective effect against invading pathogens, we asked whether a similar defense mechanism would also be in place to preserve the metabolic compartment from HCMV infection. Here, we show that gamma interferon (IFN-γ)-inducible protein 16 (IFI16), an RF able to block HCMV DNA synthesis, can also counteract HCMV-mediated metabolic reprogramming in infected primary human foreskin fibroblasts (HFFs), thereby limiting virion infectivity. Specifically, we find that IFI16 downregulates the transcriptional activation of the glucose transporter 4 (GLUT4) through cooperation with the carbohydrate-response element-binding protein (ChREBP), thereby reducing HCMV-induced transcription of lipogenic enzymes. The resulting decrease in glucose uptake and consumption leads to diminished lipid synthesis, which ultimately curbs the de novo formation of enveloped viral particles in infected HFFs. Consistently, untargeted lipidomic analysis shows enhanced cholesteryl ester levels in IFI16 KO versus wild-type (WT) HFFs. Overall, our data unveil a new role of IFI16 in the regulation of glucose and lipid metabolism upon HCMV replication and uncover new potential targets for the development of novel antiviral therapies. IMPORTANCE Human cytomegalovirus (HCMV) gathers all the substrates and enzymes necessary for the assembly of new virions from its host cell. For instance, HCMV is known to induce cellular metabolism of infected cells to favor virion assembly. Cells are, however, equipped with a first-line defense represented by restriction factors (RFs), which after sensing viral DNA can trigger innate and adaptive responses, thereby blocking HCMV replication. One such RF is IFN-γ-inducible protein 16 (IFI16), which we have shown to downregulate viral replication in human fibroblasts. Thus, we asked whether IFI16 would also play a role in preserving cellular metabolism upon HCMV infection. Our findings highlight an unprecedented role of IFI16 in opposing the metabolic changes elicited by HCMV, thus revealing new promising targets for antiviral therapy.
- Published
- 2022
- Full Text
- View/download PDF
19. Iron supplementation is sufficient to rescue skeletal muscle mass and function in cancer cachexia.
- Author
-
Wyart E, Hsu MY, Sartori R, Mina E, Rausch V, Pierobon ES, Mezzanotte M, Pezzini C, Bindels LB, Lauria A, Penna F, Hirsch E, Martini M, Mazzone M, Roetto A, Geninatti Crich S, Prenen H, Sandri M, Menga A, and Porporato PE
- Subjects
- Animals, Dietary Supplements, Humans, Iron metabolism, Mice, Muscle, Skeletal metabolism, Cachexia etiology, Cachexia metabolism, Neoplasms complications, Neoplasms drug therapy, Neoplasms metabolism
- Abstract
Cachexia is a wasting syndrome characterized by devastating skeletal muscle atrophy that dramatically increases mortality in various diseases, most notably in cancer patients with a penetrance of up to 80%. Knowledge regarding the mechanism of cancer-induced cachexia remains very scarce, making cachexia an unmet medical need. In this study, we discovered strong alterations of iron metabolism in the skeletal muscle of both cancer patients and tumor-bearing mice, characterized by decreased iron availability in mitochondria. We found that modulation of iron levels directly influences myotube size in vitro and muscle mass in otherwise healthy mice. Furthermore, iron supplementation was sufficient to preserve both muscle function and mass, prolong survival in tumor-bearing mice, and even rescues strength in human subjects within an unexpectedly short time frame. Importantly, iron supplementation refuels mitochondrial oxidative metabolism and energy production. Overall, our findings provide new mechanistic insights in cancer-induced skeletal muscle wasting, and support targeting iron metabolism as a potential therapeutic option for muscle wasting diseases., (© 2022 The Authors. Published under the terms of the CC BY 4.0 license.)
- Published
- 2022
- Full Text
- View/download PDF
20. MitoQ Inhibits Human Breast Cancer Cell Migration, Invasion and Clonogenicity.
- Author
-
Capeloa T, Krzystyniak J, d'Hose D, Canas Rodriguez A, Payen VL, Zampieri LX, Van de Velde JA, Benyahia Z, Pranzini E, Vazeille T, Fransolet M, Bouzin C, Brusa D, Michiels C, Gallez B, Murphy MP, Porporato PE, and Sonveaux P
- Abstract
To successfully generate distant metastases, metastatic progenitor cells must simultaneously possess mesenchymal characteristics, resist to anoïkis, migrate and invade directionally, resist to redox and shear stresses in the systemic circulation, and possess stem cell characteristics. These cells primarily originate from metabolically hostile areas of the primary tumor, where oxygen and nutrient deprivation, together with metabolic waste accumulation, exert a strong selection pressure promoting evasion. Here, we followed the hypothesis according to which metastasis as a whole implies the existence of metabolic sensors. Among others, mitochondria are singled out as a major source of superoxide that supports the metastatic phenotype. Molecularly, stressed cancer cells increase mitochondrial superoxide production, which activates the transforming growth factor-β pathway through src directly within mitochondria, ultimately activating focal adhesion kinase Pyk2. The existence of mitochondria-targeted antioxidants constitutes an opportunity to interfere with the metastatic process. Here, using aggressive triple-negative and HER2-positive human breast cancer cell lines as models, we report that MitoQ inhibits all the metastatic traits that we tested in vitro. Compared to other mitochondria-targeted antioxidants, MitoQ already successfully passed Phase I safety clinical trials, which provides an important incentive for future preclinical and clinical evaluations of this drug for the prevention of breast cancer metastasis.
- Published
- 2022
- Full Text
- View/download PDF
21. MitoQ Prevents Human Breast Cancer Recurrence and Lung Metastasis in Mice.
- Author
-
Capeloa T, Krzystyniak J, Rodriguez AC, Payen VL, Zampieri LX, Pranzini E, Derouane F, Vazeille T, Bouzin C, Duhoux FP, Murphy MP, Porporato PE, and Sonveaux P
- Abstract
In oncology, the occurrence of distant metastases often marks the transition from curative to palliative care. Such outcome is highly predictable for breast cancer patients, even if tumors are detected early, and there is no specific treatment to prevent metastasis. Previous observations indicated that cancer cell mitochondria are bioenergetic sensors of the tumor microenvironment that produce superoxide to promote evasion. Here, we tested whether mitochondria-targeted antioxidant MitoQ is capable to prevent metastasis in the MDA-MB-231 model of triple-negative human breast cancer in mice and in the MMTV-PyMT model of spontaneously metastatic mouse breast cancer. At clinically relevant doses, we report that MitoQ not only prevented metastatic take and dissemination, but also local recurrence after surgery. We further provide in vitro evidence that MitoQ does not interfere with conventional chemotherapies used to treat breast cancer patients. Since MitoQ already successfully passed Phase I safety clinical trials, our preclinical data collectively provide a strong incentive to test this drug for the prevention of cancer dissemination and relapse in clinical trials with breast cancer patients.
- Published
- 2022
- Full Text
- View/download PDF
22. Phosphoinositide Conversion Inactivates R-RAS and Drives Metastases in Breast Cancer.
- Author
-
Li H, Prever L, Hsu MY, Lo WT, Margaria JP, De Santis MC, Zanini C, Forni M, Novelli F, Pece S, Di Fiore PP, Porporato PE, Martini M, Belabed H, Nazare M, Haucke V, Gulluni F, and Hirsch E
- Subjects
- Cell Adhesion physiology, Female, Focal Adhesions metabolism, Focal Adhesions pathology, GTPase-Activating Proteins metabolism, Humans, Phosphatidylinositols metabolism, Breast Neoplasms
- Abstract
Breast cancer is the most prevalent cancer and a major cause of death in women worldwide. Although early diagnosis and therapeutic intervention significantly improve patient survival rate, metastasis still accounts for most deaths. Here it is reported that, in a cohort of more than 2000 patients with breast cancer, overexpression of PI3KC2α occurs in 52% of cases and correlates with high tumor grade as well as increased probability of distant metastatic events, irrespective of the subtype. Mechanistically, it is demonstrated that PI3KC2α synthetizes a pool of PI(3,4)P2 at focal adhesions that lowers their stability and directs breast cancer cell migration, invasion, and metastasis. PI(3,4)P2 locally produced by PI3KC2α at focal adhesions recruits the Ras GTPase activating protein 3 (RASA3), which inactivates R-RAS, leading to increased focal adhesion turnover, migration, and invasion both in vitro and in vivo. Proof-of-concept is eventually provided that inhibiting PI3KC2α or lowering RASA3 activity at focal adhesions significantly reduces the metastatic burden in PI3KC2α-overexpressing breast cancer, thereby suggesting a novel strategy for anti-breast cancer therapy., (© 2022 The Authors. Advanced Science published by Wiley-VCH GmbH.)
- Published
- 2022
- Full Text
- View/download PDF
23. The heme synthesis-export system regulates the tricarboxylic acid cycle flux and oxidative phosphorylation.
- Author
-
Fiorito V, Allocco AL, Petrillo S, Gazzano E, Torretta S, Marchi S, Destefanis F, Pacelli C, Audrito V, Provero P, Medico E, Chiabrando D, Porporato PE, Cancelliere C, Bardelli A, Trusolino L, Capitanio N, Deaglio S, Altruda F, Pinton P, Cardaci S, Riganti C, and Tolosano E
- Subjects
- Animals, Biological Transport, Caco-2 Cells, Heme metabolism, Humans, Membrane Transport Proteins metabolism, Mice, Inbred C57BL, Mice, SCID, Receptors, Virus metabolism, Xenograft Model Antitumor Assays, Mice, Citric Acid Cycle, Heme biosynthesis, Oxidative Phosphorylation
- Abstract
Heme is an iron-containing porphyrin of vital importance for cell energetic metabolism. High rates of heme synthesis are commonly observed in proliferating cells. Moreover, the cell-surface heme exporter feline leukemia virus subgroup C receptor 1a (FLVCR1a) is overexpressed in several tumor types. However, the reasons why heme synthesis and export are enhanced in highly proliferating cells remain unknown. Here, we illustrate a functional axis between heme synthesis and heme export: heme efflux through the plasma membrane sustains heme synthesis, and implementation of the two processes down-modulates the tricarboxylic acid (TCA) cycle flux and oxidative phosphorylation. Conversely, inhibition of heme export reduces heme synthesis and promotes the TCA cycle fueling and flux as well as oxidative phosphorylation. These data indicate that the heme synthesis-export system modulates the TCA cycle and oxidative metabolism and provide a mechanistic basis for the observation that both processes are enhanced in cells with high-energy demand., Competing Interests: Declaration of interests A.B. is a founder and a shareholder of Neophore; is an advisory board member for Roche, Inivata, Neophore, and Phoremost; has received grant support from AstraZeneca. L.T. receives research grants from Symphogen, Servier, Pfizer, Menarini, Merck KGaA, and Merus; and is in the speakers’ bureau of Eli Lilly, AstraZeneca, and Merck KGaA. E.T., V.F., A.L.A, S.P., and D.C. hold a pending patent application related to this work. The other authors declare no competing interests., (Copyright © 2021 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
24. Metabolic Aspects of Anthracycline Cardiotoxicity.
- Author
-
Russo M, Della Sala A, Tocchetti CG, Porporato PE, and Ghigo A
- Subjects
- Anthracyclines therapeutic use, Antineoplastic Agents therapeutic use, Autophagy, Biomarkers, Cell Survival, Disease Susceptibility, Fatty Acids metabolism, Glycolysis, Humans, Insulin Resistance, Iron metabolism, Myocardium metabolism, Myocardium pathology, Myocytes, Cardiac drug effects, Myocytes, Cardiac metabolism, Neoplasms drug therapy, Oxidation-Reduction, Anthracyclines adverse effects, Antineoplastic Agents adverse effects, Cardiotoxicity etiology, Cardiotoxicity metabolism, Neoplasms complications
- Abstract
Opinion Statement: Heart failure (HF) is increasingly recognized as the major complication of chemotherapy regimens. Despite the development of modern targeted therapies such as monoclonal antibodies, doxorubicin (DOXO), one of the most cardiotoxic anticancer agents, still remains the treatment of choice for several solid and hematological tumors. The insurgence of cardiotoxicity represents the major limitation to the clinical use of this potent anticancer drug. At the molecular level, cardiac side effects of DOXO have been associated to mitochondrial dysfunction, DNA damage, impairment of iron metabolism, apoptosis, and autophagy dysregulation. On these bases, the antioxidant and iron chelator molecule, dexrazoxane, currently represents the unique FDA-approved cardioprotectant for patients treated with anthracyclines.A less explored area of research concerns the impact of DOXO on cardiac metabolism. Recent metabolomic studies highlight the possibility that cardiac metabolic alterations may critically contribute to the development of DOXO cardiotoxicity. Among these, the impairment of oxidative phosphorylation and the persistent activation of glycolysis, which are commonly observed in response to DOXO treatment, may undermine the ability of cardiomyocytes to meet the energy demand, eventually leading to energetic failure. Moreover, increasing evidence links DOXO cardiotoxicity to imbalanced insulin signaling and to cardiac insulin resistance. Although anti-diabetic drugs, such as empagliflozin and metformin, have shown interesting cardioprotective effects in vitro and in vivo in different models of heart failure, their mechanism of action is unclear, and their use for the treatment of DOXO cardiotoxicity is still unexplored.This review article aims at summarizing current evidence of the metabolic derangements induced by DOXO and at providing speculations on how key players of cardiac metabolism could be pharmacologically targeted to prevent or cure DOXO cardiomyopathy.
- Published
- 2021
- Full Text
- View/download PDF
25. Inflammation-induced cholestasis in cancer cachexia.
- Author
-
Thibaut MM, Sboarina M, Roumain M, Pötgens SA, Neyrinck AM, Destrée F, Gillard J, Leclercq IA, Dachy G, Demoulin JB, Tailleux A, Lestavel S, Rastelli M, Everard A, Cani PD, Porporato PE, Loumaye A, Thissen JP, Muccioli GG, Delzenne NM, and Bindels LB
- Subjects
- Animals, Cytokines, Humans, Mice, Cachexia etiology, Cholestasis etiology, Inflammation complications, Neoplasms complications
- Abstract
Background: Cancer cachexia is a debilitating metabolic syndrome contributing to cancer death. Organs other than the muscle may contribute to the pathogenesis of cancer cachexia. This work explores new mechanisms underlying hepatic alterations in cancer cachexia., Methods: We used transcriptomics to reveal the hepatic gene expression profile in the colon carcinoma 26 cachectic mouse model. We performed bile acid, tissue mRNA, histological, biochemical, and western blot analyses. Two interventional studies were performed using a neutralizing interleukin 6 antibody and a bile acid sequestrant, cholestyramine. Our findings were evaluated in a cohort of 94 colorectal cancer patients with or without cachexia (43/51)., Results: In colon carcinoma 26 cachectic mice, we discovered alterations in five inflammatory pathways as well as in other pathways, including bile acid metabolism, fatty acid metabolism, and xenobiotic metabolism (normalized enrichment scores of -1.97, -2.16, and -1.34, respectively; all Padj < 0.05). The hepatobiliary transport system was deeply impaired in cachectic mice, leading to increased systemic and hepatic bile acid levels (+1512 ± 511.6 pmol/mg, P = 0.01) and increased hepatic inflammatory cytokines and neutrophil recruitment to the liver of cachectic mice (+43.36 ± 16.01 neutrophils per square millimetre, P = 0.001). Adaptive mechanisms were set up to counteract this bile acid accumulation by repressing bile acid synthesis and by enhancing alternative routes of basolateral bile acid efflux. Targeting bile acids using cholestyramine reduced hepatic inflammation, without affecting the hepatobiliary transporters (e.g. tumour necrosis factor α signalling via NFκB and inflammatory response pathways, normalized enrichment scores of -1.44 and -1.36, all Padj < 0.05). Reducing interleukin 6 levels counteracted the change in expression of genes involved in the hepatobiliary transport, bile acid synthesis, and inflammation. Serum bile acid levels were increased in cachectic vs. non-cachectic cancer patients (e.g. total bile acids, +5.409 ± 1.834 μM, P = 0.026) and were strongly correlated to systemic inflammation (taurochenodeoxycholic acid and C-reactive protein: ρ = 0.36, Padj = 0.017)., Conclusions: We show alterations in bile acid metabolism and hepatobiliary secretion in cancer cachexia. In this context, we demonstrate the contribution of systemic inflammation to the impairment of the hepatobiliary transport system and the role played by bile acids in the hepatic inflammation. This work paves the way to a better understanding of the role of the liver in cancer cachexia., (© 2020 The Authors. Journal of Cachexia, Sarcopenia and Muscle published by John Wiley & Sons Ltd on behalf of Society on Sarcopenia, Cachexia and Wasting Disorders.)
- Published
- 2021
- Full Text
- View/download PDF
26. Understanding the common mechanisms of heart and skeletal muscle wasting in cancer cachexia.
- Author
-
Rausch V, Sala V, Penna F, Porporato PE, and Ghigo A
- Abstract
Cachexia is a severe complication of cancer that adversely affects the course of the disease, with currently no effective treatments. It is characterized by a progressive atrophy of skeletal muscle and adipose tissue, resulting in weight loss, a reduced quality of life, and a shortened life expectancy. Although the cachectic condition primarily affects the skeletal muscle, a tissue that accounts for ~40% of total body weight, cachexia is considered a multi-organ disease that involves different tissues and organs, among which the cardiac muscle stands out for its relevance. Patients with cancer often experience severe cardiac abnormalities and manifest symptoms that are indicative of chronic heart failure, including fatigue, shortness of breath, and impaired exercise tolerance. Furthermore, cardiovascular complications are among the major causes of death in cancer patients who experienced cachexia. The lack of effective treatments for cancer cachexia underscores the need to improve our understanding of the underlying mechanisms. Increasing evidence links the wasting of the cardiac and skeletal muscles to metabolic alterations, primarily increased energy expenditure, and to increased proteolysis, ensuing from activation of the major proteolytic machineries of the cell, including ubiquitin-dependent proteolysis and autophagy. This review aims at providing an overview of the key mechanisms of cancer cachexia, with a major focus on those that are shared by the skeletal and cardiac muscles.
- Published
- 2021
- Full Text
- View/download PDF
27. Tumour acidosis evaluated in vivo by MRI-CEST pH imaging reveals breast cancer metastatic potential.
- Author
-
Anemone A, Consolino L, Conti L, Irrera P, Hsu MY, Villano D, Dastrù W, Porporato PE, Cavallo F, and Longo DL
- Subjects
- Animals, Cell Line, Tumor, Female, Hydrogen-Ion Concentration, Magnetic Resonance Imaging methods, Mice, Mice, Inbred BALB C, Breast Neoplasms chemistry, Breast Neoplasms pathology, Neoplasm Invasiveness pathology
- Abstract
Background: Tumour acidosis is considered to play a central role in promoting cancer invasion and migration, but few studies have investigated in vivo how tumour pH correlates with cancer invasion. This study aims to determine in vivo whether tumour acidity is associated with cancer metastatic potential., Methods: Breast cancer cell lines with different metastatic potentials have been characterised for several markers of aggressiveness and invasiveness. Murine tumour models have been developed and assessed for lung metastases and tumour acidosis has been assessed in vivo by a magnetic resonance imaging-based chemical exchange saturation transfer (CEST) pH imaging approach., Results: The higher metastatic potential of 4T1 and TS/A primary tumours, in comparison to the less aggressive TUBO and BALB-neuT ones, was confirmed by the highest expression of cancer cell stem markers (CD44
+ CD24- ), highlighting their propensity to migrate and invade, coinciding with the measurement obtained by in vitro assays. MRI-CEST pH imaging successfully discriminated the more aggressive 4T1 and TS/A tumours that displayed a more acidic pH. Moreover, the observed higher tumour acidity was significantly correlated with an increased number of lung metastases., Conclusions: The findings of this study indicate that the extracellular acidification is associated with the metastatic potential.- Published
- 2021
- Full Text
- View/download PDF
28. Iron: An Essential Element of Cancer Metabolism.
- Author
-
Hsu MY, Mina E, Roetto A, and Porporato PE
- Subjects
- Animals, Antioxidants metabolism, Cell Proliferation physiology, Homeostasis physiology, Humans, Oxidation-Reduction, Reactive Oxygen Species metabolism, Iron metabolism, Neoplasms metabolism
- Abstract
Cancer cells undergo considerable metabolic changes to foster uncontrolled proliferation in a hostile environment characterized by nutrient deprivation, poor vascularization and immune infiltration. While metabolic reprogramming has been recognized as a hallmark of cancer, the role of micronutrients in shaping these adaptations remains scarcely investigated. In particular, the broad electron-transferring abilities of iron make it a versatile cofactor that is involved in a myriad of biochemical reactions vital to cellular homeostasis, including cell respiration and DNA replication. In cancer patients, systemic iron metabolism is commonly altered. Moreover, cancer cells deploy diverse mechanisms to increase iron bioavailability to fuel tumor growth. Although iron itself can readily participate in redox reactions enabling vital processes, its reactivity also gives rise to reactive oxygen species (ROS). Hence, cancer cells further rely on antioxidant mechanisms to withstand such stress. The present review provides an overview of the common alterations of iron metabolism occurring in cancer and the mechanisms through which iron promotes tumor growth.
- Published
- 2020
- Full Text
- View/download PDF
29. Cachexia, a Systemic Disease beyond Muscle Atrophy.
- Author
-
Wyart E, Bindels LB, Mina E, Menga A, Stanga S, and Porporato PE
- Subjects
- Humans, Cachexia metabolism, Cachexia pathology, Cachexia therapy, Muscle, Skeletal metabolism, Muscle, Skeletal pathology, Muscular Atrophy metabolism, Muscular Atrophy pathology, Muscular Atrophy therapy, Quality of Life
- Abstract
Cachexia is a complication of dismal prognosis, which often represents the last step of several chronic diseases. For this reason, the comprehension of the molecular drivers of such a condition is crucial for the development of management approaches. Importantly, cachexia is a syndrome affecting various organs, which often results in systemic complications. To date, the majority of the research on cachexia has been focused on skeletal muscle, muscle atrophy being a pivotal cause of weight loss and the major feature associated with the steep reduction in quality of life. Nevertheless, defining the impact of cachexia on other organs is essential to properly comprehend the complexity of such a condition and potentially develop novel therapeutic approaches.
- Published
- 2020
- Full Text
- View/download PDF
30. Editorial: Metabolism Meets Function: Untangling the Cross-Talk Between Signaling and Metabolism.
- Author
-
Castegna A, McVicar DW, Campanella A, Palmieri EM, Menga A, and Porporato PE
- Published
- 2020
- Full Text
- View/download PDF
31. Glufosinate constrains synchronous and metachronous metastasis by promoting anti-tumor macrophages.
- Author
-
Menga A, Serra M, Todisco S, Riera-Domingo C, Ammarah U, Ehling M, Palmieri EM, Di Noia MA, Gissi R, Favia M, Pierri CL, Porporato PE, Castegna A, and Mazzone M
- Subjects
- Aminobutyrates, Animals, Female, Humans, Inflammation Mediators, Mice, Breast Neoplasms, Macrophages
- Abstract
Glutamine synthetase (GS) generates glutamine from glutamate and controls the release of inflammatory mediators. In macrophages, GS activity, driven by IL10, associates to the acquisition of M2-like functions. Conditional deletion of GS in macrophages inhibits metastasis by boosting the formation of anti-tumor, M1-like, tumor-associated macrophages (TAMs). From this basis, we evaluated the pharmacological potential of GS inhibitors in targeting metastasis, identifying glufosinate as a specific human GS inhibitor. Glufosinate was tested in both cultured macrophages and on mice bearing metastatic lung, skin and breast cancer. We found that glufosinate rewires macrophages toward an M1-like phenotype both at the primary tumor and metastatic site, countering immunosuppression and promoting vessel sprouting. This was also accompanied to a reduction in cancer cell intravasation and extravasation, leading to synchronous and metachronous metastasis growth inhibition, but no effects on primary tumor growth. Glufosinate treatment was well-tolerated, without liver and brain toxicity, nor hematopoietic defects. These results identify GS as a druggable enzyme to rewire macrophage functions and highlight the potential of targeting metabolic checkpoints in macrophages to treat cancer metastasis., (© 2020 The Authors. Published under the terms of the CC BY 4.0 license.)
- Published
- 2020
- Full Text
- View/download PDF
32. An EPR Study Using Cyclic Hydroxylamines To Assess The Level of Mitochondrial ROS in Superinvasive Cancer Cells.
- Author
-
Scheinok S, Capeloa T, Porporato PE, Sonveaux P, and Gallez B
- Subjects
- Cell Line, Tumor, Electron Spin Resonance Spectroscopy, Female, Humans, Hydroxylamines chemistry, Neoplasm Invasiveness, Organophosphorus Compounds chemistry, Piperidines chemistry, Superoxide Dismutase metabolism, Superoxides analysis, Mitochondria metabolism, Neoplasms metabolism, Reactive Oxygen Species metabolism, Superoxide Dismutase genetics
- Abstract
It has been proposed that a mitochondrial switch involving a high mitochondrial superoxide production is associated with cancer metastasis. We here report an EPR analysis of ROS production using cyclic hydroxylamines in superinvasive SiHa-F3 compared with less invasive SiHa wild-type human cervix cancer cells. Using the CMH probe, no significant difference was observed in the overall level of ROS between SiHa and SiHa-F3 cells. However, using mitochondria-targeted cyclic hydroxylamine probe mitoTEMPO-H, we detected a significantly higher mitochondrial ROS content in SiHa-F3 compared with the wild-type SiHa cells. To investigate the nature of mitochondrial ROS, we overexpressed superoxide dismutase 2, a SOD isoform exclusively localized in mitochondria, in SiHa-F3 superinvasive cells. A significantly lower signal was detected in SiHa-F3 cells overexpressing SOD2 compared with SiHa-F3. Despite some limitations discussed in the paper, our EPR results suggest that mitochondrial ROS (at least partly superoxide) are produced to a larger extent in superinvasive cancer cells compared with less invasive wild-type cancer cells.
- Published
- 2020
- Full Text
- View/download PDF
33. Monocarboxylate transporters in cancer.
- Author
-
Payen VL, Mina E, Van Hée VF, Porporato PE, and Sonveaux P
- Subjects
- Animals, Citric Acid Cycle genetics, Energy Metabolism genetics, Glucose metabolism, Humans, Lactic Acid metabolism, Metabolic Networks and Pathways genetics, Mice, Mice, Knockout, Neoplasms genetics, Neoplasms pathology, Monocarboxylic Acid Transporters genetics, Muscle Proteins genetics, Neoplasms metabolism, Receptors, G-Protein-Coupled genetics, Symporters genetics
- Abstract
Background: Tumors are highly plastic metabolic entities composed of cancer and host cells that can adopt different metabolic phenotypes. For energy production, cancer cells may use 4 main fuels that are shuttled in 5 different metabolic pathways. Glucose fuels glycolysis that can be coupled to the tricarboxylic acid (TCA) cycle and oxidative phosphorylation (OXPHOS) in oxidative cancer cells or to lactic fermentation in proliferating and in hypoxic cancer cells. Lipids fuel lipolysis, glutamine fuels glutaminolysis, and lactate fuels the oxidative pathway of lactate, all of which are coupled to the TCA cycle and OXPHOS for energy production. This review focuses on the latter metabolic pathway., Scope of Review: Lactate, which is prominently produced by glycolytic cells in tumors, was only recently recognized as a major fuel for oxidative cancer cells and as a signaling agent. Its exchanges across membranes are gated by monocarboxylate transporters MCT1-4. This review summarizes the current knowledge about MCT structure, regulation and functions in cancer, with a specific focus on lactate metabolism, lactate-induced angiogenesis and MCT-dependent cancer metastasis. It also describes lactate signaling via cell surface lactate receptor GPR81., Major Conclusions: Lactate and MCTs, especially MCT1 and MCT4, are important contributors to tumor aggressiveness. Analyses of MCT-deficient (MCT
+/- and MCT-/- ) animals and (MCT-mutated) humans indicate that they are druggable, with MCT1 inhibitors being in advanced development phase and MCT4 inhibitors still in the discovery phase. Imaging lactate fluxes non-invasively using a lactate tracer for positron emission tomography would further help to identify responders to the treatments., (Copyright © 2019 The Authors. Published by Elsevier GmbH.. All rights reserved.)- Published
- 2020
- Full Text
- View/download PDF
34. Annual Meeting of the International Society of Cancer Metabolism (ISCaM): Metabolic Adaptations and Targets in Cancer.
- Author
-
Avnet S, Baldini N, Brisson L, Pedersen SF, Porporato PE, Sonveaux P, Szabadkai G, and Pastorekova S
- Abstract
The metabolism of cancer cells differs from that of their normal counterparts in a spectrum of attributes, including imbalances in diverse metabolic arms and pathways, metabolic plasticity and extent of adaptive responses, levels, and activities of metabolic enzymes and their upstream regulators and abnormal fluxes of metabolic intermediates and products. These attributes endow cancer cells with the ability to survive stressors of the tumor microenvironment and enable them to landscape and exploit the host terrain, thereby facilitating cancer progression and therapy resistance. Understanding the molecular and physiological principles of cancer metabolism is one of the key prerequisites for the development of better anticancer treatments. Therefore, various aspects of cancer metabolism were addressed at the 5th annual meeting of the International Society of Cancer Metabolism (ISCaM) in Bratislava, Slovakia, on October 17-20, 2018. The meeting presentations and discussions were traditionally focused on mechanistic, translational, and clinical characteristics of metabolism and pH control in cancer, at the level of molecular pathways, cells, tissues, and organisms. In order to reflect major healthcare challenges of the current era, ISCaM has extended its scope to metabolic disorders contributing to cancer, as well as to opportunities for their prevention, intervention, and therapeutic targeting., (Copyright © 2019 Avnet, Baldini, Brisson, Pedersen, Porporato, Sonveaux, Szabadkai and Pastorekova.)
- Published
- 2019
- Full Text
- View/download PDF
35. Pro- and antitumor effects of mitochondrial reactive oxygen species.
- Author
-
Payen VL, Zampieri LX, Porporato PE, and Sonveaux P
- Subjects
- Animals, Antioxidants metabolism, Carcinogenesis metabolism, Carcinogenesis pathology, Humans, Mitochondria pathology, Neoplasms pathology, Oxidative Phosphorylation, Mitochondria metabolism, Neoplasms metabolism, Reactive Oxygen Species metabolism
- Abstract
In cancer, mitochondrial functions are commonly altered. Directly involved in metabolic reprogramming, mitochondrial plasticity confers to cancer cells a high degree of adaptability to a wide range of stresses and to the harsh tumor microenvironment. Lack of nutrients or oxygen caused by altered perfusion, metabolic needs of proliferating cells, co-option of the microenvironment, control of the immune system, cell migration and metastasis, and evasion of exogenous stress (e.g., chemotherapy) are all, at least in part, influenced by mitochondria. Mitochondria are undoubtedly one of the key contributors to cancer development and progression. Understanding their protumoral (dys)functions may pave the way to therapeutic strategies capable of turning them into innocent entities. Here, we will focus on the production and detoxification of mitochondrial reactive oxygen species (mtROS), on their impact on tumorigenesis (genetic, prosurvival, and microenvironmental effects and their involvement in autophagy), and on tumor metastasis. We will also summarize the latest therapeutic approaches involving mtROS.
- Published
- 2019
- Full Text
- View/download PDF
36. Assessing Metabolic Dysregulation in Muscle During Cachexia.
- Author
-
Hsu MY, Porporato PE, and Wyart E
- Subjects
- Humans, Mitochondria metabolism, Muscle Fibers, Skeletal metabolism, Muscle, Skeletal pathology, Muscular Atrophy, Neoplasms metabolism, Oxygen metabolism, Cachexia etiology, Cachexia metabolism, Energy Metabolism, Muscle, Skeletal metabolism, Neoplasms complications
- Abstract
Cancer cachexia is a metabolic disease characterized by a negative energy balance associated with systemic weight loss and poor quality of life.In particular, skeletal muscle, which represents almost 50% of the total body mass, is strongly affected, and metabolic alterations therein (e.g., insulin resistance and mitochondrial dysfunction) can eventually support tumor growth by facilitating nutrient scavenging by the growing mass. Interestingly, metabolic interventions on wasting muscle have been proven to be protective, advocating for the importance of metabolic regulation in the wasting muscle.Here, we will briefly define the current knowledge of metabolic regulation in cachexia and provide a protocol to grow and differentiate in vitro myotubes for the assessment of mitochondrial metabolism during cachexia.
- Published
- 2019
- Full Text
- View/download PDF
37. Annual Meeting of the International Society of Cancer Metabolism (ISCaM): Cancer Metabolism.
- Author
-
Avnet S, Baldini N, Brisson L, De Milito A, Otto AM, Pastoreková S, Porporato PE, Szabadkai G, and Sonveaux P
- Abstract
Tumors are metabolic entities wherein cancer cells adapt their metabolism to their oncogenic agenda and microenvironmental influences. Metabolically different cancer cell subpopulations collaborate to optimize nutrient delivery with respect to immediate bioenergetic and biosynthetic needs. They can also metabolically exploit host cells. These metabolic networks are directly linked with cancer progression, treatment, resistance, and relapse. Conversely, metabolic alterations in cancer are exploited for anticancer therapy, imaging, and stratification for personalized treatments. These topics were addressed at the 4th annual meeting of the International Society of Cancer Metabolism (ISCaM) in Bertinoro, Italy, on 19-21 October 2017.
- Published
- 2018
- Full Text
- View/download PDF
38. Phosphoinositide 3-Kinase Gamma Inhibition Protects From Anthracycline Cardiotoxicity and Reduces Tumor Growth.
- Author
-
Li M, Sala V, De Santis MC, Cimino J, Cappello P, Pianca N, Di Bona A, Margaria JP, Martini M, Lazzarini E, Pirozzi F, Rossi L, Franco I, Bornbaum J, Heger J, Rohrbach S, Perino A, Tocchetti CG, Lima BHF, Teixeira MM, Porporato PE, Schulz R, Angelini A, Sandri M, Ameri P, Sciarretta S, Lima-Júnior RCP, Mongillo M, Zaglia T, Morello F, Novelli F, Hirsch E, and Ghigo A
- Subjects
- Animals, Antibiotics, Antineoplastic toxicity, Autophagy-Related Proteins genetics, Autophagy-Related Proteins metabolism, Breast Neoplasms enzymology, Breast Neoplasms genetics, Breast Neoplasms pathology, Cardiotoxicity, Class Ib Phosphatidylinositol 3-Kinase genetics, Class Ib Phosphatidylinositol 3-Kinase metabolism, Cytoprotection, Disease Models, Animal, Doxorubicin toxicity, Female, Genes, erbB-2, Heart Diseases chemically induced, Heart Diseases enzymology, Heart Diseases pathology, Mice, Inbred BALB C, Mice, Transgenic, Mutation, Myocytes, Cardiac enzymology, Myocytes, Cardiac pathology, Toll-Like Receptor 9 genetics, Toll-Like Receptor 9 metabolism, Antibiotics, Antineoplastic pharmacology, Autophagy drug effects, Breast Neoplasms drug therapy, Doxorubicin pharmacology, Heart Diseases prevention & control, Myocytes, Cardiac drug effects, Phosphoinositide-3 Kinase Inhibitors, Protein Kinase Inhibitors pharmacology, Quinoxalines pharmacology, Thiazolidinediones pharmacology, Tumor Burden drug effects
- Abstract
Background: Anthracyclines, such as doxorubicin (DOX), are potent anticancer agents for the treatment of solid tumors and hematologic malignancies. However, their clinical use is hampered by cardiotoxicity. This study sought to investigate the role of phosphoinositide 3-kinase γ (PI3Kγ) in DOX-induced cardiotoxicity and the potential cardioprotective and anticancer effects of PI3Kγ inhibition., Methods: Mice expressing a kinase-inactive PI3Kγ or receiving PI3Kγ-selective inhibitors were subjected to chronic DOX treatment. Cardiac function was analyzed by echocardiography, and DOX-mediated signaling was assessed in whole hearts or isolated cardiomyocytes. The dual cardioprotective and antitumor action of PI3Kγ inhibition was assessed in mouse mammary tumor models., Results: PI3Kγ kinase-dead mice showed preserved cardiac function after chronic low-dose DOX treatment and were protected against DOX-induced cardiotoxicity. The beneficial effects of PI3Kγ inhibition were causally linked to enhanced autophagic disposal of DOX-damaged mitochondria. Consistently, either pharmacological or genetic blockade of autophagy in vivo abrogated the resistance of PI3Kγ kinase-dead mice to DOX cardiotoxicity. Mechanistically, PI3Kγ was triggered in DOX-treated hearts, downstream of Toll-like receptor 9, by the mitochondrial DNA released by injured organelles and contained in autolysosomes. This autolysosomal PI3Kγ/Akt/mTOR/Ulk1 signaling provided maladaptive feedback inhibition of autophagy. PI3Kγ blockade in models of mammary gland tumors prevented DOX-induced cardiac dysfunction and concomitantly synergized with the antitumor action of DOX by unleashing anticancer immunity., Conclusions: Blockade of PI3Kγ may provide a dual therapeutic advantage in cancer therapy by simultaneously preventing anthracyclines cardiotoxicity and reducing tumor growth.
- Published
- 2018
- Full Text
- View/download PDF
39. Long-term antigen exposure irreversibly modifies metabolic requirements for T cell function.
- Author
-
Bettonville M, d'Aria S, Weatherly K, Porporato PE, Zhang J, Bousbata S, Sonveaux P, and Braun MY
- Subjects
- Adenosine Triphosphate antagonists & inhibitors, Adenosine Triphosphate biosynthesis, Animals, Antibodies, Monoclonal pharmacology, Antimetabolites, Antineoplastic pharmacology, B7-H1 Antigen immunology, Cell Lineage drug effects, Cell Lineage genetics, DNA-Binding Proteins deficiency, DNA-Binding Proteins genetics, DNA-Binding Proteins immunology, Diazooxonorleucine pharmacology, Epoxy Compounds pharmacology, Gene Expression Profiling, Gene Expression Regulation, Glycolysis drug effects, Interferon-gamma antagonists & inhibitors, Interferon-gamma immunology, Interleukin Receptor Common gamma Subunit deficiency, Interleukin Receptor Common gamma Subunit genetics, Interleukin Receptor Common gamma Subunit immunology, Lymphocyte Activation, Male, Mice, Mice, Inbred BALB C, Mice, Knockout, Oligomycins pharmacology, Oxidative Phosphorylation drug effects, Programmed Cell Death 1 Receptor antagonists & inhibitors, Programmed Cell Death 1 Receptor immunology, Receptors, Antigen, T-Cell genetics, Receptors, Antigen, T-Cell immunology, Signal Transduction, T-Lymphocytes cytology, T-Lymphocytes drug effects, T-Lymphocytes transplantation, Transplantation, Homologous, B7-H1 Antigen genetics, Cell Lineage immunology, Interferon-gamma genetics, Programmed Cell Death 1 Receptor genetics, T-Lymphocytes immunology
- Abstract
Energy metabolism is essential for T cell function. However, how persistent antigenic stimulation affects T cell metabolism is unknown. Here, we report that long-term in vivo antigenic exposure induced a specific deficit in numerous metabolic enzymes. Accordingly, T cells exhibited low basal glycolytic flux and limited respiratory capacity. Strikingly, blockade of inhibitory receptor PD-1 stimulated the production of IFNγ in chronic T cells, but failed to shift their metabolism towards aerobic glycolysis, as observed in effector T cells. Instead, chronic T cells appeared to rely on oxidative phosphorylation (OXPHOS) and fatty acid oxidation (FAO) to produce ATP for IFNγ synthesis. Check-point blockade, however, increased mitochondrial production of superoxide and reduced viability and effector function. Thus, in the absence of a glycolytic switch, PD-1-mediated inhibition appears essential for limiting oxidative metabolism linked to effector function in chronic T cells, thereby promoting survival and functional fitness., Competing Interests: MB, Sd, KW, PP, JZ, SB, PS, MB No competing interests declared, (© 2018, Bettonville et al.)
- Published
- 2018
- Full Text
- View/download PDF
40. Signaling Pathways Regulating Redox Balance in Cancer Metabolism.
- Author
-
De Santis MC, Porporato PE, Martini M, and Morandi A
- Abstract
The interplay between rewiring tumor metabolism and oncogenic driver mutations is only beginning to be appreciated. Metabolic deregulation has been described for decades as a bystander effect of genomic aberrations. However, for the biology of malignant cells, metabolic reprogramming is essential to tackle a harsh environment, including nutrient deprivation, reactive oxygen species production, and oxygen withdrawal. Besides the well-investigated glycolytic metabolism, it is emerging that several other metabolic fluxes are relevant for tumorigenesis in supporting redox balance, most notably pentose phosphate pathway, folate, and mitochondrial metabolism. The relationship between metabolic rewiring and mutant genes is still unclear and, therefore, we will discuss how metabolic needs and oncogene mutations influence each other to satisfy cancer cells' demands. Mutations in oncogenes, i.e., PI3K/AKT/mTOR, RAS pathway, and MYC, and tumor suppressors, i.e., p53 and liver kinase B1, result in metabolic flexibility and may influence response to therapy. Since metabolic rewiring is shaped by oncogenic driver mutations, understanding how specific alterations in signaling pathways affect different metabolic fluxes will be instrumental for the development of novel targeted therapies. In the era of personalized medicine, the combination of driver mutations, metabolite levels, and tissue of origins will pave the way to innovative therapeutic interventions.
- Published
- 2018
- Full Text
- View/download PDF
41. Mitochondrial metabolism and cancer.
- Author
-
Porporato PE, Filigheddu N, Pedro JMB, Kroemer G, and Galluzzi L
- Subjects
- Animals, Antineoplastic Agents therapeutic use, Calcium metabolism, Glycolysis, Humans, Molecular Targeted Therapy, Neoplasms drug therapy, Oxidation-Reduction, Carcinogenesis immunology, Carcinogenesis metabolism, Cell Transformation, Neoplastic immunology, Cell Transformation, Neoplastic metabolism, Mitochondria immunology, Mitochondria metabolism, Neoplasms metabolism
- Abstract
Glycolysis has long been considered as the major metabolic process for energy production and anabolic growth in cancer cells. Although such a view has been instrumental for the development of powerful imaging tools that are still used in the clinics, it is now clear that mitochondria play a key role in oncogenesis. Besides exerting central bioenergetic functions, mitochondria provide indeed building blocks for tumor anabolism, control redox and calcium homeostasis, participate in transcriptional regulation, and govern cell death. Thus, mitochondria constitute promising targets for the development of novel anticancer agents. However, tumors arise, progress, and respond to therapy in the context of an intimate crosstalk with the host immune system, and many immunological functions rely on intact mitochondrial metabolism. Here, we review the cancer cell-intrinsic and cell-extrinsic mechanisms through which mitochondria influence all steps of oncogenesis, with a focus on the therapeutic potential of targeting mitochondrial metabolism for cancer therapy.
- Published
- 2018
- Full Text
- View/download PDF
42. Metabolic Alterations in a Slow-Paced Model of Pancreatic Cancer-Induced Wasting.
- Author
-
Wyart E, Reano S, Hsu MY, Longo DL, Li M, Hirsch E, Filigheddu N, Ghigo A, Riganti C, and Porporato PE
- Subjects
- Animals, Female, Mice, Mice, Inbred C57BL, Quality of Life, Cachexia physiopathology, Muscle, Skeletal physiopathology, Muscular Atrophy physiopathology, Pancreatic Neoplasms physiopathology
- Abstract
Cancer cachexia is a devastating syndrome occurring in the majority of terminally ill cancer patients. Notably, skeletal muscle atrophy is a consistent feature affecting the quality of life and prognosis. To date, limited therapeutic options are available, and research in the field is hampered by the lack of satisfactory models to study the complexity of wasting in cachexia-inducing tumors, such as pancreatic cancer. Moreover, currently used in vivo models are characterized by an explosive cachexia with a lethal wasting within few days, while pancreatic cancer patients might experience alterations long before the onset of overt wasting. In this work, we established and characterized a slow-paced model of pancreatic cancer-induced muscle wasting that promotes efficient muscular wasting in vitro and in vivo . Treatment with conditioned media from pancreatic cancer cells led to the induction of atrophy in vitro , while tumor-bearing mice presented a clear reduction in muscle mass and functionality. Intriguingly, several metabolic alterations in tumor-bearing mice were identified, paving the way for therapeutic interventions with drugs targeting metabolism.
- Published
- 2018
- Full Text
- View/download PDF
43. (+)-Catechin in a 1:2 Complex with Lysine Inhibits Cancer Cell Migration and Metastatic Take in Mice.
- Author
-
Payen VL, Porporato PE, Danhier P, Vazeille T, Blackman MCNM, May BH, Niebes P, and Sonveaux P
- Abstract
Metastasis is of dismal prognosis for cancer patients, but recent evidence in mouse models of cancer shows that metastasis prevention is a reachable clinical objective. These experiments indicate that altered mitochondrial activities are associated with the metastatic phenotype. Mitochondrial transfer from metastatic to non-metastatic cells can indeed transfer the metastatic phenotype, and metastatic progenitor cells differ from other cancer cells by a higher sublethal production of mitochondrial reactive oxygen species (ROS). Moreover, mitochondria-targeted antioxidants can prevent metastatic dissemination in mouse models of cancer. Comparatively, general antioxidants have unpredictable effects on cancer metastasis, most probably because they affect several cell types, several subcellular ROS production sites and, often, several endogenous oxidant species. Thus, targeting antioxidants to mitochondria could improve their antimetastatic activities, as previously exemplified with mitochondria-targeted mitoTEMPO and mitoQ that can prevent metastatic dissemination in cancer-bearing mice. Our objective in this study was to identify whether catechins, which are known to be potent antioxidants, can inhibit cancer cell migration in vitro and metastatic take in vivo . Comparative analysis of the response to epigallocatechin-3-gallate, (+)-catechin and (+)-catechin:lysine complexes revealed that, whereas all compounds had similar general antioxidant properties, (+)-catechin:lysine 1:2, but not epigallocatechin-3-gallate, can prevent metastatic take of melanoma cells to the lungs of mice. (+)-Catechin:lysine 1:2 possesses two net positive charges provided by lysines at physiological pH, which could provide high affinity for the negatively charged mitochondrial matrix. While this study reveals that (+)-catechin:lysine 1:2 has interesting antimetastatic effects, future experiments are needed to formally demonstrate the stability of the complex, its effective tropism for mitochondria and whether or not its activity can be globally attributed to its antioxidant activity at this precise subcellular location.
- Published
- 2017
- Full Text
- View/download PDF
44. MDA-MB-231 breast cancer cells fuel osteoclast metabolism and activity: A new rationale for the pathogenesis of osteolytic bone metastases.
- Author
-
Lemma S, Di Pompo G, Porporato PE, Sboarina M, Russell S, Gillies RJ, Baldini N, Sonveaux P, and Avnet S
- Subjects
- Animals, Cell Line, Tumor metabolism, Coumarins antagonists & inhibitors, Female, Glucose metabolism, Glycolysis, Humans, Lactates metabolism, MCF-7 Cells, Mice, Monocarboxylic Acid Transporters metabolism, Muscle Proteins metabolism, Osteoclasts pathology, Stromal Cells metabolism, Stromal Cells pathology, Symporters metabolism, Bone Neoplasms metabolism, Bone Neoplasms secondary, Breast Neoplasms metabolism, Breast Neoplasms pathology, Osteoclasts metabolism
- Abstract
Recent progress in dissecting the molecular paracrine circuits of cancer and stromal cells in bone metastases (BM) are offering new options to improve current merely palliative approach. The study of tumor-stroma metabolic interplay may further ameliorate this scenario. In this context, we demonstrated that highly glycolytic MDA-MB-231 cancer cells, that form osteolytic BM in vivo, release a large amount of lactate at a significantly higher level than MCF7 cells. Thus, we speculated that lactate released from carcinoma cells is uptaken and metabolically used by osteoclasts, the key players of osteolysis associated with BM. First, we demonstrated that the release of lactate at the bone site is mediated by monocarboxylate transporter 4 (MCT4), as revealed by immunostaining and MCT4 localization at the plasma membrane of tumor cells in mouse model of BM and in human tissue sections of BM. Then, we showed that in vitro lactate is uptaken by osteoclasts to be used as a fuel for the oxidative metabolism of osteoclasts, ultimately enhancing Type I collagen resorption. The passive transport of lactate into osteoclasts was mediated by MCT1: MCT1 expression is significantly upregulated during osteoclast differentiation and Type I collagen resorption is significantly impaired when osteoclasts are treated with 7-(N-benzyl-N-methylamino)-2-oxo-2H-chromene-3-carboxylic acid, an MCT-1 inhibitor. Together, these data demonstrate that lactate released by glycolytic breast carcinoma cells in the bone microenvironment promotes the formation of osteolytic lesions, and provide the rationale for further studies on the use of MCT1 targeting as a novel therapeutic approach in advanced cancer patients with BM., (Copyright © 2017 Elsevier B.V. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
45. Ffar2 expression regulates leukaemic cell growth in vivo.
- Author
-
Bindels LB, Porporato PE, Ducastel S, Sboarina M, Neyrinck AM, Dewulf EM, Feron O, Lestavel S, Cani PD, Staels B, Sonveaux P, and Delzenne NM
- Subjects
- Animals, Apoptosis, Biomarkers, Tumor metabolism, Female, Leukemia, Experimental metabolism, Male, Mice, Mice, Inbred BALB C, Mice, Inbred C57BL, Tumor Cells, Cultured, Cell Proliferation, Leukemia, Experimental pathology, Receptors, G-Protein-Coupled physiology
- Abstract
Background: Activation of free fatty acid receptor 2 (FFAR2) by microbiota-derived metabolites (e.g., propionate) reduces leukaemic cell proliferation in vitro. This study aims to test whether Ffar2 expression per se also influences leukaemia cell growth in vivo., Methods: Bcr-Abl-expressing BaF cells were used as a leukaemia model and the role of Ffar2 was evaluated in Balb/c mice after lentiviral shRNA transduction., Results: Our data formally establish that reduced leukaemic cell proliferation is associated with increased Ffar2 expression in vivo and in vitro. Going beyond association, we point out that decreasing Ffar2 expression fosters cancer cell growth in vitro and in vivo., Conclusions: Our data demonstrate the role of Ffar2 in the control of leukaemic cell proliferation in vivo and indicate that a modulation of Ffar2 expression through nutritional tools or pharmacological agents may constitute an attractive therapeutic approach to tackle leukaemia progression in humans.
- Published
- 2017
- Full Text
- View/download PDF
46. Monocarboxylate Transporter MCT1 Promotes Tumor Metastasis Independently of Its Activity as a Lactate Transporter.
- Author
-
Payen VL, Hsu MY, Rädecke KS, Wyart E, Vazeille T, Bouzin C, Porporato PE, and Sonveaux P
- Subjects
- Animals, Biological Transport, Cell Line, Tumor, Cell Movement physiology, Female, Humans, Mice, Mice, Inbred BALB C, Neoplasm Invasiveness, Neoplasm Metastasis, Mammary Neoplasms, Experimental metabolism, Mammary Neoplasms, Experimental pathology, Monocarboxylic Acid Transporters metabolism, Symporters metabolism, Uterine Cervical Neoplasms metabolism, Uterine Cervical Neoplasms pathology
- Abstract
Extracellular acidosis resulting from intense metabolic activities in tumors promotes cancer cell migration, invasion, and metastasis. Although host cells die at low extracellular pH, cancer cells resist, as they are well equipped with transporters and enzymes to regulate intracellular pH homeostasis. A low extracellular pH further activates proteolytic enzymes that remodel the extracellular matrix to facilitate cell migration and invasion. Monocarboxylate transporter MCT1 is a passive transporter of lactic acid that has attracted interest as a target for small-molecule drugs to prevent metastasis. In this study, we present evidence of a function for MCT1 in metastasis beyond its role as a transporter of lactic acid. MCT1 activates transcription factor NF-κB to promote cancer cell migration independently of MCT1 transporter activity. Although pharmacologic MCT1 inhibition did not modulate MCT1-dependent cancer cell migration, silencing or genetic deletion of MCT1 in vivo inhibited migration, invasion, and spontaneous metastasis. Our findings raise the possibility that pharmacologic inhibitors of MCT1-mediated lactic acid transport may not effectively prevent metastatic dissemination of cancer cells. Cancer Res; 77(20); 5591-601. ©2017 AACR ., (©2017 American Association for Cancer Research.)
- Published
- 2017
- Full Text
- View/download PDF
47. Presenilin 2-Dependent Maintenance of Mitochondrial Oxidative Capacity and Morphology.
- Author
-
Contino S, Porporato PE, Bird M, Marinangeli C, Opsomer R, Sonveaux P, Bontemps F, Dewachter I, Octave JN, Bertrand L, Stanga S, and Kienlen-Campard P
- Abstract
Mitochondrial dysfunction plays a pivotal role in the progression of Alzheimer's disease (AD), and yet the mechanisms underlying the impairment of mitochondrial function in AD remain elusive. Recent evidence suggested a role for Presenilins (PS1 or PS2) in mitochondrial function. Mutations of PSs, the catalytic subunits of the γ-secretase complex, are responsible for the majority of inherited AD cases (FAD). PSs were shown to be present in mitochondria and particularly enriched in mitochondria-associated membranes (MAM), where PS2 is involved in the calcium shuttling between mitochondria and the endoplasmic reticulum (ER). We investigated the precise contribution of PS1 and PS2 to the bioenergetics of the cell and to mitochondrial morphology in cell lines derived from wild type (PS+/+), PS1/2 double knock-out (PSdKO), PS2KO and PS1KO embryos. Our results showed a significant impairment in the respiratory capacity of PSdKO and PS2KO cells with reduction of basal oxygen consumption, oxygen utilization dedicated to ATP production and spare respiratory capacity. In line with these functional defects, we found a decrease in the expression of subunits responsible for mitochondrial oxidative phosphorylation (OXPHOS) associated with an altered morphology of the mitochondrial cristae. This OXPHOS disruption was accompanied by a reduction of the NAD
+ /NADH ratio. Still, neither ADP/ATP ratio nor mitochondrial membrane potential (ΔΨ) were affected, suggesting the existence of a compensatory mechanism for energetic balance. We observed indeed an increase in glycolytic flux in PSdKO and PS2KO cells. All these effects were truly dependent on PS2 since its stable re-expression in a PS2KO background led to a complete restoration of the parameters impaired in the absence of PS2. Our data clearly demonstrate here the crucial role of PS2 in mitochondrial function and cellular bioenergetics, pointing toward its peculiar role in the formation and integrity of the electron transport chain.- Published
- 2017
- Full Text
- View/download PDF
48. Cancer metabolism in space and time: Beyond the Warburg effect.
- Author
-
Danhier P, Bański P, Payen VL, Grasso D, Ippolito L, Sonveaux P, and Porporato PE
- Subjects
- Animals, Cell Death, Cell Division, Glycolysis, Humans, Metabolic Networks and Pathways, Mitochondria metabolism, Neoplasm Metastasis, Neoplastic Stem Cells metabolism, Stromal Cells metabolism, Energy Metabolism, Neoplasms metabolism
- Abstract
Altered metabolism in cancer cells is pivotal for tumor growth, most notably by providing energy, reducing equivalents and building blocks while several metabolites exert a signaling function promoting tumor growth and progression. A cancer tissue cannot be simply reduced to a bulk of proliferating cells. Tumors are indeed complex and dynamic structures where single cells can heterogeneously perform various biological activities with different metabolic requirements. Because tumors are composed of different types of cells with metabolic activities affected by different spatial and temporal contexts, it is important to address metabolism taking into account cellular and biological heterogeneity. In this review, we describe this heterogeneity also in metabolic fluxes, thus showing the relative contribution of different metabolic activities to tumor progression according to the cellular context. This article is part of a Special Issue entitled Mitochondria in Cancer, edited by Giuseppe Gasparre, Rodrigue Rossignol and Pierre Sonveaux., (Copyright © 2017 Elsevier B.V. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
49. Annual Meeting of the International Society of Cancer Metabolism (ISCaM): Metabolic Networks in Cancer.
- Author
-
Baldini N, De Milito A, Feron O, Gillies RJ, Michiels C, Otto AM, Pastoreková S, Pedersen SF, Porporato PE, Sonveaux P, Supuran CT, and Avnet S
- Abstract
Cancers are metabolic entities wherein cancer cells adapt their metabolism to their oncogenic agenda and microenvironmental influences. Metabolically different cancer cell subpopulations collaborate to optimize nutrient delivery with respect to immediate bioenergetic and biosynthetic needs. They can also metabolically exploit host cells. These metabolic networks are directly linked with cancer progression, treatment resistance and relapse. Conversely, metabolic alterations in cancer are exploited for anticancer therapy, imaging and personalized medicine. These topics were addressed at the 3rd annual meeting of the International Society of Cancer Metabolism (ISCaM) in Brussels, Belgium, on 26-29 October 2016.
- Published
- 2017
- Full Text
- View/download PDF
50. Multimodality Imaging Identifies Distinct Metabolic Profiles In Vitro and In Vivo.
- Author
-
Neveu MA, De Preter G, Marchand V, Bol A, Brender JR, Saito K, Kishimoto S, Porporato PE, Sonveaux P, Grégoire V, Feron O, Jordan BF, Krishna MC, and Gallez B
- Subjects
- Animals, Cell Line, Tumor, Disease Models, Animal, Electron Spin Resonance Spectroscopy, Glucose metabolism, Glycolysis, Heterografts, Humans, Magnetic Resonance Spectroscopy, Oxidation-Reduction, Oxygen metabolism, Phenotype, Positron Emission Tomography Computed Tomography, Metabolome, Multimodal Imaging, Neoplasms diagnostic imaging, Neoplasms metabolism
- Abstract
The study of alterations of tumor metabolism should allow the identification of new targets for innovative anticancer strategies. Metabolic alterations are generally established in vitro, and conclusions are often extrapolated to the in vivo situation without further tumor metabolic phenotyping. To highlight the key role of microenvironment on tumor metabolism, we studied the response of glycolytic and oxidative tumor models to metabolic modulations in vitro and in vivo. MDA-MB-231 and SiHa tumor models, characterized in vitro as glycolytic and oxidative, respectively, were studied. Theoretically, when passing from a hypoxic state to an oxygenated state, a Warburg phenotype should conserve a glycolytic metabolism, whereas an oxidative phenotype should switch from glycolytic to oxidative metabolism (Pasteur effect). This challenge was applied in vitro and in vivo to evaluate the impact of different oxic conditions on glucose metabolism.
18 F-fluorodeoxyglucose uptake, lactate production, tumor oxygenation, and metabolic fluxes were monitored in vivo using positron emission tomography, microdialysis, electron paramagnetic resonance imaging, and13 C-hyperpolarizated magnetic resonance spectroscopy, respectively. In vitro, MDA-MB-231 cells were glycolytic under both hypoxic and oxygenated conditions, whereas SiHa cells underwent a metabolic shift after reoxygenation. On the contrary, in vivo, the increase in tumor oxygenation (induced by carbogen challenge) led to a similar metabolic shift in glucose metabolism in both tumor models. The major discordance in metabolic patterns observed in vitro and in vivo highlights that any extrapolation of in vitro metabolic profiling to the in vivo situation should be taken cautiously and that metabolic phenotyping using molecular imaging is mandatory in vivo., (Copyright © 2016 The Authors. Published by Elsevier Inc. All rights reserved.)- Published
- 2016
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.