100 results on '"José M. Cuezva"'
Search Results
2. Reprogramming Oxidative Phosphorylation in Cancer: A Role for RNA-Binding Proteins
- Author
-
Pau B. Esparza-Moltó and José M. Cuezva
- Subjects
0301 basic medicine ,Cell signaling ,Physiology ,Cellular differentiation ,Clinical Biochemistry ,Oxidative phosphorylation ,Biology ,Mitochondrion ,medicine.disease_cause ,Biochemistry ,Gene Expression Regulation, Enzymologic ,Oxidative Phosphorylation ,03 medical and health sciences ,Neoplasms ,medicine ,Animals ,Humans ,Molecular Biology ,General Environmental Science ,030102 biochemistry & molecular biology ,ATP synthase ,RNA-Binding Proteins ,Cell Biology ,Mitochondria ,Cell biology ,Gene Expression Regulation, Neoplastic ,Cell Transformation, Neoplastic ,030104 developmental biology ,Organ Specificity ,Anaerobic glycolysis ,biology.protein ,General Earth and Planetary Sciences ,Disease Susceptibility ,Signal transduction ,Energy Metabolism ,Carcinogenesis ,Glycolysis - Abstract
Significance: Cancer is a major disease imposing high personal and economic burden draining large part of National Health Care and Research budgets worldwide. In the last decade, research in cancer has underscored the reprogramming of metabolism to an enhanced aerobic glycolysis as a major trait of the cancer phenotype with great potential for targeted therapy. Recent Advances: Mitochondria are essential organelles in metabolic reprogramming for controlling the production of biological energy through oxidative phosphorylation (OXPHOS) and the supply of metabolic precursors that sustain proliferation. In addition, mitochondria are critical hubs that integrate different signaling pathways that control cellular metabolism and cell fate. The mitochondrial ATP synthase plays a fundamental role in OXPHOS and cellular signaling. Critical Issues: This review overviews mitochondrial metabolism and OXPHOS, and the major changes reported in the expression and function of mitochondrial proteins of OXPHOS in oncogenesis and in cellular differentiation. We summarize the prominent role that RNA-binding proteins (RNABPs) play in the sorting and localized translation of nuclear-encoded mRNAs that help define the mitochondrial cell-type-specific phenotype. Moreover, we emphasize the mechanisms that contribute to restrain the activity and expression of the mitochondrial ATP synthase in carcinomas, and illustrate that the dysregulation of proteins that control energy metabolism correlates with patients' survival. Future Directions: Future research should elucidate the mechanisms and RNABPs that promote the specific alterations of the mitochondrial phenotype in carcinomas arising from different tissues with the final aim of developing new therapeutic strategies to treat cancer.
- Published
- 2020
- Full Text
- View/download PDF
3. Analysis of the metabolic proteome of lung adenocarcinomas by reverse-phase protein arrays (RPPA) emphasizes mitochondria as targets for therapy
- Author
-
Laura Torresano, Fulvio Santacatterina, Sonia Domínguez-Zorita, Cristina Nuevo-Tapioles, Alfonso Núñez-Salgado, Pau B. Esparza-Moltó, Lucía González-Llorente, Inés Romero-Carramiñana, Cristina Núñez de Arenas, Brenda Sánchez-Garrido, Laura Nájera, Clara Salas, Mariano Provencio, and José M. Cuezva
- Subjects
Cancer Research ,Molecular Biology - Abstract
Lung cancer is the leading cause of cancer-related death worldwide despite the success of therapies targeting oncogenic drivers and immune-checkpoint inhibitors. Although metabolic enzymes offer additional targets for therapy, the precise metabolic proteome of lung adenocarcinomas is unknown, hampering its clinical translation. Herein, we used Reverse Phase Protein Arrays to quantify the changes in enzymes of glycolysis, oxidation of pyruvate, fatty acid metabolism, oxidative phosphorylation, antioxidant response and protein oxidative damage in 128 tumors and paired non-tumor adjacent tissue of lung adenocarcinomas to profile the proteome of metabolism. Steady-state levels of mitochondrial proteins of fatty acid oxidation, oxidative phosphorylation and of the antioxidant response are independent predictors of survival and/or of disease recurrence in lung adenocarcinoma patients. Next, we addressed the mechanisms by which the overexpression of ATPase Inhibitory Factor 1, the physiological inhibitor of oxidative phosphorylation, which is an independent predictor of disease recurrence, prevents metastatic disease. We highlight that IF1 overexpression promotes a more vulnerable and less invasive phenotype in lung adenocarcinoma cells. Finally, and as proof of concept, the therapeutic potential of targeting fatty acid assimilation or oxidation in combination with an inhibitor of oxidative phosphorylation was studied in mice bearing lung adenocarcinomas. The results revealed that this therapeutic approach significantly extended the lifespan and provided better welfare to mice than cisplatin treatments, supporting mitochondrial activities as targets of therapy in lung adenocarcinoma patients.
- Published
- 2022
4. Exploiting the passenger ACO1-deficiency arising from 9p21 deletions to kill T-cell lymphoblastic neoplasia cells
- Author
-
José Luis Marín-Rubio, Konstantinos Stamatakis, Javier Santos, María Villa-Morales, José M. Cuezva, José Fernández-Piqueras, Eduardo Salido, Pilar Llamas, Iria Gonzalez-Vasconcellos, José Luis López-Lorenzo, Laura González-Sánchez, María Ángeles Cobos-Fernández, Irene Vázquez-Domínguez, Pilar López-Nieva, Instituto de Salud Carlos III, Ministerio de Economía y Competitividad (España), Ministerio de Ciencia e Innovación (España), Universidad Autónoma de Madrid, Asociación Española Contra el Cáncer, Fundación Ramón Areces, and Banco Santander
- Subjects
0301 basic medicine ,Cancer Research ,Skin Neoplasms ,Cell Survival ,T cell ,Mice, Nude ,Biology ,Precursor T-Cell Lymphoblastic Leukemia-Lymphoma ,Aconitase ,Mice ,03 medical and health sciences ,0302 clinical medicine ,CDKN2A ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Citrates ,Iron Regulatory Protein 1 ,Enzyme Inhibitors ,Melanoma ,Gene ,Cyclin-Dependent Kinase Inhibitor p16 ,Gene knockdown ,General Medicine ,medicine.disease ,030104 developmental biology ,medicine.anatomical_structure ,Drug Resistance, Neoplasm ,Cell culture ,030220 oncology & carcinogenesis ,Cancer research ,Heterografts ,Female ,Ectopic expression ,Chromosomes, Human, Pair 9 ,Gene Deletion - Abstract
Precursor T-cell lymphoblastic neoplasms are aggressive malignancies in need for more effective and specific therapeutic treatments. A significant fraction of these neoplasms harbor deletions on the locus 9p21, targeting the tumor suppressor CDKN2A but also deleting the aconitase 1 gene (ACO1), a neighboring housekeeping gene involved in cytoplasm and mitochondrial metabolism. Here we show that reducing the aconitase activity with fluorocitrate decreases the viability of T-cell lymphoblastic neoplasia cells in correlation to the differential aconitase expression. The consequences of the treatment were evidenced in vitro using T-cell lymphoblastic neoplasia cell lines exhibiting 9p21 deletions and variable levels of ACO1 expression or activity. Similar results were observed in melanoma cell lines, suggesting a true potential for fluorocitrate in different cancer types. Notably, ectopic expression of ACO1 alleviated the susceptibility of cell lines to fluorocitrate and, conversely, knockdown experiments increased susceptibility of resistant cell lines. These findings were confirmed in vivo on athymic nude-mice by using tumor xenografts derived from two T-cell lines with different levels of ACO1. Taken together, our results indicate that the non-targeted ACO1 deficiency induced by common deletions exerts a collateral cellular lethality that can be used as a novel therapeutic strategy in the treatment of several types of cancer., Instituto de Salud Carlos III (ACCI-CIBERER-17); Spanish Ministerio de Economía y Competitividad (SAF2015-70561-R; MINECO/FEDER, EU); Spanish Ministerio de Ciencia, Innovación y Universidades (RTI2018-093330-B-I00; MCIU/FEDER, EU); Universidad Autónoma de Madrid, Spain (B2017/BMD-3778; LINFOMAS-CM); Spanish Association Against Cancer (AECC, 2018; PROYE18054PIRI); Fundación Ramón Areces (CIVP19S7917); Institutional grants from Fundación Ramón Areces and Banco de Santander
- Published
- 2019
- Full Text
- View/download PDF
5. Changes in the Turnover of the Cellular Proteome during Metabolic Reprogramming: A Role for mtROS in Proteostasis
- Author
-
Ana García-Aguilar, José M. Cuezva, Inmaculada Martínez-Reyes, Ministerio de Economía y Competitividad (España), Instituto de Salud Carlos III, Fundación Ramón Areces, Consejo Superior de Investigaciones Científicas (España), García-Aguilar, Ana [0000-0003-1648-7143], Martínez-Reyes, Inmaculada [0000-0003-0479-1535], Cuezva, José M. [0000-0003-1118-248X], García-Aguilar, Ana, Martínez-Reyes, Inmaculada, and Cuezva, José M.
- Subjects
Mitochondrial ROS ,Proteome ,mTORC1 ,Protein degradation ,SILAC ,Biochemistry ,chemistry.chemical_compound ,Humans ,MitoQ ,Prolyl hydroxylases ,Chemistry ,Protein turnover ,Translation (biology) ,General Chemistry ,Cellular Reprogramming ,Cell Hypoxia ,Mitochondria ,Cell biology ,Proteostasis ,ATP synthase ,Signal transduction ,Energy Metabolism ,Reactive Oxygen Species ,Glycolysis ,Signal Transduction - Abstract
The role played by protein turnover in metabolic reprogramming is unknown. Herein, using a SILAC approach, we have studied the changes in the half-life of 266 proteins of energy metabolism and of translation during the metabolic switch induced by the prolyl hydroxylases inhibitor dimethyloxalylglycine (DMOG). DMOG induces HIF-1α expression and triggers the activation of glycolysis and the concurrent inhibition of mitochondrial respiration in colon cancer cells. Changes in the activity of energy provision pathways correlated with increased turnover rates of glycolytic enzymes and the stabilization of mitochondrial proteins. Moreover, reprogramming also stabilized the proteins of translation. The partial DMOG-mediated arrest of the synthesis of mitochondrial and translation proteins results from the inhibition of the mTORC1/p70SK/S6 signaling pathway. In contrast, DMOG stimulated the synthesis of glycolytic enzymes, emphasizing the opposite and differential regulation of the two pathways of energy provision. Addition of MitoQ, a mitochondrial reactive oxygen species (mtROS) scavenger, stabilized the turnover of cellular proteins similarly as when protein degradation is inhibited with leupeptin, a serine-protease inhibitor. Overall, the results show that the higher the activity of a pathway the lower is the half-life of the proteins involved and suggest a role for mtROS in cellular proteostasis. Data are available via ProteomeXchange with identifier PXD013482. ©, MINECO (SAF2013-41945-R and SAF2016-75916-R), CIBERER-ISCIII, and Fundación Ramón Areces, Spain, We acknowledge support by the CSIC Open Access Publication Initiative through its Unit of Information Resources for Research (URICI).
- Published
- 2019
- Full Text
- View/download PDF
6. Critical requirement of SOS1 RAS-GEF function for mitochondrial dynamics, metabolism, and redox homeostasis
- Author
-
Rósula García-Navas, Fernando C. Baltanás, P. Liceras-Boillos, Nuria Calzada, José M. Cuezva, Eugenio Santos, Carmela Gómez, Cristina Nuevo-Tapioles, UAM. Departamento de Biología Molecular, Instituto de Salud Carlos III, Junta de Castilla y León, Fundación Ramón Areces, European Commission, Universidad de Sevilla. Departamento de Fisiología Médica y Biofísica, European Commission (EC). Fondo Europeo de Desarrollo Regional (FEDER), Instituto de Salud Carlos III FIS PI19/00934, CIBERONC-CB16/12/00352, Gobierno regional de Castilla y Leon SA264P18-UIC 076, and Fundación Ramon Areces CIVP19A5942
- Subjects
0301 basic medicine ,Cancer Research ,Fission ,Molecular biology ,Activation ,Endogeny ,Oxidative phosphorylation ,Mitochondrion ,Biology ,medicine.disease_cause ,Mitochondrial Dynamics ,Article ,Induction ,03 medical and health sciences ,0302 clinical medicine ,Genetics ,medicine ,Homeostasis ,Glycolysis ,Fusion ,Pathways ,Cancer ,Cell-Proliferation ,Superoxide ,Metabolism ,Oncogenes ,Mass ,Biología y Biomedicina / Biología ,Electron transport chain ,Cell biology ,Oxidative Stress ,030104 developmental biology ,030220 oncology & carcinogenesis ,ras Guanine Nucleotide Exchange Factors ,Oxidative stress ,Intracellular ,Cell signalling - Abstract
© The Author(s) 2021., SOS1 ablation causes specific defective phenotypes in MEFs including increased levels of intracellular ROS. We showed that the mitochondria-targeted antioxidant MitoTEMPO restores normal endogenous ROS levels, suggesting predominant involvement of mitochondria in generation of this defective SOS1-dependent phenotype. The absence of SOS1 caused specific alterations of mitochondrial shape, mass, and dynamics accompanied by higher percentage of dysfunctional mitochondria and lower rates of electron transport in comparison to WT or SOS2-KO counterparts. SOS1-deficient MEFs also exhibited specific alterations of respiratory complexes and their assembly into mitochondrial supercomplexes and consistently reduced rates of respiration, glycolysis, and ATP production, together with distinctive patterns of substrate preference for oxidative energy metabolism and dependence on glucose for survival. RASless cells showed defective respiratory/metabolic phenotypes reminiscent of those of SOS1-deficient MEFs, suggesting that the mitochondrial defects of these cells are mechanistically linked to the absence of SOS1-GEF activity on cellular RAS targets. Our observations provide a direct mechanistic link between SOS1 and control of cellular oxidative stress and suggest that SOS1-mediated RAS activation is required for correct mitochondrial dynamics and function., This work was supported by grants from ISCIII-MCUI (FIS PI19/00934 and CIBERONCCB16/12/00352), Junta de Castilla y Leon (SA264P18-UIC 076), and Ramon Areces Foundation (CIVP19A5942). The research was co-financed by FEDER funds.
- Published
- 2021
7. Specific Effects of Trabectedin and Lurbinectedin on Human Macrophage Function and Fate-Novel Insights
- Author
-
Lisardo Boscá, Eduardo López-Collazo, José M. Cuezva, Adrián Povo-Retana, Adrian B Stremtan, Cristina Nuevo-Tapioles, José Manuel Molina-Guijarro, José Avendaño-Ortiz, Marina Mojena, Victoria Fernández-García, Ana Gómez-Sáez, Juan F. Martínez-Leal, Ministerio de Economía y Competitividad (España), Ministerio de Ciencia, Innovación y Universidades (España), Agencia Estatal de Investigación (España), Centro de Investigación Biomédica en Red Enfermedades Cardiovaculares (España), PharmaMar, Fundación Ramón Areces, Comunidad de Madrid, European Commission, and UAM. Departamento de Biología Molecular
- Subjects
Lurbinectedin ,0301 basic medicine ,Cell viability ,Cancer Research ,Macrophage ,lurbinectedin ,Apoptosis ,Tumor-associated macrophage ,macrophage ,lcsh:RC254-282 ,Calcium dynamics ,Article ,03 medical and health sciences ,tumor-associated macrophage ,0302 clinical medicine ,Immune system ,medicine ,Viability assay ,Caspase ,Trabectedin ,cell viability ,biology ,Respiration ,apoptosis ,calcium dynamics ,Biología y Biomedicina / Biología ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Mitochondria ,Immunosurveillance ,mitochondria ,ATP ,030104 developmental biology ,Oncology ,caspases ,Caspases ,030220 oncology & carcinogenesis ,trabectedin ,biology.protein ,Cancer research ,respiration ,medicine.drug - Abstract
© 2020 by the authors., Background: Tumor-associated macrophages (TAMs) play a crucial role in suppressing the immunosurveillance function of the immune system that prevents tumor growth. Indeed, macrophages can also be targeted by different chemotherapeutic agents improving the action over immune checkpoints to fight cancer. Here we describe the effect of trabectedin and lurbinectedin on human macrophage cell viability and function. Methods: Blood monocytes from healthy donors were differentiated into macrophages and exposed to different stimuli promoting functional polarization and differentiation into tumor-associated macrophages. Cells were challenged with the chemotherapeutic drugs and the effects on cell viability and function were analyzed. Results: Human macrophages exhibit at least two different profiles in response to these drugs. One-fourth of the blood donors assayed (164 individuals) were extremely sensitive to trabectedin and lurbinectedin, which promoted apoptotic cell death. Macrophages from other individuals retained viability but responded to the drugs increasing reactive oxygen production and showing a rapid intracellular calcium rise and a loss of mitochondrial oxygen consumption. Cell-membrane exposure of programmed-death ligand 1 (PD-L1) significantly decreased after treatment with therapeutic doses of these drugs, including changes in the gene expression profile of hypoxia-inducible factor 1 alpha (HIF-1α)-dependent genes, among other. Conclusions: The results provide evidence of additional onco-therapeutic actions for these drugs., This research was supported by the Ministerio Economía, Industria y Competitividad (SAF2017-82436-R/RTC2017-6283-1 and PID2019-108674RB-100), Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares (CB16/11/00222), PharmaMar (contract 164249), Fundación Ramón Areces (CIUP18A3864), Consorcio de Investigación en Red de la Comunidad de Madrid, S2017/BMD-3686 and Fondo Europeo de Desarrollo Regional.
- Published
- 2020
8. Tissue‐specific expression and post‐transcriptional regulation of the ATPase inhibitory factor 1 (IF1) in human and mouse tissues
- Author
-
José M. Cuezva, Laura Nájera, Laura Torresano, Pau B. Esparza-Moltó, Fulvio Santacatterina, Cristina Nuevo-Tapioles, and Margarita Chamorro
- Subjects
Male ,0301 basic medicine ,Oxidative phosphorylation ,Mitochondrion ,Biochemistry ,Cell Line ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Genetics ,Animals ,Humans ,Protein phosphorylation ,RNA, Messenger ,Phosphorylation ,RNA Processing, Post-Transcriptional ,Molecular Biology ,Post-transcriptional regulation ,Mice, Knockout ,chemistry.chemical_classification ,ATP synthase ,biology ,Proteins ,Cell biology ,Mice, Inbred C57BL ,030104 developmental biology ,Enzyme ,chemistry ,biology.protein ,Pentatricopeptide repeat ,Female ,030217 neurology & neurosurgery ,Biotechnology - Abstract
The ATPase inhibitory factor 1 (IF1) is an intrinsically disordered protein that regulates the activity of the mitochondrial ATP synthase. Phosphorylation of S39 in IF1 prevents it from binding to the enzyme and thus abolishes its inhibitory activity. Dysregulation of IF1 is linked to different human diseases, providing a relevant biomarker of cancer progression. However, the tissue content of IF1 relative to the abundance of the ATP synthase is unknown. In this study, we characterized the tissue-specific expression of IF1 in human and mouse tissues and quantitated the content of IF1 and of ATP synthase. We found relevant differences in IF1 expression between human and mouse tissues and found that in high-energy-demanding tissues, the molar content of IF1 exceeds that of the ATP synthase. In these tissues, a fraction of IF1 is bound to the enzyme, and the other fraction is phosphorylated and hence is unable to bind the enzyme. Post-transcriptional control accounts for most of the regulated expression of IF1, especially in mouse heart, where IF1 mRNA translation is repressed by the leucine-rich pentatricopeptide repeat containing protein. Overall, these findings enlighten the cellular biology of IF1 and pave the way to development of additional models that address its role in pathophysiology.-Esparza-Moltó, P. B., Nuevo-Tapioles, C., Chamorro, M., Nájera, L., Torresano, L., Santacatterina, F., Cuezva, J. M. Tissue-specific expression and post-transcriptional regulation of the ATPase inhibitory factor 1 (IF1) in human and mouse tissues.
- Published
- 2018
- Full Text
- View/download PDF
9. Metformin as an adjuvant to photodynamic therapy in resistant basal cell carcinoma cells
- Author
-
Cristina Nuevo-Tapioles, Edgar Abarca-Lachen, Salvador González, Pablo Delgado-Wicke, Marta Mascaraque, Tamara Gracia-Cazaña, Angeles Juarranz, José M. Cuezva, Yolanda Gilaberte, Instituto de Salud Carlos III, Ministerio de Ciencia, Innovación y Universidades (España), Ministerio de Economía y Competitividad (España), UAM. Departamento de Biología, and UAM. Departamento de Biología Molecular
- Subjects
0301 basic medicine ,Cancer Research ,Cell Survival ,Cellular differentiation ,Aerobic Glycolysis ,Protein Expression ,Resistance ,Terapia fotodinámica ,Oxidative phosphorylation ,PKM2 ,lcsh:RC254-282 ,Cell Drug Determination ,Article ,Photodynamic therapy ,03 medical and health sciences ,Carcinoma de células basales ,0302 clinical medicine ,Glucose Metabolism ,Resistencia ,G1 Phase Cell Cycle Checkpoint ,Metformina ,PI3K/AKT/mTOR pathway ,Chemistry ,Marcadores metabólicos ,Animal Experiment ,Biología y Biomedicina / Biología ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Warburg effect ,Metformin ,Animal Cell ,Cell Metabolism ,Metabolic markers ,Cancer Resistance ,030104 developmental biology ,Oncology ,Anaerobic glycolysis ,Cell culture ,030220 oncology & carcinogenesis ,Cancer cell ,Basal cell carcinoma ,Cancer research ,Célula - Abstract
© 2020 by the authors., Photodynamic Therapy (PDT) with methyl-aminolevulinate (MAL-PDT) is being used for the treatment of Basal Cell Carcinoma (BCC), although resistant cells may appear. Normal differentiated cells depend primarily on mitochondrial oxidative phosphorylation (OXPHOS) to generate energy, but cancer cells switch this metabolism to aerobic glycolysis (Warburg effect), influencing the response to therapies. We have analyzed the expression of metabolic markers (β-F1-ATPase/GAPDH (glyceraldehyde-3-phosphate dehydrogenase) ratio, pyruvate kinase M2 (PKM2), oxygen consume ratio, and lactate extracellular production) in the resistance to PDT of mouse BCC cell lines (named ASZ and CSZ, heterozygous for ptch1). We have also evaluated the ability of metformin (Metf), an antidiabetic type II compound that acts through inhibition of the AMP-activated protein kinase (AMPK)/mammalian target of rapamycin (mTOR) pathway to sensitize resistant cells to PDT. The results obtained indicated that resistant cells showed an aerobic glycolysis metabolism. The treatment with Metf induced arrest in the G0/G1 phase and a reduction in the lactate extracellular production in all cell lines. The addition of Metf to MAL-PDT improved the cytotoxic effect on parental and resistant cells, which was not dependent on the PS protoporphyrin IX (PpIX) production. After Metf + MAL-PDT treatment, activation of pAMPK was detected, suppressing the mTOR pathway in most of the cells. Enhanced PDT-response with Metf was also observed in ASZ tumors. In conclusion, Metf increased the response to MAL-PDT in murine BCC cells resistant to PDT with aerobic glycolysis., This research was funded by Spanish grants from Instituto de Salud Carlos III MINECO and Feder Funds (FIS PI15/00974 and PI18/00708) and Ministerio de Ciencia, Innovación y Universidades (SAF2016-75916-R).
- Published
- 2020
10. Dysfunctional oxidative phosphorylation shunts branched-chain amino acid catabolism onto lipogenesis in skeletal muscle
- Author
-
José M. Cuezva, Sandra Serrano Sanz, Ana Ramírez de Molina, Juan Cruz Herrero Martín, Marta P. Pereira, Laura Formentini, Cristina Sánchez-González, Cristina Nuevo-Tapioles, UAM. Departamento de Biología Molecular, Ministerio de Economía, Industria y Competitividad (España), Centro de Investigación Biomédica en Red Enfermedades Raras (España), Ministerio de Economía y Competitividad (España), and Comunidad de Madrid
- Subjects
medicine.medical_specialty ,Mice, Transgenic ,Oxidative phosphorylation ,Mitochondrion ,Oxidative Phosphorylation ,Article ,General Biochemistry, Genetics and Molecular Biology ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,Lipid droplet ,Edaravone ,medicine ,Animals ,Muscle, Skeletal ,Musculoskeletal System ,Molecular Biology ,030304 developmental biology ,0303 health sciences ,General Immunology and Microbiology ,ATP synthase ,biology ,Catabolism ,Lipogenesis ,General Neuroscience ,Skeletal muscle ,Lipid metabolism ,Insulin resistance ,Articles ,Biología y Biomedicina / Biología ,Mitochondria ,Acetyl‐CoA ,Metabolism ,Endocrinology ,medicine.anatomical_structure ,Acetyl-CoA ,biology.protein ,Amino Acids, Branched-Chain ,030217 neurology & neurosurgery - Abstract
It is controversial whether mitochondrial dysfunction in skeletal muscle is the cause or consequence of metabolic disorders. Herein, we demonstrate that in vivo inhibition of mitochondrial ATP synthase in muscle alters whole‐body lipid homeostasis. Mice with restrained mitochondrial ATP synthase activity presented intrafiber lipid droplets, dysregulation of acyl‐glycerides, and higher visceral adipose tissue deposits, poising these animals to insulin resistance. This mitochondrial energy crisis increases lactate production, prevents fatty acid β‐oxidation, and forces the catabolism of branched‐chain amino acids (BCAA) to provide acetyl‐CoA for de novo lipid synthesis. In turn, muscle accumulation of acetyl‐CoA leads to acetylation‐dependent inhibition of mitochondrial respiratory complex II enhancing oxidative phosphorylation dysfunction which results in augmented ROS production. By screening 702 FDA‐approved drugs, we identified edaravone as a potent mitochondrial antioxidant and enhancer. Edaravone administration restored ROS and lipid homeostasis in skeletal muscle and reinstated insulin sensitivity. Our results suggest that muscular mitochondrial perturbations are causative of metabolic disorders and that edaravone is a potential treatment for these diseases., Inhibition of ATP synthase in the muscle alters whole‐body lipid metabolism and induces insulin resistance in mice, two phenotypes that can be reverted by edavarone treatment.
- Published
- 2020
11. Metabolic reprogramming and disease progression in cancer patients
- Author
-
Cristina Nuevo-Tapioles, Laura Torresano, Fulvio Santacatterina, and José M. Cuezva
- Subjects
0301 basic medicine ,Carcinogenesis ,medicine.medical_treatment ,Down-Regulation ,Antineoplastic Agents ,Oxidative phosphorylation ,Mitochondrion ,Oxidative Phosphorylation ,Targeted therapy ,03 medical and health sciences ,0302 clinical medicine ,Downregulation and upregulation ,medicine ,Biomarkers, Tumor ,Animals ,Humans ,Molecular Biology ,Glutaminolysis ,business.industry ,Lipogenesis ,Carcinoma ,Prognosis ,Phenotype ,Mitochondria ,Up-Regulation ,Gene Expression Regulation, Neoplastic ,Survival Rate ,Disease Models, Animal ,Oxidative Stress ,030104 developmental biology ,030220 oncology & carcinogenesis ,Mutation ,Cancer research ,Disease Progression ,Molecular Medicine ,business ,Energy Metabolism ,Reactive Oxygen Species ,Reprogramming ,Glycolysis - Abstract
Genomics has contributed to the treatment of a fraction of cancer patients. However, there is a need to profile the proteins that define the phenotype of cancer and its pathogenesis. The reprogramming of metabolism is a major trait of the cancer phenotype with great potential for prognosis and targeted therapy. This review overviews the major changes reported in the steady-state levels of proteins of metabolism in primary carcinomas, paying attention to those enzymes that correlate with patients' survival. The upregulation of enzymes of glycolysis, pentose phosphate pathway, lipogenesis, glutaminolysis and the antioxidant defense is concurrent with the downregulation of mitochondrial proteins involved in oxidative phosphorylation, emphasizing the potential of mitochondrial metabolism as a promising therapeutic target in cancer. We stress that high-throughput quantitative expression profiling of differentially expressed proteins in large cohorts of carcinomas paired with normal tissues will accelerate translation of metabolism to a successful personalized medicine in cancer.
- Published
- 2019
12. Coordinate β-adrenergic inhibition of mitochondrial activity and angiogenesis arrest tumor growth
- Author
-
Konstantinos Stamatakis, José M. Cuezva, Fulvio Santacatterina, Marta Gómez de Cedrón, Cristina Núñez de Arenas, Cristina Nuevo-Tapioles, Laura Formentini, Ministerio de Economía y Competitividad (España), Centro de Investigación Biomédica en Red Enfermedades Raras (España), Instituto de Salud Carlos III, and Fundación Ramón Areces
- Subjects
0301 basic medicine ,Male ,Adrenergic Antagonists ,Angiogenesis ,Molecular biology ,Science ,General Physics and Astronomy ,Breast Neoplasms ,Oxidative phosphorylation ,Mitochondrion ,medicine.disease_cause ,Biochemistry ,General Biochemistry, Genetics and Molecular Biology ,Oxidative Phosphorylation ,Article ,Nebivolol ,03 medical and health sciences ,0302 clinical medicine ,Cell Line, Tumor ,medicine ,Humans ,lcsh:Science ,Cell Proliferation ,Cancer ,Multidisciplinary ,ATP synthase ,biology ,Molecular medicine ,Chemistry ,Proteins ,NADH Dehydrogenase ,General Chemistry ,Mitochondrial Proton-Translocating ATPases ,Mitochondria ,Endothelial stem cell ,030104 developmental biology ,030220 oncology & carcinogenesis ,Cancer cell ,Colonic Neoplasms ,biology.protein ,Cancer research ,Phosphorylation ,lcsh:Q ,Angiogenesis Inducing Agents ,Female ,Oxidative stress ,Biomarkers - Abstract
Mitochondrial metabolism has emerged as a promising target against the mechanisms of tumor growth. Herein, we have screened an FDA-approved library to identify drugs that inhibit mitochondrial respiration. The β1-blocker nebivolol specifically hinders oxidative phosphorylation in cancer cells by concertedly inhibiting Complex I and ATP synthase activities. Complex I inhibition is mediated by interfering the phosphorylation of NDUFS7. Inhibition of the ATP synthase is exerted by the overexpression and binding of the ATPase Inhibitory Factor 1 (IF1) to the enzyme. Remarkably, nebivolol also arrests tumor angiogenesis by arresting endothelial cell proliferation. Altogether, targeting mitochondria and angiogenesis triggers a metabolic and oxidative stress crisis that restricts the growth of colon and breast carcinomas. Nebivolol holds great promise to be repurposed for the treatment of cancer patients., FPI-MINECO and Fondo Social Europeo. L.F. received support from the Ramón y Cajal Program (RyC-2013-13693). The work was supported by grants from MINECO (SAF2016-75916-R and PID2019-108674RB-100), CIBERER-ISCIII (CB06/07/0017) and Fundación Ramón Areces
- Published
- 2019
13. Mitochondrial ROS Production Protects the Intestine from Inflammation through Functional M2 Macrophage Polarization
- Author
-
Laura Formentini, Michael P. Murphy, David López-Martínez, José M. Cuezva, Fulvio Santacatterina, Manuel Fresno, Angela Logan, Ron Smits, Konstantinos Stamatakis, Cristina Núñez de Arenas, Fundación Ramón Areces, Centro de Investigación Biomédica en Red Enfermedades Raras (España), Ministerio de Educación y Ciencia (España), Murphy, Mike [0000-0003-1115-9618], Apollo - University of Cambridge Repository, and Gastroenterology & Hepatology
- Subjects
0301 basic medicine ,Mitochondrial ROS ,STAT3 Transcription Factor ,Inflammation ,Biology ,Mitochondrion ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,chemistry.chemical_compound ,Mice ,medicine ,Animals ,lcsh:QH301-705.5 ,innate immunity ,PI3K/AKT/mTOR pathway ,Cells, Cultured ,MitoQ ,Innate immune system ,ATP synthase ,Macrophages ,TOR Serine-Threonine Kinases ,NF-kappa B ,Macrophage Activation ,Mitochondrial Proton-Translocating ATPases ,cytokines ,Immunity, Innate ,3. Good health ,Cell biology ,Mitochondria ,Intestines ,Mice, Inbred C57BL ,030104 developmental biology ,Phenotype ,lcsh:Biology (General) ,chemistry ,biology.protein ,Colitis, Ulcerative ,medicine.symptom ,Signal transduction ,Reactive Oxygen Species ,Proto-Oncogene Proteins c-akt ,Signal Transduction - Abstract
Mitochondria are signaling hubs in cellular physiology that play a role in inflammatory diseases. We found that partial inhibition of the mitochondrial ATP synthase in the intestine of transgenic mice triggers an anti-inflammatory response through NFκB activation mediated by mitochondrial mtROS. This shielding phenotype is revealed when mice are challenged by DSS-induced colitis, which, in control animals, triggers inflammation, recruitment of M1 pro-inflammatory macrophages, and the activation of the pro-oncogenic STAT3 and Akt/mTOR pathways. In contrast, transgenic mice can polarize macrophages to the M2 anti-inflammatory phenotype. Using the mitochondria-targeted antioxidant MitoQ to quench mtROS in vivo, we observe decreased NFκB activation, preventing its cellular protective effects. These findings stress the relevance of mitochondrial signaling to the innate immune system and emphasize the potential role of the ATP synthase as a therapeutic target in inflammatory and other related diseases., MEC (SAF2013-41945-R and SAF2016-75916-R), CIBERER-ISCIII, Comunidad Madrid (S2011/BMD-2402), and Fundación Ramón Areces, Spain. M.P.M.
- Published
- 2017
14. The ATPase Inhibitory Factor 1 (IF1): A master regulator of energy metabolism and of cell survival
- Author
-
José M. Cuezva and Javier Garcia-Bermudez
- Subjects
0301 basic medicine ,Bioenergetics ,Cellular differentiation ,Biophysics ,Oxidative phosphorylation ,Mitochondrion ,Biology ,Biochemistry ,Oxidative Phosphorylation ,03 medical and health sciences ,0302 clinical medicine ,Neoplasms ,Serine ,Humans ,Phosphorylation ,Hypoxia ,Protein kinase A ,Cyclic AMP-Dependent Protein Kinase Catalytic Subunits ,ATP synthase ,Proteins ,Cell Biology ,Mitochondrial Proton-Translocating ATPases ,Cellular Reprogramming ,Mitochondria ,Cell biology ,Protein Subunits ,Cell Transformation, Neoplastic ,030104 developmental biology ,Gene Expression Regulation ,030220 oncology & carcinogenesis ,biology.protein ,Protons ,Signal transduction ,Reactive Oxygen Species ,Signal Transduction - Abstract
In this contribution we summarize most of the findings reported for the molecular and cellular biology of the physiological inhibitor of the mitochondrial H(+)-ATP synthase, the engine of oxidative phosphorylation (OXPHOS) and gate of cell death. We first describe the structure and major mechanisms and molecules that regulate the activity of the ATP synthase placing the ATPase Inhibitory Factor 1 (IF1) as a major determinant in the regulation of the activity of the ATP synthase and hence of OXPHOS. Next, we summarize the post-transcriptional mechanisms that regulate the expression of IF1 and emphasize, in addition to the regulation afforded by the protonation state of histidine residues, that the activity of IF1 as an inhibitor of the ATP synthase is also regulated by phosphorylation of a serine residue. Phosphorylation of S39 in IF1 by the action of a mitochondrial cAMP-dependent protein kinase A hampers its interaction with the ATP synthase, i.e., only dephosphorylated IF1 interacts with the enzyme. Upon IF1 interaction with the ATP synthase both the synthetic and hydrolytic activities of the engine of OXPHOS are inhibited. These findings are further placed into the physiological context to stress the emerging roles played by IF1 in metabolic reprogramming in cancer, in hypoxia and in cellular differentiation. We review also the implication of IF1 in other cellular situations that involve the malfunctioning of mitochondria. Special emphasis is given to the role of IF1 as driver of the generation of a reactive oxygen species signal that, emanating from mitochondria, is able to reprogram the nucleus of the cell to confer by various signaling pathways a cell-death resistant phenotype against oxidative stress. Overall, our intention is to highlight the urgent need of further investigations in the molecular and cellular biology of IF1 and of its target, the ATP synthase, to unveil new therapeutic strategies in human pathology. This article is part of a Special Issue entitled 'EBEC 2016: 19th European Bioenergetics Conference, Riva del Garda, Italy, July 2-6, 2016', edited by Prof. Paolo Bernardi.
- Published
- 2016
- Full Text
- View/download PDF
15. In vivo evidence of mitochondrial dysfunction and altered redox homeostasis in a genetic mouse model of propionic acidemia: Implications for the pathophysiology of this disorder
- Author
-
Lourdes R. Desviat, Eva Richard, Michael P. Murphy, Belén Pérez, Sonia Medina, C. Cuevas-Martín, Angela Logan, Angel Gil-Izquierdo, Lorena Gallego-Villar, José M. Cuezva, Adrián González-Quintana, Ana Rivera-Barahona, Michael A. Barry, Miguel Martín, and Adam J. Guenzel
- Subjects
0301 basic medicine ,GPX1 ,Methylmalonyl-CoA Decarboxylase ,Propionic Acidemia ,Protein Array Analysis ,Oxidative phosphorylation ,Mitochondrion ,Biology ,medicine.disease_cause ,DNA, Mitochondrial ,Biochemistry ,Antioxidants ,Mice ,03 medical and health sciences ,chemistry.chemical_compound ,Superoxides ,Physiology (medical) ,medicine ,Animals ,Homeostasis ,Humans ,Propionic acidemia ,chemistry.chemical_classification ,Reactive oxygen species ,Superoxide ,Hydrogen Peroxide ,medicine.disease ,Mitochondria ,Disease Models, Animal ,Oxidative Stress ,030104 developmental biology ,Mitochondrial biogenesis ,chemistry ,Lipid Peroxidation ,Reactive Oxygen Species ,Oxidation-Reduction ,Oxidative stress - Abstract
Accumulation of toxic metabolites has been described to inhibit mitochondrial enzymes, thereby inducing oxidative stress in propionic acidemia (PA), an autosomal recessive metabolic disorder caused by the deficiency of mitochondrial propionyl-CoA carboxylase. PA patients exhibit neurological deficits and multiorgan complications including cardiomyopathy. To investigate the role of mitochondrial dysfunction in the development of these alterations we have used a hypomorphic mouse model of PA that mimics the biochemical and clinical hallmarks of the disease. We have studied the tissue-specific bioenergetic signature by Reverse Phase Protein Microarrays and analysed OXPHOS complex activities, mtDNA copy number, oxidative damage, superoxide anion and hydrogen peroxide levels. The results show decreased levels and/or activity of several OXPHOS complexes in different tissues of PA mice. An increase in mitochondrial mass and OXPHOS complexes was observed in brain, possibly reflecting a compensatory mechanism including metabolic reprogramming. mtDNA depletion was present in most tissues analysed. Antioxidant enzymes were also found altered. Lipid peroxidation was present along with an increase in hydrogen peroxide and superoxide anion production. These data support the hypothesis that oxidative damage may contribute to the pathophysiology of PA, opening new avenues in the identification of therapeutic targets and paving the way for in vivo evaluation of compounds targeting mitochondrial biogenesis or reactive oxygen species production.
- Published
- 2016
- Full Text
- View/download PDF
16. PD-1 signaling affects cristae morphology and leads to mitochondrial dysfunction in human CD8+ T lymphocytes
- Author
-
Jesús Ogando, Cristina Nuevo-Tapioles, Antonio González-Pérez, Marta Gut, Anna Esteve-Codina, Santos Mañes, Simon Heath, José M. Cuezva, Rosa Ana Lacalle, Javier Santos, María Eugenia Sáez, UAM. Departamento de Biología Molecular, Ministerio de Economía y Competitividad (España), Agencia Estatal de Investigación (España), Ministerio de Ciencia, Innovación y Universidades (España), Fundación Merck Salud, European Commission, Instituto de Salud Carlos III, and Comunidad de Madrid
- Subjects
0301 basic medicine ,Cancer Research ,Mitochondrial DNA ,T cell ,Immunology ,Oxidative phosphorylation ,Mitochondrion ,lcsh:RC254-282 ,Cristae ,03 medical and health sciences ,0302 clinical medicine ,PD-1 ,medicine ,Immunology and Allergy ,Receptor ,Pharmacology ,Chemistry ,CD28 ,CD8 ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Biología y Biomedicina / Biología ,Cell biology ,MICOS ,030104 developmental biology ,medicine.anatomical_structure ,Metabolism ,Oncology ,030220 oncology & carcinogenesis ,Molecular Medicine ,Signal transduction ,RNA-seq - Abstract
[Background] Binding of the programmed death-1 (PD-1) receptor to its ligands (PD-L1/2) transduces inhibitory signals that promote exhaustion of activated T cells. Blockade of the PD-1 pathway is widely used for cancer treatment, yet the inhibitory signals transduced by PD-1 in T cells remain elusive., [Methods] Expression profiles of human CD8+ T cells in resting, activated (CD3 + CD28) and PD-1-stimulated cells (CD3 + CD28 + PD-L1-Fc) conditions were evaluated by RNA-seq. Bioinformatic analyses were used to identify signaling pathways differentially regulated in PD-1-stimulated cells. Metabolic analyses were performed with SeaHorse technology, and mitochondrial ultrastructure was determined by transmission electron microscopy. PD-1-regulated mitochondrial genes were silenced using short-hairpin RNA in primary cells. Blue native gel electrophoresis was used to determine respiratory supercomplex assembly., [Results] PD-1 engagement in human CD8+ T cells triggers a specific, progressive genetic program different from that found in resting cells. Gene ontology identified metabolic processes, including glycolysis and oxidative phosphorylation (OXPHOS), as the main pathways targeted by PD-1. We observed severe functional and structural alterations in the mitochondria of PD-1-stimulated cells, including a reduction in the number and length of mitochondrial cristae. These cristae alterations were associated with reduced expression of CHCHD3 and CHCHD10, two proteins that form part of the mitochondrial contact site and cristae organizing system (MICOS). Although PD-1-stimulated cells showed severe cristae alterations, assembly of respiratory supercomplexes was unexpectedly greater in these cells than in activated T cells. CHCHD3 silencing in primary CD8+ T cells recapitulated some effects induced by PD-1 stimulation, including reduced mitochondrial polarization and interferon-γ production following T cell activation with anti-CD3 and -CD28 activating antibodies., [Conclusions] Our results suggest that mitochondria are the main targets of PD-1 inhibitory activity. PD-1 reprograms CD8+ T cell metabolism for efficient use of fatty acid oxidation; this mitochondrial phenotype might explain the long-lived phenotype of PD-1-engaged T cells., This work was funded by grants from the Spanish Ministerio de Economía y Competitividad (MINECO) (SAF2014–54475-R and SAF2017–83732-R to SM; SAF2016–75916-R to JMC; AEI/FEDER, EU), the Instituto de Salud Carlos III (PT17 PT17/0009/0019, AEI/FEDER, EU), the Comunidad de Madrid (B2017/BMD-3733; Inmunothercan-CM, to SM), and the Merck-Salud Foundation (to SM). JO, JS and CN-T are supported by predoctoral fellowships from the MINECO and the EU European Social Fund.
- Published
- 2019
17. PD-1 signaling affects cristae morphology and leads to mitochondrial dysfunction in human CD8
- Author
-
Jesús, Ogando, María Eugenia, Sáez, Javier, Santos, Cristina, Nuevo-Tapioles, Marta, Gut, Anna, Esteve-Codina, Simon, Heath, Antonio, González-Pérez, José M, Cuezva, Rosa Ana, Lacalle, and Santos, Mañes
- Subjects
Programmed Cell Death 1 Receptor ,Gene Expression ,CD8 ,CD8-Positive T-Lymphocytes ,B7-H1 Antigen ,Oxidative Phosphorylation ,Mitochondria ,Cristae ,MICOS ,Mitochondrial Proteins ,HEK293 Cells ,Metabolism ,PD-1 ,Leukocytes, Mononuclear ,Humans ,RNA-seq ,Mitochondrion ,Glycolysis ,Cells, Cultured ,Signal Transduction ,Research Article - Abstract
Background Binding of the programmed death-1 (PD-1) receptor to its ligands (PD-L1/2) transduces inhibitory signals that promote exhaustion of activated T cells. Blockade of the PD-1 pathway is widely used for cancer treatment, yet the inhibitory signals transduced by PD-1 in T cells remain elusive. Methods Expression profiles of human CD8+ T cells in resting, activated (CD3 + CD28) and PD-1-stimulated cells (CD3 + CD28 + PD-L1-Fc) conditions were evaluated by RNA-seq. Bioinformatic analyses were used to identify signaling pathways differentially regulated in PD-1-stimulated cells. Metabolic analyses were performed with SeaHorse technology, and mitochondrial ultrastructure was determined by transmission electron microscopy. PD-1-regulated mitochondrial genes were silenced using short-hairpin RNA in primary cells. Blue native gel electrophoresis was used to determine respiratory supercomplex assembly. Results PD-1 engagement in human CD8+ T cells triggers a specific, progressive genetic program different from that found in resting cells. Gene ontology identified metabolic processes, including glycolysis and oxidative phosphorylation (OXPHOS), as the main pathways targeted by PD-1. We observed severe functional and structural alterations in the mitochondria of PD-1-stimulated cells, including a reduction in the number and length of mitochondrial cristae. These cristae alterations were associated with reduced expression of CHCHD3 and CHCHD10, two proteins that form part of the mitochondrial contact site and cristae organizing system (MICOS). Although PD-1-stimulated cells showed severe cristae alterations, assembly of respiratory supercomplexes was unexpectedly greater in these cells than in activated T cells. CHCHD3 silencing in primary CD8+ T cells recapitulated some effects induced by PD-1 stimulation, including reduced mitochondrial polarization and interferon-γ production following T cell activation with anti-CD3 and -CD28 activating antibodies. Conclusions Our results suggest that mitochondria are the main targets of PD-1 inhibitory activity. PD-1 reprograms CD8+ T cell metabolism for efficient use of fatty acid oxidation; this mitochondrial phenotype might explain the long-lived phenotype of PD-1-engaged T cells. Electronic supplementary material The online version of this article (10.1186/s40425-019-0628-7) contains supplementary material, which is available to authorized users.
- Published
- 2018
18. Down-regulation of oxidative phosphorylation in the liver by expression of the ATPase inhibitory factor 1 induces a tumor-promoter metabolic state
- Author
-
Ángela M. Valverde, Juan M. Zapata, Carlos B. Rueda, Jorgina Satrústegui, José M. Cuezva, Laura Sánchez-Cenizo, Fulvio Santacatterina, Cristina Núñez de Arenas, Maysa A. Mobasher, María Sánchez-Aragó, Inmaculada Martínez-Reyes, Estela Casas, Javier Garcia-Bermudez, Laura Formentini, Ministerio de Economía y Competitividad (España), Comunidad de Madrid, and Fundación Ramón Areces
- Subjects
0301 basic medicine ,Programmed cell death ,Cell Survival ,Transgene ,p38 mitogen-activated protein kinases ,Blotting, Western ,Down-Regulation ,Gene Expression ,Apoptosis ,Mice, Transgenic ,Oxidative phosphorylation ,AMP-Activated Protein Kinases ,Mitochondrion ,p38 Mitogen-Activated Protein Kinases ,Oxidative Phosphorylation ,03 medical and health sciences ,AMP-activated protein kinase ,Animals ,Humans ,Acetaminophen ,Cancer ,ATPase inhibitory factor 1 ,biology ,Reverse Transcriptase Polymerase Chain Reaction ,Kinase ,Liver Neoplasms ,Proteins ,AMPK ,Energy metabolism ,Mitochondria ,Cell biology ,Microscopy, Electron ,030104 developmental biology ,Liver ,Microscopy, Fluorescence ,Oncology ,Mutation ,biology.protein ,Reactive oxygen species ,Research Paper - Abstract
The ATPase Inhibitory Factor 1 (IF1) is an inhibitor of the mitochondrial H+-ATP synthase that regulates the activity of both oxidative phosphorylation (OXPHOS) and cell death. Here, we have developed transgenic Tet-On and Tet-Off mice that express a mutant active form of hIF1 in the hepatocytes to restrain OXPHOS in the liver to investigate the relevance of mitochondrial activity in hepatocarcinogenesis. The expression of hIF1 promotes the inhibition of OXPHOS in both Tet-On and Tet- Off mouse models and induces a state of metabolic preconditioning guided by the activation of the stress kinases AMPK and p38 MAPK. Expression of the transgene significantly augmented proliferation and apoptotic resistance of carcinoma cells, which contributed to an enhanced diethylnitrosamine-induced liver carcinogenesis. Moreover, the expression of hIF1 also diminished acetaminophen-induced apoptosis, which is unrelated to differences in permeability transition pore opening. Mechanistically, cell survival in hIF1-preconditioned hepatocytes results from a nuclear factor-erythroid 2-related factor (Nrf2)-guided antioxidant response. The results emphasize in vivo that a metabolic phenotype with a restrained OXPHOS in the liver is prone to the development of cancer, MINECO (SAF2013-41945-R to JMC and SAF2012-33243 to AMV), CIBERER-ISCIII to JMC and CIBERDEM-ISCIII to AMV, Comunidad de Madrid (S2011/BMD-2402 to JMC and S2010/BMD-2423 to AMV), Fundación Ramón Areces (FRA)
- Published
- 2015
- Full Text
- View/download PDF
19. PKA Phosphorylates the ATPase Inhibitory Factor 1 and Inactivates Its Capacity to Bind and Inhibit the Mitochondrial H+-ATP Synthase
- Author
-
Araceli del Arco, Beatriz Soldevilla, José M. Cuezva, María Sánchez-Aragó, Cristina Nuevo-Tapioles, Javier Garcia-Bermudez, Comunidad de Madrid, Fundación Ramón Areces, Ministerio de Economía y Competitividad (España), and UAM. Departamento de Biología Molecular
- Subjects
Models, Molecular ,Oxidative phosphorylation ,Mitochondrion ,Mitochondria, Heart ,Oxidative Phosphorylation ,General Biochemistry, Genetics and Molecular Biology ,Mice ,chemistry.chemical_compound ,Adenosine Triphosphate ,Mitochondrial proton-translocating ATPases ,Protein kinases ,Animals ,Humans ,Glycolysis ,Phosphorylation ,lcsh:QH301-705.5 ,Enzyme Assays ,Sulfonamides ,Binding Sites ,ATP synthase ,biology ,Kinase ,Myocardium ,Colforsin ,Proteins ,Mitochondrial Proton-Translocating ATPases ,Biología y Biomedicina / Biología ,HCT116 Cells ,Isoquinolines ,Cyclic AMP-Dependent Protein Kinases ,Mitochondria ,Cell biology ,Kinetics ,lcsh:Biology (General) ,Bucladesine ,Gene Expression Regulation ,chemistry ,Biochemistry ,Anaerobic glycolysis ,biology.protein ,Protein IF1 ,Adenosine triphosphate ,Protein Binding ,Signal Transduction - Abstract
The mitochondrial H+ -ATP synthase synthesizes most of cellular ATP requirements by oxidative phosphorylation (OXPHOS). The ATPase Inhibitory Factor 1 (IF1) is known to inhibit the hydrolase activity of the H+ -ATP synthase in situations that compromise OXPHOS. Herein, we demonstrate that phosphorylation of S39 in IF1 by mitochondrial protein kinase A abolishes its capacity to bind the H+ -ATP synthase. Only dephosphorylated IF1 binds and inhibits both the hydrolase and synthase activities of the enzyme. The phosphorylation status of IF1 regulates the flux of aerobic glycolysis and ATP production through OXPHOS in hypoxia and during the cell cycle. Dephosphorylated IF1 is present in human carcinomas. Remarkably, mouse heart contains a large fraction of dephosphorylated IF1 that becomes phosphorylated and inactivated upon in vivo β-adrenergic stimulation. Overall, we demonstrate the essential function of the phosphorylation of IF1 in regulating energy metabolism and speculate that dephosho-IF1 might play a role in signaling mitohormesis., This work was supported by grants from Ministerio de Economía y Competitividad (SAF2013-41945-R); Comunidad de Madrid (S2011/BMD-2402); Fundación Ramón Areces (FRA), Spain. The CBMSO received an institutional grant from the FRA
- Published
- 2015
- Full Text
- View/download PDF
20. Sensitivity to anti-Fas is independent of increased cathepsin D activity and adrenodoxin reductase expression occurring in NOS-3 overexpressing HepG2 cells
- Author
-
Jordi Muntané, Manuel de la Mata, Manuel Rodríguez-Perálvarez, José Luis Montero-Álvarez, Clara I. Linares, Gustavo Ferrín, Patricia Aguilar-Melero, Eduardo Chicano-Gálvez, José M. Cuezva, María Sánchez-Aragó, Sandra González-Rubio, UAM. Departamento de Biología Molecular, Instituto de Salud Carlos III, and Centro de Investigación Biomédica en Red Enfermedades Hepáticas y Digestivas (España)
- Subjects
Proteomics ,Programmed cell death ,Hepatocarcinoma ,Nitro-oxidative stress ,Nitric Oxide Synthase Type III ,Proteome ,Cell Respiration ,Gene Dosage ,Cathepsin D ,Biology ,DNA, Mitochondrial ,Models, Biological ,Oxidative Phosphorylation ,Adrenodoxin reductase ,chemistry.chemical_compound ,Downregulation and upregulation ,Humans ,fas Receptor ,Molecular Biology ,Cell Death ,Fas-mediated apoptosis ,Hep G2 Cells ,Mitochondrial Turnover ,Cell Biology ,Biología y Biomedicina / Biología ,Molecular biology ,Ferredoxin-NADP Reductase ,Oxidative Stress ,Protein Transport ,Mitochondrial biogenesis ,chemistry ,Apoptosis ,Mitochondrial Membranes ,HSP60 ,Endothelial nitric oxide synthase ,Pepstatin - Abstract
© 2015 Elsevier B.V. Stable overexpression of endothelial nitric oxide synthase (NOS-3) in HepG2 cells (4TO-NOS) leads to increased nitro-oxidative stress and upregulation of the cell death mediators p53 and Fas. Thus, NOS-3 overexpression has been suggested as a useful antiproliferative mechanism in hepatocarcinoma cells. We aimed to identify the underlying mechanism of cell death induced by NOS-3 overexpression at basal conditions and with anti-Fas treatment. The intracellular localization of NOS-3, the nitro-oxidative stress and the mitochondrial activity were analysed. In addition, the protein expression profile in 4TO-NOS was screened for differentially expressed proteins potentially involved in the induction of apoptosis. NOS-3 localization in the mitochondrial outer membrane was not associated with changes in the respiratory cellular capacity, but was related to the mitochondrial biogenesis increase and with a higher protein expression of mitochondrial complex IV. Nitro-oxidative stress and cell death in NOS-3 overexpressing cells occurred with the expression increase of pro-apoptotic genes and a higher expression/activity of the enzymes adrenodoxin reductase mitochondrial (AR) and cathepsin D (CatD). CatD overexpression in 4TO-NOS was related to the apoptosis induction independently of its catalytic activity. In addition, CatD activity inhibition by pepstatin A was not effective in blocking apoptosis induced by anti-Fas. In summary, NOS-3 overexpression resulted in an increased sensitivity to anti-Fas induced cell death, independently of AR expression and CatD activity., Instituto de Salud Carlos III (FIS 09/00185). G. Ferrín was supported by the Networked Biomedical Research Center Hepatic and Digestive Diseases (CIBEREHD)
- Published
- 2015
- Full Text
- View/download PDF
21. MYC Induces a Hybrid Energetics Program Early in Cell Reprogramming
- Author
-
Javier Prieto, Martina Palomino-Schätzlein, Azahara Vallet-Sánchez, José M. Cuezva, Xavier Ponsoda, Fulvio Santacatterina, Josema Torres, Marian León, Laura Torresano, Jennifer Lippincott-Schwartz, Karen Giménez, Antonio Pineda-Lucena, Arnold Y. Seo, Ministerio de Economía y Competitividad (España), and Fundación Ramón Areces
- Subjects
0301 basic medicine ,Cell signaling ,Somatic cell ,Cèl·lules ,Cell ,Oxidative phosphorylation ,cell reprogramming, cell signaling, metabolism, mitochondrial dynamics ,Biology ,Hybrid Cells ,Biochemistry ,Mitochondrial Dynamics ,Article ,Oxidative Phosphorylation ,Mitocondris ,Proto-Oncogene Proteins c-myc ,03 medical and health sciences ,Metabolomics ,CDC2 Protein Kinase ,Genetics ,medicine ,cell signaling ,Animals ,Humans ,Glycolysis ,Phosphorylation ,lcsh:QH301-705.5 ,Membrane potential ,Membrane Potential, Mitochondrial ,lcsh:R5-920 ,cell reprogramming ,Cell Biology ,Cellular Reprogramming ,Cell biology ,Mitochondria ,Mice, Inbred C57BL ,030104 developmental biology ,medicine.anatomical_structure ,lcsh:Biology (General) ,lcsh:Medicine (General) ,Reprogramming ,metabolism ,Developmental Biology - Abstract
Summary Cell reprogramming is thought to be associated with a full metabolic switch from an oxidative- to a glycolytic-based metabolism. However, neither the dynamics nor the factors controlling this metabolic switch are fully understood. By using cellular, biochemical, protein array, metabolomic, and respirometry analyses, we found that c-MYC establishes a robust bivalent energetics program early in cell reprogramming. Cells prone to undergo reprogramming exhibit high mitochondrial membrane potential and display a hybrid metabolism. We conclude that MYC proteins orchestrate a rewiring of somatic cell metabolism early in cell reprogramming, whereby somatic cells acquire the phenotypic plasticity necessary for their transition to pluripotency in response to either intrinsic or external cues., Graphical Abstract, Highlights • Endogenous MYC biological activity is necessary for cell reprogramming • MYC drives mitochondrial fission early in cell reprogramming • MYC establishes a bivalent energetics state necessary for cell reprogramming • MYC polarizes mitochondria, poising cells for reprogramming, Torres and colleagues describe an MYC-dependent remodeling of mitochondrial dynamics and metabolism during the first stage of cell reprogramming. Endogenous MYC activity was found to be necessary for cell reprogramming, likely by establishing a hybrid metabolic state characterized by elevated glycolytic flux and a somatic oxidative metabolic rate. MYC polarized mitochondria by increasing ATPIF1 and labeled cells prone to cell reprogramming.
- Published
- 2018
22. Different mitochondrial genetic defects exhibit the same protein signature of metabolism in skeletal muscle of PEO and MELAS patients: A role for oxidative stress
- Author
-
Miguel A. Martín, Pau B. Esparza-Moltó, Alberto Blázquez, Laura Torresano, Fulvio Santacatterina, Montse Olivé, Eduard Gallardo, Alfonso Núñez-Salgado, José M. Cuezva, Adrián González-Quintana, and Elena García-Arumí
- Subjects
0301 basic medicine ,Ophthalmoplegia, Chronic Progressive External ,Mitochondrial Diseases ,Support Vector Machine ,Biopsy ,medicine.disease_cause ,Biochemistry ,Antioxidants ,MELAS Syndrome ,Protein arrays ,Glycolysis ,Child ,Beta oxidation ,biology ,Middle Aged ,Catalase ,Healthy Volunteers ,Child, Preschool ,Lactic acidosis ,Adult ,medicine.medical_specialty ,Adolescent ,SOD2 ,Mitochondrial diseases ,Oxidative phosphorylation ,DNA, Mitochondrial ,Young Adult ,03 medical and health sciences ,Metabolic enzymes ,Physiology (medical) ,Internal medicine ,medicine ,Humans ,Muscle, Skeletal ,Aged ,Superoxide Dismutase ,Mn-superoxide dismutase ,medicine.disease ,Citric acid cycle ,Oxidative Stress ,030104 developmental biology ,Endocrinology ,Gene Expression Regulation ,Oxidative stress ,biology.protein ,Reactive Oxygen Species ,Biomarkers - Abstract
A major challenge in mitochondrial diseases (MDs) is the identification of biomarkers that could inform of the mechanisms involved in the phenotypic expression of genetic defects. Herein, we have investigated the protein signature of metabolism and of the antioxidant response in muscle biopsies of clinically and genetically diagnosed patients with Progressive External Ophthalmoplegia due to single large-scale (PEO-sD) or multiple (PEO-mD) deletions of mtDNA and Mitochondrial Encephalopathy Lactic Acidosis and Stroke-like episode (MELAS) syndrome, and healthy donors. A high-throughput immunoassay technique that quantitates the expression of relevant proteins of glycolysis, glycogenolysis, pentose phosphate pathway, oxidative phosphorylation, pyruvate and fatty acid oxidation, tricarboxylic acid cycle and the antioxidant response in two large independent and retrospectively collected cohorts of PEO-sD, PEO-mD and MELAS patients revealed that despite the heterogeneity of the genetic alterations, the three MDs showed the same metabolic signatures in both cohorts of patients, which were highly divergent from those of healthy individuals. Linear Discriminant Analysis and Support Vector Machine classifier provided a minimum of four biomarkers to discriminate healthy from pathological samples. Regardless of the induction of a large number of enzymes involved in ameliorating oxidative stress, the down-regulation of mitochondrial superoxide dismutase (SOD2) and catalase expression favored the accumulation of oxidative damage in patients’ proteins. Down-regulation of SOD2 and catalase expression in MD patients is not due to relevant changes in the availability of their mRNAs, suggesting that oxidative stress regulates the expression of the two enzymes post-transcriptionally. We suggest that SOD2 and catalase could provide specific targets to improve the detoxification of reactive oxygen species that affects muscle proteins in these patients.
- Published
- 2018
23. The Role of Mitochondrial H+-ATP Synthase in Cancer
- Author
-
José M. Cuezva, Pau B. Esparza-Moltó, Ministerio de Economía y Competitividad (España), Centro de Investigación Biomédica en Red Cáncer (España), and Fundación Ramón Areces
- Subjects
0301 basic medicine ,Mitochondrial ROS ,Cancer Research ,Programmed cell death ,Mini Review ,mitohormesis ,oxidative phosphorylation ,Oxidative phosphorylation ,lcsh:RC254-282 ,03 medical and health sciences ,metabolic reprogramming ,Glycolysis ,ATPase inhibitory factor 1 ,ATP synthase ,biology ,Chemistry ,hepatocarcinogenesis ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Cell biology ,030104 developmental biology ,Oncology ,Anaerobic glycolysis ,inflammation ,Cancer cell ,biology.protein ,Signal transduction - Abstract
Cancer cells reprogram energy metabolism by boosting aerobic glycolysis as a main pathway for the provision of metabolic energy and of precursors for anabolic purposes. Accordingly, the relative expression of the catalytic subunit of the mitochondrial H-ATP synthase-the core hub of oxidative phosphorylation-is downregulated in human carcinomas when compared with its expression in normal tissues. Moreover, some prevalent carcinomas also upregulate the ATPase inhibitory factor 1 (IF1), which is the physiological inhibitor of the H-ATP synthase. IF1 overexpression, both in cells in culture and in tissue-specific mouse models, is sufficient to reprogram energy metabolism to an enhanced glycolysis by limiting ATP production by the H-ATP synthase. Furthermore, the IF1-mediated inhibition of the H-ATP synthase promotes the production of mitochondrial ROS (mtROS). mtROS modulate signaling pathways favoring cellular proliferation and invasion, the activation of antioxidant defenses, resistance to cell death, and modulation of the tissue immune response, favoring the acquisition of several cancer traits. Consistently, IF1 expression is an independent marker of cancer prognosis. By contrast, inhibition of the H-ATP synthase by α-ketoglutarate and the oncometabolite 2-hydroxyglutarate, reduces mTOR signaling, suppresses cancer cell growth, and contributes to lifespan extension in several model organisms. Hence, the H-ATP synthase appears as a conserved hub in mitochondria-to-nucleus signaling controlling cell fate. Unraveling the molecular mechanisms responsible for IF1 upregulation in cancer and the signaling cascades that are modulated by the H-ATP synthase are of utmost interest to decipher the metabolic and redox circuits contributing to cancer origin and progression., MINECO (SAF2016-75916-R), CIBERER-ISCIII, and Fundación Ramón Areces, Spain.
- Published
- 2018
24. The mitochondrial ATP synthase is a shared drug target for aging and dementia
- Author
-
Eric P. Ratliff, Pau B. Esparza-Moltó, Marguerite Prior, Michael Petrascheck, David Schubert, Wolfgang H. Fischer, Pamela Maher, Kim D. Finley, Richard Dargusch, Daniel Daugherty, José M. Cuezva, Antonio Currais, Joshua Goldberg, Chandramouli Chiruta, Nomis Foundation, Edward N. and Della L. Thome Memorial Foundation, Bundy Family Foundation, Hewitt Foundation, Salk Institute for Biological Studies, and UAM. Departamento de Biología Molecular
- Subjects
0301 basic medicine ,Aging ,Phenotypic screening ,Mitochondrion ,Biology ,03 medical and health sciences ,Aginga, Alzheimer’s diseasea, ATP synthase ,medicine ,Aging brain ,Humans ,Neurodegeneration ,PI3K/AKT/mTOR pathway ,CAMKK2 ,ATP synthase ,Kinase ,Cell Biology ,Original Articles ,Mitochondrial Proton-Translocating ATPases ,Alzheimer's disease ,medicine.disease ,Biología y Biomedicina / Biología ,3. Good health ,Cell biology ,Mitochondria ,030104 developmental biology ,Metabolism ,biology.protein ,Original Article ,Dementia - Abstract
Aging is a major driving force underlying dementia, such as that caused by Alzheimer's disease (AD). While the idea of targeting aging as a therapeutic strategy is not new, it remains unclear how closely aging and age-associated diseases are coupled at the molecular level. Here, we discover a novel molecular link between aging and dementia through the identification of the molecular target for the AD drug candidate J147. J147 was developed using a series of phenotypic screening assays mimicking disease toxicities associated with the aging brain. We have previously demonstrated the therapeutic efficacy of J147 in several mouse models of AD. Here, we identify the mitochondrial α-F-ATP synthase (ATP5A) as a target for J147. By targeting ATP synthase, J147 causes an increase in intracellular calcium leading to sustained calcium/calmodulin-dependent protein kinase kinase β (CAMKK2)-dependent activation of the AMPK/mTOR pathway, a canonical longevity mechanism. Accordingly, modulation of mitochondrial processes by J147 prevents age-associated drift of the hippocampal transcriptome and plasma metabolome in mice and extends lifespan in drosophila. Our results link aging and age-associated dementia through ATP synthase, a molecular drug target that can potentially be exploited for the suppression of both. These findings demonstrate that novel screens for new AD drug candidates identify compounds that act on established aging pathways, suggesting an unexpectedly close molecular relationship between the two., NIH R01AG046153 (D.S) and NIH/NIA SBIR 2R44AG033427 (K.F. and E.R.), the Nomis Foundation (AC), AI104034 and the Della Thome Foundation (PM), Bundy Foundation (DD), the Hewitt Foundation (JG), the Paul F. Glenn Center for Aging Research at the Salk Institute (JG)
- Published
- 2017
25. Overexpression of Mitochondrial IF1 Prevents Metastatic Disease of Colorectal Cancer by Enhancing Anoikis and Tumor Infiltration of NK Cells
- Author
-
Fulvio Santacatterina, Sara González-García, María L. Toribio, José M. Cuezva, Lucía González-Llorente, Zuzana Tirpakova, Ana García-Aguilar, Cristina Nuevo-Tapioles, UAM. Departamento de Biología Molecular, Ministerio de Ciencia, Innovación y Universidades (España), Centro de Investigación Biomédica en Red Enfermedades Raras (España), and Fundación Ramón Areces
- Subjects
0301 basic medicine ,Cancer Research ,Colorectal cancer ,Cell ,colorectal cancer ,Biology ,Article ,Metastasis ,law.invention ,Transcriptome ,ATPase Inhibitory Factor 1 ,03 medical and health sciences ,0302 clinical medicine ,law ,In vivo ,medicine ,metastasis ,Anoikis ,ATPase Inhibitor Factor 1 ,immune surveillance ,Prognosis ,Biología y Biomedicina / Biología ,medicine.disease ,In vitro ,Immune surveillance ,030104 developmental biology ,medicine.anatomical_structure ,Oncology ,030220 oncology & carcinogenesis ,Cancer research ,Suppressor ,prognosis - Abstract
Increasing evidences show that the ATPase Inhibitory Factor 1 (IF1), the physiological inhibitor of the ATP synthase, is overexpressed in a large number of carcinomas contributing to metabolic reprogramming and cancer progression. Herein, we show that in contrast to the findings in other carcinomas, the overexpression of IF1 in a cohort of colorectal carcinomas (CRC) predicts less chances of disease recurrence, IF1 being an independent predictor of survival. Bioinformatic and gene expression analyses of the transcriptome of colon cancer cells with differential expression of IF1 indicate that cells overexpressing IF1 display a less aggressive behavior than IF1 silenced (shIF1) cells. Proteomic and functional in vitro migration and invasion assays confirmed the higher tumorigenic potential of shIF1 cells. Moreover, shIF1 cells have increased in vivo metastatic potential. The higher metastatic potential of shIF1 cells relies on increased cFLIP-mediated resistance to undergo anoikis after cell detachment. Furthermore, tumor spheroids of shIF1 cells have an increased ability to escape from immune surveillance by NK cells. Altogether, the results reveal that the overexpression of IF1 acts as a tumor suppressor in CRC with an important anti-metastatic role, thus supporting IF1 as a potential therapeutic target in CRC., Ministerio de Ciencia, Innovación y Universidades (SAF2013-41945-R, SAF2016-75916-R and SAF2016-75452-R), CIBERER-ISCIII (CB06/07/0017) and Fundación Ramón Areces, Spain
- Published
- 2019
- Full Text
- View/download PDF
26. Overexpression of the ATPase Inhibitory Factor 1 Favors a Non-metastatic Phenotype in Breast Cancer
- Author
-
Cristina Nuevo-Tapioles, Lucia Garcia-Ledo, Inmaculada Martínez-Reyes, Beatriz Soldevilla, Lucía González-Llorente, Carmen Cuevas-Martín, José M. Cuezva, Consejo Superior de Investigaciones Científicas (España), Ministerio de Economía y Competitividad (España), Comunidad de Madrid, and Fundación Ramón Areces
- Subjects
0301 basic medicine ,Cancer Research ,medicine.medical_specialty ,extracellular matrix ,Mitochondrion ,Biology ,03 medical and health sciences ,0302 clinical medicine ,Breast cancer ,breast cancer ,cellular migration ,Internal medicine ,medicine ,Epithelial–mesenchymal transition ,Original Research ,ATPase inhibitory factor 1 ,gene expression analysis ,Cancer ,Cell migration ,medicine.disease ,cellular invasion ,mitochondria ,030104 developmental biology ,Endocrinology ,Oncology ,Anaerobic glycolysis ,030220 oncology & carcinogenesis ,Cancer cell ,Cancer research ,ATP synthase ,Signal transduction - Abstract
Partial suppression of mitochondrial oxidative phosphorylation and the concurrent activation of aerobic glycolysis is a hallmark of proliferating cancer cells. Overexpression of the ATPase inhibitory factor 1 (IF1), an in vivo inhibitor of the mitochondrial ATP synthase, is observed in most prevalent human carcinomas favoring metabolic rewiring to an enhanced glycolysis and cancer progression. Consistently, a high expression of IF1 in hepatocarcinomas and in carcinomas of the lung, bladder, and stomach and in gliomas is a biomarker of bad patient prognosis. In contrast to these findings, we have previously reported that a high expression level of IF1 in breast carcinomas is indicative of less chance to develop metastatic disease. This finding is especially relevant in the bad prognosis group of patients bearing triple-negative breast carcinomas. To investigate the molecular mechanisms that underlie the differential behavior of IF1 in breast cancer progression, we have developed the triple-negative BT549 breast cancer cell line that overexpresses IF1 stably. When compared to controls, IF1-cells partially shut down respiration and enhance aerobic glycolysis. Transcriptomic analysis suggested that migration and invasion were specifically inhibited in IF1-overexpressing breast cancer cells. Analysis of gene expression by qPCR and western blotting indicate that IF1 overexpression supports the maintenance of components of the extracellular matrix (ECM) and E-cadherin concurrently with the downregulation of components and signaling pathways involved in epithelial to mesenchymal transition. The overexpression of IF1 in breast cancer cells has no effect in the rates of cellular proliferation and in the cell death response to staurosporine and hydrogen peroxide. However, the overexpression of IF1 significantly diminishes the ability of the cells to grow in soft agar and to migrate and invade when compared to control cells. Overall, the results indicate that IF1 overexpression despite favoring a metabolic phenotype prone to cancer progression in the specific case of breast cancer cells also promotes the maintenance of the ECM impeding metastatic disease. These findings hence provide a mechanistic explanation to the better prognosis of breast cancer patients bearing tumors with high expression level of IF1., CN-T and IM-R were supported by pre-doctoral FPI-MEC and JAE-CSIC fellowships, respectively. This work was supported by grants from the Ministerio de Economía y Competitividad (SAF2013-41945-R; SAF2016-75916-R), Comunidad Madrid (S2011/BMD-2402), and Fundación Ramón Areces 2015, Spain.
- Published
- 2017
27. Pyruvate kinase M2 and the mitochondrial ATPase Inhibitory Factor 1 provide novel biomarkers of dermatomyositis: a metabolic link to oncogenesis
- Author
-
Francesc Cardellach, Fulvio Santacatterina, Marc Catalán-García, Cristina Núñez de Arenas, Josep M. Grau, María Sánchez-Aragó, José M. Cuezva, Glòria Garrabou, Comunidad de Madrid, Ministerio de Economía y Competitividad (España), Centro de Investigación Biomédica en Red Enfermedades Raras (España), Fundación Ramón Areces, Fundació Privada Cellex, Instituto de Salud Carlos III, and Universitat de Barcelona
- Subjects
0301 basic medicine ,Carcinogenesis ,Biopsy ,Mitochondrion ,Polymyositis ,ATPase Inhibitory Factor 1 ,0302 clinical medicine ,Cluster Analysis ,Glycolysis ,Pyruvate kinase M2 ,Medicine(all) ,Muscles ,Biochemical markers ,General Medicine ,Mitochondria ,030220 oncology & carcinogenesis ,Marcadors bioquímics ,Biomarker (medicine) ,HSP60 ,Subcellular Fractions ,medicine.medical_specialty ,Pyruvate Kinase ,Protein Array Analysis ,Biology ,PKM2 ,General Biochemistry, Genetics and Molecular Biology ,Dermatomyositis ,Antibodies ,03 medical and health sciences ,Muscular Diseases ,Internal medicine ,medicine ,Humans ,Malalties musculars ,Inflammation ,Dermatomiositis ,Myositis ,Biochemistry, Genetics and Molecular Biology(all) ,Research ,Proteins ,Reproducibility of Results ,Energy metabolism ,medicine.disease ,030104 developmental biology ,Endocrinology ,Inflammatory myopathies ,Miositis ,Pyruvate kinase ,Biomarkers - Abstract
[Background] Metabolic alterations play a role in the development of inflammatory myopathies (IMs). Herein, we have investigated through a multiplex assay whether proteins of energy metabolism could provide biomarkers of IMs., [Methods] A cohort of thirty-two muscle biopsies and forty plasma samples comprising polymyositis (PM), dermatomyositis (DM) and sporadic inclusion body myositis (sIBM) and control donors was interrogated with monoclonal antibodies against proteins of energy metabolism using reverse phase protein microarrays (RPPA), [Results] When compared to controls the expression of the proteins is not significantly affected in the muscle of PM patients. However, the expression of β-actin is significantly increased in DM and sIBM in consistence with muscle and fiber regeneration. Concurrently, the expression of some proteins involved in glucose metabolism displayed a significant reduction in muscle of sIBM suggesting a repression of glycolytic metabolism in these patients. In contrasts to these findings, the expression of the glycolytic pyruvate kinase isoform M2 (PKM2) and of the mitochondrial ATPase Inhibitor Factor 1 (IF1) and Hsp60 were significantly augmented in DM when compared to other IMs in accordance with a metabolic shift prone to cancer development. PKM2 alone or in combination with other biomarkers allowed the discrimination of control and IMs with very high (>95%) sensitivity and specificity. Unfortunately, plasma levels of PKM2 were not significantly altered in DM patients to recommend its use as a non-invasive biomarker of the disease., [Conclusions] Expression of proteins of energy metabolism in muscle enabled discrimination of patients with IMs. RPPA identified the glycolysis promoting PKM2 and IF1 proteins as specific biomarkers of dermatomyositis, providing a biochemical link of this IM with oncogenesis., FS was supported by a pre-doctoral fellowship from FPI-UAM Spain. The work was supported by Grants from the Ministerio de Economía y Competitividad (SAF2013-41945-R; SAF2016-75916-R), Fundación Ramón Areces (FRA), Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), ISCIII, Fundación CELLEX and Comunidad de Madrid (S2011/BMD-2402), Spain. The CBMSO receives an institutional grant from FRA.
- Published
- 2017
28. Regulation of the H+-ATP synthase by IF1: a role in mitohormesis
- Author
-
Cristina Nuevo-Tapioles, Pau B. Esparza-Moltó, and José M. Cuezva
- Subjects
0301 basic medicine ,Cell signaling ,Cellular homeostasis ,Context (language use) ,Mitochondrion ,Biology ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,Hormesis ,Neoplasms ,Animals ,Humans ,Glycolysis ,Molecular Biology ,Cancer ,chemistry.chemical_classification ,Pharmacology ,Cell Nucleus ,Reactive oxygen species ,ATP synthase ,Lifespan ,Metabolic reprogramming ,Reactive oxygen species (ROS) ,Proteins ,Cell Biology ,Mitochondrial Proton-Translocating ATPases ,Cell biology ,Mitochondria ,030104 developmental biology ,Biochemistry ,chemistry ,ATPase inhibitory factor 1 (IF1) ,biology.protein ,Molecular Medicine ,Signal transduction - Abstract
The mitochondrial H+-ATP synthase is a primary hub of cellular homeostasis by providing the energy required to sustain cellular activity and regulating the production of signaling molecules that reprogram nuclear activity needed for adaption to changing cues. Herein, we summarize findings regarding the regulation of the activity of the H+-ATP synthase by its physiological inhibitor, the ATPase inhibitory factor 1 (IF1) and their functional role in cellular homeostasis. First, we outline the structure and the main molecular mechanisms that regulate the activity of the enzyme. Next, we describe the molecular biology of IF1 and summarize the regulation of IF1 expression and activity as an inhibitor of the H+-ATP synthase emphasizing the role of IF1 as a main driver of energy rewiring and cellular signaling in cancer. Findings in transgenic mice in vivo indicate that the overexpression of IF1 is sufficient to reprogram energy metabolism to an enhanced glycolysis and activate reactive oxygen species (ROS)-dependent signaling pathways that promote cell survival. These findings are placed in the context of mitohormesis, a program in which a mild mitochondrial stress triggers adaptive cytoprotective mechanisms that improve lifespan. In this regard, we emphasize the role played by the H+-ATP synthase in modulating signaling pathways that activate the mitohormetic response, namely ATP, ROS and target of rapamycin (TOR). Overall, we aim to highlight the relevant role of the H+-ATP synthase and of IF1 in cellular physiology and the need of additional studies to decipher their contributions to aging and age-related diseases.
- Published
- 2017
29. Regulation of the H
- Author
-
Pau B, Esparza-Moltó, Cristina, Nuevo-Tapioles, and José M, Cuezva
- Subjects
Cell Nucleus ,Lifespan ,Metabolic reprogramming ,Proteins ,Reactive oxygen species (ROS) ,Review ,Mitochondrial Proton-Translocating ATPases ,Mitochondria ,Hormesis ,Neoplasms ,ATPase inhibitory factor 1 (IF1) ,Animals ,Humans ,Cancer - Abstract
The mitochondrial H+-ATP synthase is a primary hub of cellular homeostasis by providing the energy required to sustain cellular activity and regulating the production of signaling molecules that reprogram nuclear activity needed for adaption to changing cues. Herein, we summarize findings regarding the regulation of the activity of the H+-ATP synthase by its physiological inhibitor, the ATPase inhibitory factor 1 (IF1) and their functional role in cellular homeostasis. First, we outline the structure and the main molecular mechanisms that regulate the activity of the enzyme. Next, we describe the molecular biology of IF1 and summarize the regulation of IF1 expression and activity as an inhibitor of the H+-ATP synthase emphasizing the role of IF1 as a main driver of energy rewiring and cellular signaling in cancer. Findings in transgenic mice in vivo indicate that the overexpression of IF1 is sufficient to reprogram energy metabolism to an enhanced glycolysis and activate reactive oxygen species (ROS)-dependent signaling pathways that promote cell survival. These findings are placed in the context of mitohormesis, a program in which a mild mitochondrial stress triggers adaptive cytoprotective mechanisms that improve lifespan. In this regard, we emphasize the role played by the H+-ATP synthase in modulating signaling pathways that activate the mitohormetic response, namely ATP, ROS and target of rapamycin (TOR). Overall, we aim to highlight the relevant role of the H+-ATP synthase and of IF1 in cellular physiology and the need of additional studies to decipher their contributions to aging and age-related diseases.
- Published
- 2016
30. Correction to: Mitochondrial H+-ATP synthase in human skeletal muscle: contribution to dyslipidaemia and insulin resistance
- Author
-
Theodore P. Ciaraldi, Laura Formentini, José M. Cuezva, Francisco Villarreal, Leslie Carter, Pam R. Taub, John D. Lapek, Manuel Gálvez-Santisteban, Robert R. Henry, David Gonzalez, and Alexander J. Ryan
- Subjects
medicine.medical_specialty ,business.industry ,Endocrinology, Diabetes and Metabolism ,Skeletal muscle ,Human physiology ,medicine.disease ,Endocrinology ,Insulin resistance ,medicine.anatomical_structure ,Internal medicine ,Internal Medicine ,medicine ,H atp synthase ,business - Abstract
Owing to an oversight, the authors omitted to note that Dr Taub is a co-founder of and equity holder in Cardero Therapeutics.
- Published
- 2018
- Full Text
- View/download PDF
31. Dysfunction of the immune system in conditional IF1 (Atp5if1) knockout mice in colon
- Author
-
Sonia Domínguez-Zorita, Pau B. Esparza-Moltó, José M. Cuezva, Fulvio Santacatterina, and Cristina Nuevo-Tapioles
- Subjects
Immune system ,Immunology ,Knockout mouse ,Biophysics ,Cell Biology ,Biology ,Biochemistry - Published
- 2018
- Full Text
- View/download PDF
32. RNA-binding proteins regulate cell respiration and coenzyme Q biosynthesis by post-transcriptional regulation of COQ7
- Author
-
Myriam Gorospe, Guillermo López-Lluch, Marina Valenzuela-Villatoro, Ji Heon Noh, Daniel J. M. Fernández-Ayala, Imke M. Willers, Emilio Siendones, Gloria Brea, José M. Cuezva, Kotb Abdelmohsen, Plácido Navas, María V. Cascajo, Ministerio de Economía y Competitividad (España), Instituto de Salud Carlos III, National Institutes of Health (US), and Ministerio de Sanidad, Servicios Sociales e Igualdad (España)
- Subjects
0301 basic medicine ,Untranslated region ,Ubiquinone ,RNA-binding protein ,RNA-binding proteins ,Oxidative phosphorylation ,COQ7 ,Biology ,Oxidative Phosphorylation ,ELAV-Like Protein 1 ,03 medical and health sciences ,Oxygen Consumption ,coenzyme Q10 ,mitochondrial respiration ,Humans ,Molecular Biology ,3' Untranslated Regions ,Gene knockdown ,030102 biochemistry & molecular biology ,Post-transcriptional regulation ,Heterogeneous-Nuclear Ribonucleoprotein Group C ,Cell Biology ,030104 developmental biology ,Mitochondrial respiratory chain ,Biochemistry ,Gene Expression Regulation ,Coenzyme Q – cytochrome c reductase ,Respirasome ,HuR ,Coenzyme Q10 ,hnRNP C1/C2 ,Research Article ,Research Paper ,post-transcriptional regulation ,HeLa Cells ,Mitochondrial respiration - Abstract
et al., Coenzyme Q (CoQ) is a key component of the mitochondrial respiratory chain carrying electrons from complexes I and II to complex III and it is an intrinsic component of the respirasome. CoQ concentration is highly regulated in cells in order to adapt the metabolism of the cell to challenges of nutrient availability and stress stimuli. At least 10 proteins have been shown to be required for CoQ biosynthesis in a multi-peptide complex and COQ7 is a central regulatory factor of this pathway. We found that the first 765 bp of the 3′-untranslated region (UTR) of COQ7 mRNA contains cis-acting elements of interaction with RNA-binding proteins (RBPs) HuR and hnRNP C1/C2. Binding of hnRNP C1/C2 to COQ7 mRNA was found to require the presence of HuR, and hnRNP C1/C2 silencing appeared to stabilize COQ7 mRNA modestly. By contrast, lowering HuR levels by silencing or depriving cells of serum destabilized and reduced the half-life of COQ7 mRNA, thereby reducing COQ7 protein and CoQ biosynthesis rate. Accordingly, HuR knockdown decreased oxygen consumption rate and mitochondrial production of ATP, and increased lactate levels. Taken together, our results indicate that a reduction in COQ7 mRNA levels by HuR depletion causes mitochondrial dysfunction and a switch toward an enhanced aerobic glycolysis, the characteristic phenotype exhibited by primary deficiency of CoQ. Thus HuR contributes to efficient oxidative phosphorylation by regulating of CoQ biosynthesis., This research was supported by grants from the Spanish Ministry of Health, Instituto de Salud Carlos III (ISCIII), FIS PI14–01962 to PN, the Spanish Ministry of Economy and Competitively, SAF2013–41945-R to JMC, and the Intramural Research Program of the NIA, NIH (KA, MG).
- Published
- 2016
33. Determination of the H+-ATP Synthase and Hydrolytic Activities
- Author
-
José M. Cuezva, Cristina Nuevo-Tapioles, and Javier Garcia-Bermudez
- Subjects
ATP Hydrolase ,ATP synthase ,biology ,Chemistry ,Strategy and Management ,Mechanical Engineering ,Metals and Alloys ,Oxidative phosphorylation ,Mitochondrion ,Industrial and Manufacturing Engineering ,Enzyme assay ,Hydrolysis ,Biochemistry ,biology.protein ,H atp synthase - Published
- 2016
- Full Text
- View/download PDF
34. Adalimumab Reduces Photoreceptor Cell Death in A Mouse Model of Retinal Degeneration
- Author
-
Enrique J. de la Rosa, Cristina Martinez-Fernandez de la Camara, José M. Cuezva, David Hervás, David Salom, Alberto M. Hernández-Pinto, Regina Rodrigo, María Sánchez-Aragó, Carmen Cuevas-Martín, José M. Millán, Lorena Olivares-González, UAM. Departamento de Biología Molecular, and Ministerio de Economía y Competitividad (España)
- Subjects
Retinal degeneration ,Programmed cell death ,Poly ADP ribose polymerase ,Cell Count ,Biology ,Photoreceptor cell ,Antioxidants ,Retina ,Article ,Mice ,Retinitis pigmentosa ,medicine ,Animals ,Gliosis ,Inflammation ,Multidisciplinary ,Photoreceptor ,Cell Death ,Retinal Degeneration ,Adalimumab ,medicine.disease ,Biología y Biomedicina / Biología ,Disease Models, Animal ,medicine.anatomical_structure ,Immunology ,Cancer research ,Disease Progression ,Tumor necrosis factor alpha ,medicine.symptom ,Poly(ADP-ribose) Polymerases ,Energy Metabolism ,Retinitis Pigmentosa ,Photoreceptor Cells, Vertebrate - Abstract
13 p.-5 fig.-1 tab. Cristina Martínez-Fernández de la Cámara et alt., Growing evidence suggests that inflammation is involved in the progression of retinitis pigmentosa (RP) both in patients and in animal models. The aim of this study was to investigate the effect of Adalimumab, a monoclonal anti-TNFα antibody, on retinal degeneration in a murine model of human autosomal recessive RP, the rd10 mice at postnatal day (P) 18. In our housing conditions, rd10 retinas were seriously damaged at P18. Adalimumab reduced photoreceptor cell death, as determined by scoring the number of TUNEL-positive cells. In addition, nuclear poly (ADP) ribose (PAR) content, an indirect measure of PAR polymerase (PARP) activity, was also reduced after treatment. The blockade of TNFα ameliorated reactive gliosis, as visualized by decreased GFAP and IBA1 immunolabelling (Müller cell and microglial markers, respectively) and decreased up-regulation of TNFα gene expression. Adalimumab also improved antioxidant response by restoring total antioxidant capacity and superoxide dismutase activity. Finally, we observed that Adalimumab normalized energetic and metabolic pattern in rd10 mouse retinas. Our study suggests that the TNFα blockade could be a successful therapeutic approach to increase photoreceptor survival during the progression of RP. Further studies are needed to characterize its effect along the progression of the disease., This work was supported by the European Regional Development Fund, Institute of Health Carlos III, PI12/0481, SAF2013-41059-R and SAF2013-41945 from the Spanish Ministry of Economy and Competitiveness (MEC).
- Published
- 2015
35. Epigenetic regulation of the binding activity of translation inhibitory proteins that bind the 3′ untranslated region of β-F1-ATPase mRNA by adenine nucleotides and the redox state
- Author
-
José M. Cuezva and José M. Izquierdo
- Subjects
Untranslated region ,Ultraviolet Rays ,Biophysics ,RNA-binding protein ,Biology ,Models, Biological ,Biochemistry ,Gene Expression Regulation, Enzymologic ,Epigenesis, Genetic ,Fetus ,Pregnancy ,Adenine nucleotide ,Animals ,RNA, Messenger ,Rats, Wistar ,3' Untranslated Regions ,Molecular Biology ,Psychological repression ,Messenger RNA ,Adenine Nucleotides ,Three prime untranslated region ,RNA-Binding Proteins ,RNA ,Translation (biology) ,Rats ,Proton-Translocating ATPases ,Cross-Linking Reagents ,Liver ,Protein Biosynthesis ,Female ,Oxidation-Reduction ,Protein Binding - Abstract
Here, we describe the binding affinities and the regulation of the binding activities of the fetal liver proteins that interact with the 3' untranslated region of beta-F1-ATPase mRNA (beta-mRNA). These proteins (3'beta FBPs), which are involved in the repression of beta-mRNA translation during fetal development, have poly(A)-binding activity. Reducing agents do not affect the RNA-binding activity of 3'beta FBPs. In contrast, oxidizing and alkylating reagents abolished the binding activity of 3'beta FBPs to its target RNA element, an effect that is partially prevented by the presence of reducing agents. Interestingly, the availability of adenine nucleotides regulates in a concentration-dependent manner the binding activities of 3'beta FBPs. The results suggest that epigenetic changes that occur at the time of birth affecting both the redox and energy state of the liver play a relevant role in the regulation of the binding activities of 3'beta FBPs and therefore in the translation of beta-mRNA.
- Published
- 2005
- Full Text
- View/download PDF
36. Quantitative analysis of proteins of metabolism by reverse phase protein microarrays identifies potential biomarkers of rare neuromuscular diseases
- Author
-
Miguel A. Martín, Fulvio Santacatterina, Margarita Chamorro, Carmen Navarro, Cristina Núñez de Arenas, María Sánchez-Aragó, José M. Cuezva, UAM. Departamento de Biología Molecular, Ministerio de Economía y Competitividad (España), Centro de Investigación Biomédica en Red Enfermedades Raras (España), Comunidad de Madrid, Fundación Ramón Areces, CSIC - Unidad de Recursos de Información Científica para la Investigación (URICI), and Ministerio de Ciencia e Innovación (España)
- Subjects
Male ,Proteomics ,Biopsy ,Protein Array Analysis ,Oxidative phosphorylation ,Mitochondrion ,Biology ,Bioinformatics ,Antibodies ,General Biochemistry, Genetics and Molecular Biology ,medicine ,Animals ,Humans ,skin and connective tissue diseases ,Myopathy ,Medicine(all) ,Mice, Inbred BALB C ,Biochemistry, Genetics and Molecular Biology(all) ,Research ,Muscles ,Reproducibility of Results ,Física ,Neuromuscular Diseases ,General Medicine ,Energy metabolism ,medicine.disease ,Biología y Biomedicina / Biología ,Mitochondria ,Rare diseases ,Citric acid cycle ,Neuromuscular diseases ,Immunology ,Protein microarray ,Female ,Neuronal ceroid lipofuscinosis ,sense organs ,medicine.symptom ,Biomarkers ,Limb-girdle muscular dystrophy - Abstract
Muscle diseases have been associated with changes in the expression of proteins involved in energy metabolism. To this aim we have developed a number of monoclonal antibodies against proteins of energy metabolism. Methods: Herein, we have used Reverse Phase Protein Microarrays (RPMA), a high throughput technique, to investigate quantitative changes in protein expression with the aim of identifying potential biomarkers in rare neuromuscular diseases. A cohort of 73 muscle biopsies that included samples from patients diagnosed of Duchenne (DMD), Becker (BMD), symptomatic forms of DMD and BMD in female carriers (Xp21 Carriers), Limb Girdle Muscular Dystrophy Type 2C (LGMD2C), neuronal ceroid lipofuscinosis (NCL), glycogenosis type V (Mc Ardle disease), isolated mitochondrial complex I deficiency, intensive care unit myopathy and control donors were investigated. The nineteen proteins of energy metabolism studied included members of the mitochondrial oxidation of pyruvate, the tricarboxylic acid cycle, ß-oxidation of fatty acids, electron transport and oxidative phosphorylation, glycogen metabolism, glycolysis and oxidative stress using highly specific antibodies. Results: The results indicate that the phenotype of energy metabolism offers potential biomarkers that could be implemented to refine the understanding of the biological principles of rare diseases and, eventually, the management of these patients. Conclusions: We suggest that some biomarkers of energy metabolism could be translated into the clinics to contribute to the improvement of the clinical handling of patients affected by rare diseases., We thank Dr. Sébastien Tosi (Advanced Digital Microscopy - ADM, IRB Barcelona) for the development of the Image Analysis tool in ImageJ macro language and support of the publication fee by the CSIC Open Access Publication Support Initiative through its Unit of Information Resources for Research (URICI). FS was supported by a pre-doctoral fellowship from FPI-UAM Spain. This work was supported by grants from the Ministerio de Ciencia e Innovación (TREAT-CMT), Ministerio de Economía y Competitividad (SAF2013-41945-R), the Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), ISCIII, Madrid, Ministerio de Economía y Competitividad (FIS-ISCIII PI 12/01683 and PI 10/02628), and Comunidad de Madrid (S2011/BMD-2402), Spain. The CBMSO receives an institutional grant from Fundación Ramón Areces.
- Published
- 2015
37. The Relevance of the Mitochondrial H+-ATP Synthase in Cancer Biology
- Author
-
Inmaculada Martínez-Reyes and José M. Cuezva
- Subjects
Metabolic pathway ,biology ,ATP synthase ,Anaerobic glycolysis ,Chemistry ,ATPase ,Cancer cell ,biology.protein ,Glycolysis ,Mitochondrion ,Glyceraldehyde 3-phosphate dehydrogenase ,Cell biology - Abstract
Cancer cells depend on metabolic changes to cover the increased energy and metabolite demands that sustain proliferation. The enhanced rate of aerobic glycolysis and the activation of other metabolic pathways provide the energy and building blocks that support cell division. These changes occurred in response to the partial silencing of the bioenergetic function of mitochondria, specifically of the H+-ATP synthase, which is the engine that produces most of the ATP that sustains cellular activities in normal differentiated aerobic cells. Changes in the bioenergetic phenotype of carcinomas can be assessed by the determination of the expression of the catalytic subunit of the H+-ATP synthase (β-F1-ATPase) relative to the expression of the enzyme of glycolysis glyceraldehyde-3-phosphate dehydrogenase (GAPDH). The β-F1-ATPase/GAPDH ratio provides a bioenergetic signature of the tumor with clinical relevance as a molecular marker of the prognosis of different cancer patients as well as of the tumor response to chemotherapy. Energy metabolism of cancer cells has become an attractive target for cancer therapy because it is a common phenotypic trait of most carcinomas. In addition, silencing of the H+-ATP synthase in most prevalent carcinomas is also exerted at the activity level by overexpression of the ATPase inhibitory factor 1 (IF1), a protein that contributes to metabolic rewiring and the signaling of cell death-resistant phenotypes in cancer cells.
- Published
- 2015
- Full Text
- View/download PDF
38. Assembly of the ribonucleoprotein complex containing the mRNA of the β-subunit of the mitochondrial H+-ATP synthase requires the participation of two distal cis-acting elements and a complex set of cellular trans-acting proteins
- Author
-
José M. Izquierdo, José M. Cuezva, Javier Ricart, Carlo Maria Di Liegro, Ricart, J., Izquierdo, J., Di Liegro, C., and Cuezva, J.
- Subjects
Male ,Translation ,Blotting, Western ,Mitochondria, Liver ,RNA-binding protein ,Biochemistry ,Reticulocyte ,Pregnancy ,Polysome ,P-bodies ,medicine ,Animals ,Oxidative phosphorylation ,RNA, Messenger ,Rats, Wistar ,3' Untranslated Regions ,Molecular Biology ,In Situ Hybridization ,Messenger RNA ,ATP synthase ,biology ,Three prime untranslated region ,RNA-Binding Proteins ,RNA ,Cell Biology ,Immunohistochemistry ,Rats ,Proton-Translocating ATPases ,medicine.anatomical_structure ,biology.protein ,mRNA localization ,Female ,Research Article - Abstract
The mRNA encoding the beta-subunit of the mitochondrial H(+)-ATP synthase (beta-F1-ATPase) is localized in an approx. 150 nm structure of the hepatocyte of mammals. In the present study, we have investigated the cis- and trans-acting factors involved in the generation of the ribonucleoprotein complex containing beta-F1-ATPase mRNA. Two cis-acting elements (beta1.2 and 3'beta) have been identified. The beta1.2 element is placed in the open reading frame, downstream of the region encoding the mitochondrial pre-sequence of the protein. The 3'beta element is the 3' non-translated region of the mRNA. Complex sets of proteins from the soluble and non-soluble fractions of the liver interact with the beta1.2 and 3'beta elements. A soluble p88, present also in reticulocyte lysate, displays binding specificity for both the cis-acting elements. Sedimentation and high-resolution in situ hybridization experiments showed that the structure containing the rat liver beta-F1-ATPase mRNA is found in fractions of high sucrose concentration, where large polysomes sediment. Treatment of liver extracts with EDTA promoted the mobilization of beta-F1-ATPase mRNA to fractions of lower sucrose concentration, suggesting that the structure containing beta-F1-ATPase mRNA is a large polysome. Finally, in vitro reconstitution experiments with reticulocyte lysate, using either the full-length, mutant or chimaeric versions of beta-F1-ATPase mRNA, reveal that the assembly of the beta-F1-ATPase mRNA polysome requires the co-operation of both the cis-acting mRNA determinants. The present study illustrates the existence of an intramolecular RNA cross-talking required for the association of the mRNA with the translational machinery.
- Published
- 2002
- Full Text
- View/download PDF
39. Plasma metabolome and skin proteins in Charcot-Marie-Tooth 1A patients
- Author
-
Clara Ibáñez, Teresa Sevilla, Carlos Casasnovas, Carmen Cuevas-Martín, José M. Cuezva, Carolina Simó, Carmen Espinós, Fulvio Santacatterina, Samuel I. Pascual, Francesc Palau, Beatriz Soldevilla, Celedonio Márquez-Infante, María Sánchez-Aragó, Maria Antonia Alberti, UAM. Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa (CBM), Instituto de Investigación Sanitaria Hospital Universitario de La Paz (IdiPAZ), Instituto de Salud Carlos III, and Universitat de Barcelona
- Subjects
0301 basic medicine ,Pathology ,Peripheral neuropathy ,Physiology ,Biopsy ,Cell Membranes ,Protein metabolism ,Linear Discriminant Analysis ,Biochemistry ,chemistry.chemical_compound ,0302 clinical medicine ,Malalties hereditàries ,Metabolites ,Medicine and Health Sciences ,Membrane Metabolism ,Medicine ,Prospective Studies ,Prospective cohort study ,Skin ,Protein Metabolism ,Multidisciplinary ,medicine.diagnostic_test ,Middle Aged ,Biología y Biomedicina / Biología ,Body Fluids ,Ultrahigh liquid chromatography ,Protein catabolism ,Blood ,CMT1A ,Neurology ,Metabolome ,Cellular Structures and Organelles ,Anatomy ,medicine.symptom ,Research Article ,Genetic diseases ,Adult ,congenital, hereditary, and neonatal diseases and abnormalities ,medicine.medical_specialty ,Medicina ,Science ,Surgical and Invasive Medical Procedures ,Inflammation ,macromolecular substances ,Amiotròfia neural progressiva de Charcot-Marie-Tooth ,Charcot-Marie-Tooth disease ,Potential biomarkers in plasma ,Blood Plasma ,03 medical and health sciences ,Metabolomics ,Humans ,Clinical genetics ,business.industry ,Proteins ,Biology and Life Sciences ,Cell Biology ,medicine.disease ,nervous system diseases ,Neuropathy ,Metabolism ,030104 developmental biology ,chemistry ,Sarcopenia ,Energy Metabolism ,business ,Biomarkers ,030217 neurology & neurosurgery - Abstract
Objective: Charcot-Marie-Tooth 1A (CMT1A) disease is the most common inherited neuropathy that lacks of therapy and of molecular markers to assess disease severity. Herein, we have pursued the identification of potential biomarkers in plasma samples and skin biopsies that could define the phenotype of CMT1A patients at mild (Mi), moderate (Mo) and severe (Se) stages of disease as assessed by the CMT neuropathy score to contribute to the understanding of CMT pathophysiology and eventually inform of the severity of the disease. Methods: We have used: (i) a high-throughput untargeted metabolomic approach of plasma samples in a cohort of 42 CMT1A patients and 15 healthy controls (CRL) using ultrahigh liquid chromatography coupled to mass spectrometry and (ii) reverse phase protein microarrays to quantitate the expression of some proteins of energy metabolism and of the antioxidant response in skin biopsies of a cohort of 70 CMT1A patients and 13 healthy controls. Results: The metabolomic approach identified 194 metabolites with significant differences among the four groups (Mi, Mo, Se, CRL) of samples. A multivariate Linear Discriminant Analysis model using 12 metabolites afforded the correct classification of the samples. These metabolites indicate an increase in protein catabolism and the mobilization of membrane lipids involved in signaling inflammation with severity of CMT1A. A concurrent depletion of leucine, which is required for the biogenesis of the muscle, is also observed in the patients. Protein expression in skin biopsies indicates early loss of mitochondrial and antioxidant proteins in patients' biopsies. Conclusion: The findings indicate that CMT1A disease is associated with a metabolic state resembling inflammation and sarcopenia suggesting that it might represent a potential target to prevent the nerve and muscle wasting phenotype in these patients. The observed changes in metabolites could be useful as potential biomarkers of CMT1A disease after appropriate validation in future longitudinal studies., ISCIII grant IR11/TREAT-CMT, Instituto de Salud Carlos III (to CC, CM, TS, SIP, CE, FP and JMC
- Published
- 2017
- Full Text
- View/download PDF
40. Mitochondria signal the oncogenic phenotype
- Author
-
José M. Cuezva
- Subjects
education ,Biophysics ,Cell Biology ,Mitochondrion ,Biology ,Phenotype ,Signal ,Biochemistry ,humanities ,Cell biology - Published
- 2014
- Full Text
- View/download PDF
41. Internal-ribosome-entry-site functional activity of the 3′-untranslated region of the mRNA for the β subunit of mitochondrial H+-ATP synthase
- Author
-
José M. Izquierdo and José M. Cuezva
- Subjects
Untranslated region ,Internal ribosome entry site ,Messenger RNA ,Five prime untranslated region ,Three prime untranslated region ,Eukaryotic initiation factor ,Cytoplasmic translation ,EIF4E ,Cell Biology ,Biology ,Molecular Biology ,Biochemistry ,Molecular biology - Abstract
Translation in vitro of the mammalian nucleus-encoded mRNA for the beta subunit of mitochondrial H(+)-ATP synthase (beta-mRNA) of oxidative phosphorylation is promoted by a 150 nt translational enhancer sequence in the 3'-untranslated region (3' UTR). Titration of the eukaryotic initiation factor eIF4E with cap analogue revealed that translation of capped beta-mRNA was pseudo-cap independent. The 3' UTR of beta-mRNA stimulates the translation of heterologous uncapped mRNA species, both when the 3' UTR is placed at the 3' end and at the 5' end of the transcripts. The 3' UTRs of the alpha subunit of mitochondrial H(+)-ATP synthase (alpha-F1-ATPase) and subunit IV of cytochrome c oxidase (COX IV) mRNA species, other nucleus-encoded transcripts of oxidative phosphorylation, do not have the same activity in translation as the 3' UTR of beta-mRNA. On dicistronic RNA species, the 3' UTR of beta-mRNA, and to a smaller extent that of COX IV mRNA, is able to promote the translation of the second cistron to a level comparable to the activity of internal ribosome entry sites (IRESs) described in picornavirus mRNA species. These results indicate that the 3' UTRs of certain mRNA species of oxidative phosphorylation have IRES-like functional activity. Riboprobes of the active 3' UTRs on dicistronic assays formed specific RNA-protein complexes when cross-linked by UV to proteins of the lysate, suggesting that cytoplasmic translation of the mRNA species bearing an active 3' UTR is assisted by specific RNA-protein interactions.
- Published
- 2000
- Full Text
- View/download PDF
42. Short-term exposure of nontumorigenic human bronchial epithelial cells to carcinogenic chromium(VI) compromises their respiratory capacity and alters their bioenergetic signature
- Author
-
Ana M. Urbano, María Sánchez-Aragó, José M. Cuezva, Joana F. Cerveira, Comunidad de Madrid, Fundação para a Ciência e a Tecnologia (Portugal), Ministerio de Ciencia e Innovación (España), and UAM. Departamento de Biología Molecular
- Subjects
Cellular respiration ,Bioenergetics ,DCF, 2′,7′-dichlorofluorescein ,EDTA, ethylenediaminetetracetic acid ,OXPHOS, oxidative phosphorylation ,PBS, phosphate-buffered saline ,TCA, tricarboxylic acid ,Cr(IV), tetravalent chromium ,Oxidative phosphorylation ,General Biochemistry, Genetics and Molecular Biology ,Article ,Cr(III), trivalent chromium ,PI, propidium iodide ,ETC, mitochondrial electron transport chain ,ROS, reactive oxygen species ,2-DG, 2-deoxyglucose ,Glycolysis ,OCR, oxygen consumption rate ,Cr(V), pentavalent chromium ,Cellular energy status ,lcsh:QH301-705.5 ,Glyceraldehyde 3-phosphate dehydrogenase ,Cellular bioenergetic index ,biology ,Cr(VI), hexavalent chromium ,IARC, International Agency for Research on Cancer ,Metabolism ,Biología y Biomedicina / Biología ,Chromate lung cancer ,Biochemistry ,lcsh:Biology (General) ,Anaerobic glycolysis ,GAPDH, glyceraldehyde-3-phosphate dehydrogenase ,biology.protein ,Aerobic glycolysis ,2,4-DNP, 2,4-dinitrophenol ,Warburg effect ,Intracellular ,β-F1-ATPase, catalytic subunit (subunit β) of the mitochondrial H+-ATP synthase - Abstract
Previous studies on the impact of hexavalent chromium [Cr(VI)] on mammalian cell energetics revealed alterations suggestive of a shift to a more fermentative metabolism. Aiming at a more defined understanding of the metabolic effects of Cr(VI) and of their molecular basis, we assessed the impact of a mild Cr(VI) exposure on critical bioenergetic parameters (lactate production, oxygen consumption and intracellular ATP levels). Cells derived from normal human bronchial epithelium (BEAS-2B cell line), the main in vivo target of Cr(VI) carcinogenicity, were subjected for 48h to 1¿M Cr(VI). We could confirm a shift to a more fermentative metabolism, resulting from the simultaneous inhibition of respiration and stimulation of glycolysis. This shift was accompanied by a decrease in the protein levels of the catalytic subunit (subunit ß) of the mitochondrial H+-ATP synthase (ß-F1-ATPase) and a concomitant marked increase in those of glyceraldehyde-3-phosphate dehydrogenase (GAPDH). The corresponding alteration in the ß-F1-ATPase/GAPDH protein ratio (viewed as a bioenergetic signature) upon Cr(VI) exposure was in agreement with the observed attenuation of cellular respiration and enhancement of glycolytic flux. Altogether, these results constitute a novel finding in terms of the molecular mechanisms of Cr(VI) effects. © 2014 The Authors., MICINN (BFU2010-18903) and Comunidad de Madrid (S2011/BMD-2402) (from Spain,to J.M.C.), and CIMAGO (CIMAGO 26/07) and Fundação para a Ciência e a Tecnologia(Portuga)
- Published
- 2014
43. The H+-ATP synthase: A gate to ROS-mediated cell death or cell survival
- Author
-
Inmaculada Martínez-Reyes, José M. Cuezva, and UAM. Departamento de Biología Molecular
- Subjects
Bioenergetics ,Cell Survival ,Biophysics ,Apoptosis ,Oxidative phosphorylation ,Mitochondrion ,Biology ,medicine.disease_cause ,Biochemistry ,Neoplasms ,medicine ,Animals ,Humans ,Cancer ,chemistry.chemical_classification ,Reactive oxygen species ,Reactive oxygen species (ROS) ,Metabolism ,Cell Biology ,H+-ATP synthase ,Biología y Biomedicina / Biología ,Cell biology ,Mitochondria ,Proton-Translocating ATPases ,ROS signaling ,chemistry ,Anaerobic glycolysis ,Signal transduction ,ATPase Inhibitory Factor 1 (IF1) ,Reactive Oxygen Species ,Oxidative stress - Abstract
This article is part of a Special Issue entitled: 18th European Bioenergetic Conference, Cellular oxidative stress results from the increased generation of reactive oxygen species and/or the dysfunction of the antioxidant systems. Most intracellular reactive oxygen species derive from superoxide radical although the majority of the biological effects of reactive oxygen species are mediated by hydrogen peroxide. In this contribution we overview the major cellular sites of reactive oxygen species production, with special emphasis in the mitochondrial pathways. Reactive oxygen species regulate signaling pathways involved in promoting survival and cell death, proliferation, metabolic regulation, the activation of the antioxidant response, the control of iron metabolism and Ca2 + signaling. The reversible oxidation of cysteines in transducers of reactive oxygen species is the primary mechanism of regulation of the activity of these proteins. Next, we present the mitochondrial H+-ATP synthase as a core hub in energy and cell death regulation, defining both the rate of energy metabolism and the reactive oxygen species-mediated cell death in response to chemotherapy. Two main mechanisms that affect the expression and activity of the H+-ATP synthase down-regulate oxidative phosphorylation in prevalent human carcinomas. In this context, we emphasize the prominent role played by the ATPase Inhibitory Factor 1 in human carcinogenesis as an inhibitor of the H+-ATP synthase activity and a mediator of cell survival. The ATPase Inhibitory Factor 1 promotes metabolic rewiring to an enhanced aerobic glycolysis and the subsequent production of mitochondrial reactive oxygen species. The generated reactive oxygen species are able to reprogram the nucleus to support tumor development by arresting cell death. Overall, we discuss the cross-talk between reactive oxygen species signaling and mitochondrial function that is crucial in determining the cellular fate, Work in the authors’ laboratory was supported by grants from the Ministerio de Educación y Ciencia (BFU2010-18903), by the Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), ISCIII and by Comunidad de Madrid (S/2011-BMD-2402), Spain. The CBMSO receives an institutional grant from Fundación Ramón Areces
- Published
- 2014
44. Control of the Translational Efficiency of β-ATPase mRNA Depends on the Regulation of a Protein That Binds the 3′ Untranslated Region of the mRNA
- Author
-
José M. Izquierdo and José M. Cuezva
- Subjects
Untranslated region ,Messenger RNA ,Five prime untranslated region ,Translational efficiency ,Three prime untranslated region ,Translational regulation ,P-bodies ,Protein biosynthesis ,Cell Biology ,Biology ,Molecular Biology ,Molecular biology - Abstract
The expression of the nucleus-encoded beta-F1-ATPase gene of oxidative phosphorylation is developmentally regulated in the liver at both the transcriptional and posttranscriptional levels. In this study we have analyzed the potential mechanisms that control the cytoplasmic expression of beta-F1-ATPase mRNA during liver development. Remarkably, a full-length 3' untranslated region (UTR) of the transcript is required for its efficient in vitro translation. When the 3' UTR of beta-F1-ATPase mRNA is placed downstream of a reporter construct, it functions as a translational enhancer. In vitro translation experiments with full-length beta-F1-ATPase mRNA and with a chimeric reporter construct containing the 3' UTR of beta-F1-ATPase mRNA suggested the existence of an inhibitor of beta-F1-ATPase mRNA translation in the fetal liver. Electrophoretic mobility shift assays and UV cross-linking experiments allowed the identification of an acutely regulated protein (3'betaFBP) of the liver that binds at the 3' UTR of beta-F1-ATPase mRNA. The developmental profile of 3'betaFBP parallels the reported changes in the translational efficiency of beta-F1-ATPase mRNA during development. Fractionation of fetal liver extracts revealed that the inhibitory activity of beta-F1-ATPase mRNA translation cofractionates with 3'-UTR band-shifting activity. Compared to other tissues of the adult rat, kidney and spleen extracts showed very high expression levels of 3'betaFBP. Translation of beta-F1-ATPase mRNA in the presence of kidney and spleen extracts further supported a translational inhibitory role for 3'betaFBP. Mapping experiments and a deletion mutant of the 3' UTR revealed that the cis-acting element for binding 3'betaFBP is located within a highly conserved region of the 3' UTR of mammalian beta-F1-ATPase mRNAs. Overall, we have identified a mechanism of translational control that regulates the rapid postnatal differentiation of liver mitochondria.
- Published
- 1997
- Full Text
- View/download PDF
45. [Untitled]
- Author
-
C M Di Liegro, L.K. Ostronoff, José M. Izquierdo, Javier Ricart, José M. Cuezva, and M López de Heredia
- Subjects
Regulation of gene expression ,Mitochondrial DNA ,Mitochondrial biogenesis ,Physiology ,Gene expression ,Cell Biology ,Oxidative phosphorylation ,Mitochondrion ,Biology ,Phenotype ,Biogenesis ,Cell biology - Abstract
The analysis of the expression of oxidative phosphorylation genes in the liver during development reveals the existence of two biological programs involved in the biogenesis of mitochondria. Differentiation is a short-term program of biogenesis that is controlled at post-transcriptional levels of gene expression and is responsible for the rapid changes in the bioenergetic phenotype of mitochondria. In contrast, proliferation is a long-term program controlled both at the transcriptional and post-transcriptional levels of gene expression and is responsible for the increase in mitochondrial mass in the hepatocyte. Recently, a specific subcellular structure involved in the localization and control of the translation of the mRNA encoding the β-catalytic subunit of the H+-ATP synthase (β-mRNA) has been identified. It is suggested that this structure plays a prominent role in the control of mitochondrial biogenesis at post-transcriptional levels. The fetal liver has many phenotypic manifestations in common with highly glycolytic tumor cells. In addition, both have a low mitochondrial content despite a paradoxical increase in the cellular representation of oxidative phosphorylation transcripts. Based on the paradigm provided by the fetal liver we hypothesize that the aberrant mitochondrial phenotype of fast-growing hepatomas represents a reversion to a fetal program of expression of oxidative phosphorylation genes by the activation, or increased expression, of an inhibitor of β-mRNA translation.
- Published
- 1997
- Full Text
- View/download PDF
46. Impaired mitochondrial oxidative phosphorylation in the peroxisomal disease X-linked adrenoleukodystrophy
- Author
-
Isidro Ferrer, Daniel Cacabelos, Manuel Portero-Otin, Montserrat Ruiz, Jone López-Erauskin, Aurora Pujol, Jordi Boada, Isabel Fabregat, Reinald Pamplona, Francesc Villarroya, Juan José Martínez, Stéphane Fourcade, Jorge Galino, José M. Cuezva, and UAM. Departamento de Biología Molecular
- Subjects
Nervous system ,Mitochondrial ROS ,Free Radicals ,Mice, Transgenic ,Oxidative phosphorylation ,Mitochondrion ,Biology ,medicine.disease_cause ,ATP Binding Cassette Transporter, Subfamily D, Member 1 ,DNA, Mitochondrial ,Oxidative Phosphorylation ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Genetics ,medicine ,Peroxisomes ,Animals ,Humans ,Adrenoleukodystrophy ,Molecular Biology ,Genetics (clinical) ,Cells, Cultured ,030304 developmental biology ,0303 health sciences ,Metabolic disorder ,Fatty Acids ,Brain ,General Medicine ,Peroxisome ,Fibroblasts ,Biología y Biomedicina / Biología ,medicine.disease ,3. Good health ,Cell biology ,Mitochondria ,Mice, Inbred C57BL ,Oxidative Stress ,medicine.anatomical_structure ,Biochemistry ,Spinal Cord ,ATP-Binding Cassette Transporters ,Oxidation-Reduction ,030217 neurology & neurosurgery ,Oxidative stress - Abstract
X-linked adrenoleukodystrophy (X-ALD) is an inherited metabolic disorder of the nervous systemcharacterized by axonopathy in spinal cords and/or cerebral demyelination, adrenal insufficiency and accumulation of very long-chain fatty acids (VLCFAs) in plasma and tissues. The disease is caused by malfunction of the ABCD1 gene, which encodes a peroxisomal transporter of VLCFAs or VLCFA-CoA. In the mouse, Abcd1 loss causes late onset axonal degeneration in the spinal cord, associated with locomotor disability resembling the most common phenotype in patients, adrenomyeloneuropathy. We have formerly shown that an excess of the VLCFA C26:0 induces oxidative damage, which underlies the axonal degeneration exhibited by the Abcd1- mice. In the present study, we sought to investigate the noxious effects of C26:0 on mitochondria function. Ourdata indicate that in X-ALDpatients' fibroblasts,excessof C26:0 generatesmt DNA oxidation and specifically impairs oxidative phosphorylation (OXPHOS) triggering mitochondrialROSproduction from electron transport chain complexes. This correlates with impaired complex V phosphorylative activity, as visualized by high-resolution respirometry on spinal cord slices of Abcd1- mice. Further, we identified a marked oxidation of key OXPHOS system subunits in Abcd1- mouse spinal cords at presymptomatic stages. Altogether, our results illustratesomeof themechanistic intricacies bywhichthe excessof a fatty acid targeted to peroxisomesactivates a deleterious process of oxidative damage to mitochondria, leading to amultifaceted dysfunction of this organelle. These findings may be of relevance for patient management while unveiling novel therapeutic targets for X-ALD. © The Author 2013. Published by Oxford University Press. All rights reserved., European Commission (FP7-241622); European Leukodystrophy Association (ELA2009-036C5, ELA2008-040C4, ELA 2010-020F1); Spanish Institute for Health Carlos III (FIS PI080991 and FIS PI11/01043); Autonomous Government of Catalonia (2009SGR85); Spanish Institute for Health Carlos III (Miguel Servet program CP11/00080); CIBER on Rare Diseases (CIBERER); COST action BM0604 ; Department of Education, Universities and Research of the Basque Country Government (BFI07.126); European Leukodystrophy Association ; Spanish Ministry of Science and Innovation (BFU2009-11879/BFI); Spanish Ministry of Health (PI11/1532); the Autonomous Government of Catalonia (2009SGR735); the ‘La Caixa’ Foundation and COST B35 Action of the European Union. D.C. is a fellow from the Spanish Ministry of Health (FI08-00707); SAF2008-01896 and SAF2011-23636 from the Spanish Ministry of Science and Innovation
- Published
- 2013
47. Expression, regulation and clinical relevance of the ATPase inhibitory factor 1 in human cancers
- Author
-
L. Nájera, Javier Garcia-Bermudez, Laura Formentini, José M. Cuezva, Carmen Navarro, Angeles Juarranz, E C López, Inmaculada Martínez-Reyes, Enrique Espinosa, M. Aldea, María Sánchez-Aragó, Imke M. Willers, J. Clofent, Laura Sánchez-Cenizo, Fulvio Santacatterina, Instituto de Salud Carlos III, Comunidad de Madrid, Fundación Ramón Areces, and Ministerio de Educación y Ciencia (España)
- Subjects
Cancer Research ,ATPase inhibitory factor 1 ,Predictive marker ,Colorectal cancer ,Cancer ,Biology ,Cell cycle ,Mitochondrion ,H+-ATP synthase ,medicine.disease ,medicine.disease_cause ,cancer prognosis ,mitochondria ,ROS signaling ,Anaerobic glycolysis ,energy metabolism ,Immunology ,medicine ,Cancer research ,Original Article ,Protein stabilization ,Carcinogenesis ,Molecular Biology - Abstract
Recent findings in colon cancer cells indicate that inhibition of the mitochondrial Hþ-adenosine triphosphate (ATP) synthase by the ATPase inhibitory factor 1 (IF1) promotes aerobic glycolysis and a reactive oxygen species (ROS)-mediated signal that enhances proliferation and cell survival. Herein, we have studied the expression, biological relevance, mechanism of regulation and potential clinical impact of IF1 in some prevalent human carcinomas. We show that IF1 is highly overexpressed in most (490%) of the colon (n¼64), lung (n¼30), breast (n¼129) and ovarian (n¼10) carcinomas studied as assessed by different approaches in independent cohorts of cancer patients. The expression of IF1 in the corresponding normal tissues is negligible. By contrast, the endometrium, stomach and kidney show high expression of IF1 in the normal tissue revealing subtle differences by carcinogenesis. The overexpression of IF1 also promotes the activation of aerobic glycolysis and a concurrent ROS signal in mitochondria of the lung, breast and ovarian cancer cells mimicking the activity of oligomycin. IF1-mediated ROS signaling activates cell-type specific adaptive responses aimed at preventing death in these cell lines. Remarkably, regulation of IF1 expression in the colon, lung, breast and ovarian carcinomas is exerted at post-transcriptional levels. We demonstrate that IF1 is a short-lived protein (t1/2B100 min) strongly implicating translation and/or protein stabilization as main drivers of metabolic reprogramming and cell survival in these human cancers. Analysis of tumor expression of IF1 in cohorts of breast and colon cancer patients revealed its relevance as a predictive marker for clinical outcome, emphasizing the high potential of IF1 as therapeutic target., This work was supported by grants from the Ministerio de Educacion y Ciencia (BFU2010-18903); the Centro de Investigacion Biomedica en Red de Enfermedades Raras (CIBERER); ISCIII, Madrid and Comunidad de Madrid (S2011/BMD-2402); The CBMSO receives an institutional grant from the Fundacion Ramon Areces
- Published
- 2013
48. Hypothyroidism Affects the Expression of the beta-F1-ATPase Gene and Limits Mitochondrial Proliferation in Rat Liver at All Stages of Development
- Author
-
José M. Izquierdo, José M. Cuezva, and Eugenio Jiménez
- Subjects
Thyroid Hormones ,endocrine system ,medicine.medical_specialty ,endocrine system diseases ,Mitochondria, Liver ,Mitochondrion ,Biochemistry ,Hypothyroidism ,Pregnancy ,Internal medicine ,Organelle ,medicine ,Animals ,Euthyroid ,RNA, Messenger ,Rats, Wistar ,Beta (finance) ,ATP synthase ,biology ,ATPase Gene ,Gene Expression Regulation, Developmental ,Animals, Suckling ,Rats ,Proton-Translocating ATPases ,Endocrinology ,Mitochondrial biogenesis ,biology.protein ,Female ,hormones, hormone substitutes, and hormone antagonists ,ATP synthase alpha/beta subunits - Abstract
In order to analyze the role of thyroid hormones in mitochondrial biogenesis, we have studied the expression pattern of the beta subunit of the mitochondrial ATP-synthase complex in liver and in isolated mitochondria during postnatal development of hypothyroid rats. Chemically induced hypothyroidism promoted a significant reduction in body and liver masses at all stages of development. Furthermore, plasma 3,5,3'-triiodo-L-thyronine (T3) and 3,5,3',5'-tetraiodo-L-thyronine (T4) concentrations were significantly reduced in hypothyroid animals when compared to euthyroid animals. Remarkably, steady-state beta-F1-ATPase mRNA levels in livers of hypothyroid animals showed an approximately 50% reduction when compared to age-matched euthyroid rats at all stages of development. The relative amounts of beta-F1-ATPase protein determined in isolated mitochondria of 1-day-old and adult hypothyroid animals were similar to those determined in mitochondria of age-matched euthyroids, indicating that hypothyroidism does not affect organelle differentiation in the liver of suckling and adult rats. In contrast, the relative amount of beta-F1-ATPase protein in liver homogenates varied (0-30% reduction) due to the hypothyroid condition during development. These findings suggest the existence of compensatory mechanisms operating at the translational and/or post-translational levels which promote proliferation of mitochondria in the hypothyroid liver. However, when the liver mass was considered, hypothyroidism significantly reduced overall mitochondrial proliferation in rat liver. Interestingly, the effects of thyroid hormones on the biogenesis of the ATP synthase complex at latter stages of development provide an example in which the hypothyroid condition limits the expression of the nuclear-encoded gene with no apparent effect on the expression of the mitochondrial-encoded genes (ATP synthase subunits 6-8).
- Published
- 1995
- Full Text
- View/download PDF
49. ATPase Inhibitory Factor 1 (IF1): A main driver of metabolic reprogramming and nuclear signaling in cancer
- Author
-
Pau B. Esparza, Cristina Nuevo-Tapioles, Beatriz Soldevilla, José M. Cuezva, Fulvio Santacatterina, Lucía González, and Laura Formentini
- Subjects
ATPase INHIBITORY FACTOR 1 ,Metabolic reprogramming ,Biophysics ,medicine ,Cancer ,Cell Biology ,Biology ,medicine.disease ,Biochemistry ,Cell biology - Published
- 2016
- Full Text
- View/download PDF
50. Mitochondria-mediated energy adaption in cancer: the H(+)-ATP synthase-geared switch of metabolism in human tumors
- Author
-
María Sánchez-Aragó, Laura Formentini, José M. Cuezva, and UAM. Departamento de Biología Molecular
- Subjects
Programmed cell death ,Physiology ,Clinical Biochemistry ,Oxidative phosphorylation ,Mitochondrion ,Biochemistry ,Mitochondrial Proton-Translocating ATPases ,Oxidative Phosphorylation ,Neoplasms ,medicine ,Tumor Microenvironment ,Animals ,Humans ,Molecular Biology ,General Environmental Science ,ATP synthase ,biology ,Cell Death ,Tumor Suppressor Proteins ,Carcinoma ,Cancer ,Cell Biology ,medicine.disease ,Biología y Biomedicina / Biología ,Forum Review Articles ,Cell biology ,Mitochondria ,Anaerobic glycolysis ,Cancer cell ,biology.protein ,General Earth and Planetary Sciences ,Energy Metabolism - Abstract
Significance: Since the signing of the National Cancer Act in 1971, cancer still remains a major cause of death despite significant progresses made in understanding the biology and treatment of the disease. After many years of ostracism, the peculiar energy metabolism of tumors has been recognized as an additional phenotypic trait of the cancer cell. Recent Advances: While the enhanced aerobic glycolysis of carcinomas has already been translated to bedside for precise tumor imaging and staging of cancer patients, accepting that an impaired bioenergetic function of mitochondria is pivotal to understand energy metabolism of tumors and in its progression is debated. However, mitochondrial bioenergetics and cell death are tightly connected. Critical Issues: Recent clinical findings indicate that H+-ATP synthase, a core component of mitochondrial oxidative phosphorylation, is repressed at both the protein and activity levels in human carcinomas. This review summarizes the relevance that mitochondrial function has to understand energy metabolism of tumors and explores the connection between the bioenergetic function of the organelle and the activity of mitochondria as tumor suppressors. Future Directions: The reversible nature of energy metabolism in tumors highlights the relevance that the microenvironment has for tumor progression. Moreover, the stimulation of mitochondrial activity or the inhibition of glycolysis suppresses tumor growth. Future research should elucidate the mechanisms promoting the silencing of oxidative phosphorylation in carcinomas. The aim is the development of new therapeutic strategies tackling energy metabolism to eradicate tumors or at least, to maintain tumor dormancy and transform cancer into a chronic disease. Antioxid. Redox Signal. 19, 285-298, Supported by JCI2009-03918 Juan de la Cierva Grant, Ministerio de Educación y Ciencia, Spain. Work in the authors’ laboratory was supported by grants from the Ministerio de Educación y Ciencia (BFU2010-18903), by the Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), ISCIII and by Comunidad de Madrid (S2011/BMD-2402), Spain. The CBMSO receives an institutional grant from Fundación Ramón Areces
- Published
- 2012
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.