Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
1.
Hum Mol Genet ; 30(24): 2441-2455, 2021 11 30.
Article in English | MEDLINE | ID: mdl-34274972

ABSTRACT

Charcot-Marie-Tooth (CMT) disease is a neuropathy that lacks effective therapy. CMT patients show degeneration of peripheral nerves, leading to muscle weakness and loss of proprioception. Loss of mitochondrial oxidative phosphorylation proteins and enzymes of the antioxidant response accompany degeneration of nerves in skin biopsies of CMT patients. Herein, we followed a drug-repurposing approach to find drugs in a Food and Drug Administration-approved library that could prevent development of CMT disease in the Gdap1-null mouse model. We found that the antibiotic florfenicol is a mitochondrial uncoupler that prevents the production of reactive oxygen species and activates respiration in human GDAP1-knockdown neuroblastoma cells and in dorsal root ganglion neurons of Gdap1-null mice. Treatment of CMT-affected Gdap1-null mice with florfenicol has no beneficial effect in the course of the disease. However, administration of florfenicol, or the antioxidant MitoQ, to pre-symptomatic GDAP1-null mice prevented weight gain and ameliorated the motor coordination deficiencies that developed in the Gdap1-null mice. Interestingly, both florfenicol and MitoQ halted the decay in mitochondrial and redox proteins in sciatic nerves of Gdap1-null mice, supporting that oxidative damage is implicated in the etiology of the neuropathy. These findings support the development of clinical trials for translation of these drugs for treatment of CMT patients.


Subject(s)
Charcot-Marie-Tooth Disease , Animals , Charcot-Marie-Tooth Disease/drug therapy , Charcot-Marie-Tooth Disease/genetics , Humans , Mice , Mice, Knockout , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Mutation , Nerve Tissue Proteins/genetics
2.
FASEB J ; 33(2): 1836-1851, 2019 02.
Article in English | MEDLINE | ID: mdl-30204502

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.


Subject(s)
Proteins/physiology , RNA Processing, Post-Transcriptional , Animals , Cell Line , Female , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Phosphorylation , Proteins/genetics , Proteins/metabolism , RNA, Messenger/genetics , ATPase Inhibitory Protein
3.
EMBO J ; 33(7): 762-78, 2014 Apr 01.
Article in English | MEDLINE | ID: mdl-24521670

ABSTRACT

A key transducer in energy conservation and signaling cell death is the mitochondrial H(+)-ATP synthase. The expression of the ATPase inhibitory factor 1 (IF1) is a strategy used by cancer cells to inhibit the activity of the H(+)-ATP synthase to generate a ROS signal that switches on cellular programs of survival. We have generated a mouse model expressing a mutant of human IF1 in brain neurons to assess the role of the H(+)-ATP synthase in cell death in vivo. The expression of hIF1 inhibits the activity of oxidative phosphorylation and mediates the shift of neurons to an enhanced aerobic glycolysis. Metabolic reprogramming induces brain preconditioning affording protection against quinolinic acid-induced excitotoxicity. Mechanistically, preconditioning involves the activation of the Akt/p70S6K and PARP repair pathways and Bcl-xL protection from cell death. Overall, our findings provide the first in vivo evidence highlighting the H(+)-ATP synthase as a target to prevent neuronal cell death.


Subject(s)
Gene Expression Regulation, Enzymologic , Mitochondrial Proton-Translocating ATPases/antagonists & inhibitors , Proteins/genetics , Signal Transduction , Animals , Apoptosis , Behavior, Animal , Brain/cytology , Brain/drug effects , Brain/enzymology , Glycolysis/drug effects , Humans , Male , Metabolic Networks and Pathways , Mice , Mice, Transgenic , Mitochondria/drug effects , Mitochondria/enzymology , Mitochondrial Proton-Translocating ATPases/metabolism , Models, Animal , Mutation, Missense , Neurons/cytology , Neurons/drug effects , Neurons/enzymology , Neurotoxins/pharmacology , Oxidative Phosphorylation , Promoter Regions, Genetic/genetics , Proteins/metabolism , Quinolinic Acid/pharmacology , Reactive Oxygen Species/metabolism , ATPase Inhibitory Protein
4.
J Transl Med ; 15(1): 29, 2017 02 10.
Article in English | MEDLINE | ID: mdl-28183315

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 ß-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.


Subject(s)
Carcinogenesis/metabolism , Dermatomyositis/metabolism , Mitochondria/metabolism , Proteins/metabolism , Pyruvate Kinase/metabolism , Antibodies/metabolism , Biomarkers/metabolism , Biopsy , Cluster Analysis , Energy Metabolism , Humans , Inflammation/diagnosis , Inflammation/pathology , Muscles/metabolism , Muscles/pathology , Protein Array Analysis , Reproducibility of Results , Subcellular Fractions/metabolism , ATPase Inhibitory Protein
5.
J Transl Med ; 13: 65, 2015 Feb 18.
Article in English | MEDLINE | ID: mdl-25880557

ABSTRACT

BACKGROUND: 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.


Subject(s)
Biomarkers/metabolism , Energy Metabolism , Neuromuscular Diseases/metabolism , Protein Array Analysis/methods , Proteomics/methods , Animals , Antibodies/metabolism , Biopsy , Female , Humans , Male , Mice, Inbred BALB C , Muscles/pathology , Neuromuscular Diseases/diagnosis , Rare Diseases/metabolism , Reproducibility of Results
6.
EMBO Rep ; 14(7): 638-44, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23722655

ABSTRACT

Differentiation of human mesenchymal stem cells (hMSCs) requires the rewiring of energy metabolism. Herein, we demonstrate that the ATPase inhibitory factor 1 (IF1) is expressed in hMSCs and in prostate and colon stem cells but is not expressed in the differentiated cells. IF1 inhibits oxidative phosphorylation and regulates the activity of aerobic glycolysis in hMSCs. Silencing of IF1 in hMSCs mimics the metabolic changes observed in osteocytes and accelerates cellular differentiation. Activation of IF1 degradation acts as the switch that regulates energy metabolism during differentiation. We conclude that IF1 is a stemness marker important for maintaining the quiescence state.


Subject(s)
Energy Metabolism , Mesenchymal Stem Cells/metabolism , Osteocytes/metabolism , Proteins/genetics , Stem Cells/metabolism , Biomarkers/metabolism , Cell Differentiation , Cells, Cultured , Colon/cytology , Colon/metabolism , Gene Expression Regulation , Gene Silencing , Glycolysis , Humans , Male , Mesenchymal Stem Cells/cytology , Osteocytes/cytology , Osteogenesis/genetics , Oxidative Phosphorylation , Prostate/cytology , Prostate/metabolism , Proteins/antagonists & inhibitors , Proteins/metabolism , Proteolysis , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Stem Cells/cytology , ATPase Inhibitory Protein
7.
Plast Reconstr Surg Glob Open ; 12(5): e5792, 2024 May.
Article in English | MEDLINE | ID: mdl-38726041

ABSTRACT

Background: Augmentation and reshaping of body volume, particularly in the gluteal area, presents a significant challenge in aesthetic surgery. Hyaluronic acid (HA) fillers have emerged as an effective and safe tool for such indications, but literature examining nonsurgical gluteal reshaping with HA remains limited. This study aims to evaluate the long-term safety of using recommended volumes of HA body fillers for nonsurgical gluteal augmentation. Methods: A retrospective, observational study was carried out across multiple centers in Italy and the United Arab Emirates. The study involved participants between 22 and 53 years of age who underwent gluteal augmentation using HA body filler (HYAcorp MLF1/2) between 2017 and 2021, with up to 4 years and 7 months of follow-up. Participants and investigators independently evaluated the procedure's effectiveness by comparing pre- and posttreatment photographs. The Global Aesthetic Improvement Scale was used to assess posttreatment satisfaction by both participants and investigators. All adverse effects (AEs) were recorded. Results: The study included a diverse group of 91 participants. No serious adverse events were reported, with the majority of AE occurring shortly after treatment and resolving in 1 week. AEs were more frequently observed in participants with previous treatments using different substances in the treatment area. Conclusions: The real-world application of HA body filler (HYAcorp MLF1/2) for gluteal augmentation in the participants of this study showed the treatment's effectiveness, with no severe adverse events reported among the participants. High levels of satisfaction were reported among both participants and investigators.

8.
Cell Death Dis ; 14(7): 413, 2023 07 12.
Article in English | MEDLINE | ID: mdl-37433784

ABSTRACT

ATPase Inhibitory Factor 1 (IF1) regulates the activity of mitochondrial ATP synthase. The expression of IF1 in differentiated human and mouse cells is highly variable. In intestinal cells, the overexpression of IF1 protects against colon inflammation. Herein, we have developed a conditional IF1-knockout mouse model in intestinal epithelium to investigate the role of IF1 in mitochondrial function and tissue homeostasis. The results show that IF1-ablated mice have increased ATP synthase/hydrolase activities, leading to profound mitochondrial dysfunction and a pro-inflammatory phenotype that impairs the permeability of the intestinal barrier compromising mouse survival upon inflammation. Deletion of IF1 prevents the formation of oligomeric assemblies of ATP synthase and alters cristae structure and the electron transport chain. Moreover, lack of IF1 promotes an intramitochondrial Ca2+ overload in vivo, minimizing the threshold to Ca2+-induced permeability transition (mPT). Removal of IF1 in cell lines also prevents the formation of oligomeric assemblies of ATP synthase, minimizing the threshold to Ca2+-induced mPT. Metabolomic analyses of mice serum and colon tissue highlight that IF1 ablation promotes the activation of de novo purine and salvage pathways. Mechanistically, lack of IF1 in cell lines increases ATP synthase/hydrolase activities and installs futile ATP hydrolysis in mitochondria, resulting in the activation of purine metabolism and in the accumulation of adenosine, both in culture medium and in mice serum. Adenosine, through ADORA2B receptors, promotes an autoimmune phenotype in mice, stressing the role of the IF1/ATP synthase axis in tissue immune responses. Overall, the results highlight that IF1 is required for ATP synthase oligomerization and that it acts as a brake to prevent ATP hydrolysis under in vivo phosphorylating conditions in intestinal cells.


Subject(s)
Adenosine , Inflammation , Mitochondrial Proteins , Animals , Humans , Mice , Adenosine Triphosphate , Cell Differentiation , Mice, Knockout , Mitochondrial Proton-Translocating ATPases/metabolism , Mitochondrial Proteins/metabolism , ATPase Inhibitory Protein
9.
Oncogenesis ; 11(1): 24, 2022 May 09.
Article in English | MEDLINE | ID: mdl-35534478

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.

10.
Biochim Biophys Acta Mol Basis Dis ; 1866(5): 165721, 2020 05 01.
Article in English | MEDLINE | ID: mdl-32057942

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.


Subject(s)
Antineoplastic Agents/therapeutic use , Biomarkers, Tumor/metabolism , Carcinoma/pathology , Energy Metabolism/drug effects , Mitochondria/enzymology , Animals , Antineoplastic Agents/pharmacology , Biomarkers, Tumor/genetics , Carcinogenesis/drug effects , Carcinogenesis/metabolism , Carcinoma/drug therapy , Carcinoma/genetics , Carcinoma/mortality , Disease Models, Animal , Disease Progression , Down-Regulation , Energy Metabolism/genetics , Gene Expression Regulation, Neoplastic , Glycolysis/drug effects , Glycolysis/genetics , Humans , Lipogenesis/drug effects , Lipogenesis/genetics , Mitochondria/drug effects , Mutation , Oxidative Phosphorylation/drug effects , Oxidative Stress/drug effects , Oxidative Stress/genetics , Prognosis , Reactive Oxygen Species/metabolism , Survival Rate , Up-Regulation
11.
Nat Commun ; 11(1): 3606, 2020 07 17.
Article in English | MEDLINE | ID: mdl-32681016

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 ß1-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.


Subject(s)
Adrenergic Antagonists/pharmacology , Angiogenesis Inducing Agents/pharmacology , Breast Neoplasms/physiopathology , Colonic Neoplasms/metabolism , Colonic Neoplasms/physiopathology , Mitochondria/drug effects , Nebivolol/pharmacology , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Colonic Neoplasms/drug therapy , Colonic Neoplasms/genetics , Female , Humans , Male , Mitochondria/genetics , Mitochondria/metabolism , Mitochondrial Proton-Translocating ATPases/genetics , Mitochondrial Proton-Translocating ATPases/metabolism , NADH Dehydrogenase/genetics , NADH Dehydrogenase/metabolism , Oxidative Phosphorylation/drug effects , Proteins/genetics , Proteins/metabolism , ATPase Inhibitory Protein
12.
Cancers (Basel) ; 12(1)2019 Dec 19.
Article in English | MEDLINE | ID: mdl-31861681

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.

13.
Free Radic Biol Med ; 126: 235-248, 2018 10.
Article in English | MEDLINE | ID: mdl-30138712

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.


Subject(s)
DNA, Mitochondrial/genetics , MELAS Syndrome/metabolism , Mitochondrial Diseases/metabolism , Ophthalmoplegia, Chronic Progressive External/metabolism , Adolescent , Adult , Aged , Antioxidants/metabolism , Biomarkers/metabolism , Biopsy , Child , Child, Preschool , Gene Expression Regulation , Glycolysis , Healthy Volunteers , Humans , MELAS Syndrome/genetics , MELAS Syndrome/pathology , Middle Aged , Mitochondrial Diseases/genetics , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Ophthalmoplegia, Chronic Progressive External/genetics , Ophthalmoplegia, Chronic Progressive External/pathology , Oxidative Stress , Reactive Oxygen Species , Superoxide Dismutase/genetics , Support Vector Machine , Young Adult
14.
Stem Cell Reports ; 11(6): 1479-1492, 2018 12 11.
Article in English | MEDLINE | ID: mdl-30472011

ABSTRACT

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.


Subject(s)
Cellular Reprogramming , Hybrid Cells/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Animals , CDC2 Protein Kinase/metabolism , Glycolysis , Humans , Membrane Potential, Mitochondrial , Mice, Inbred C57BL , Mitochondria/metabolism , Mitochondrial Dynamics , Oxidative Phosphorylation , Phosphorylation
15.
Cell Rep ; 19(6): 1202-1213, 2017 05 09.
Article in English | MEDLINE | ID: mdl-28494869

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.


Subject(s)
Colitis, Ulcerative/immunology , Intestines/immunology , Macrophage Activation , Macrophages/immunology , Mitochondria/metabolism , Reactive Oxygen Species/metabolism , Animals , Cells, Cultured , Immunity, Innate , Intestines/cytology , Macrophages/cytology , Mice , Mice, Inbred C57BL , Mitochondrial Proton-Translocating ATPases/metabolism , NF-kappa B/metabolism , Phenotype , Proto-Oncogene Proteins c-akt/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/metabolism
16.
PLoS One ; 12(6): e0178376, 2017.
Article in English | MEDLINE | ID: mdl-28575008

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.


Subject(s)
Charcot-Marie-Tooth Disease/blood , Charcot-Marie-Tooth Disease/metabolism , Metabolome , Proteins/analysis , Skin/pathology , Adult , Biomarkers/analysis , Biomarkers/blood , Biomarkers/metabolism , Charcot-Marie-Tooth Disease/pathology , Energy Metabolism , Humans , Metabolomics , Middle Aged , Prospective Studies , Proteins/metabolism , Skin/metabolism
17.
Oncotarget ; 7(1): 490-508, 2016 Jan 05.
Article in English | MEDLINE | ID: mdl-26595676

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.


Subject(s)
Down-Regulation , Liver Neoplasms/metabolism , Liver/metabolism , Oxidative Phosphorylation , Proteins/metabolism , AMP-Activated Protein Kinases/metabolism , Acetaminophen/pharmacology , Animals , Apoptosis/drug effects , Apoptosis/genetics , Blotting, Western , Cell Survival/genetics , Gene Expression , Humans , Liver/pathology , Liver/ultrastructure , Liver Neoplasms/genetics , Mice, Transgenic , Microscopy, Electron , Microscopy, Fluorescence , Mitochondria/genetics , Mitochondria/metabolism , Mutation , Proteins/genetics , Reverse Transcriptase Polymerase Chain Reaction , p38 Mitogen-Activated Protein Kinases/metabolism , ATPase Inhibitory Protein
SELECTION OF CITATIONS
SEARCH DETAIL