Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters











Publication year range
1.
Immunity ; 55(11): 2059-2073.e8, 2022 11 08.
Article in English | MEDLINE | ID: mdl-36351375

ABSTRACT

T memory stem cells (TSCM) display increased self-renewal and prolonged survival capabilities, thus preventing T cell exhaustion and promoting effective anti-tumor T cell responses. TSCM cells can be expanded by Urolithin A (UA), which is produced by the commensal gut microbiome from foods rich in ellagitannins and is known to improve mitochondrial health. Oral UA administration to tumor-bearing mice conferred strong anti-tumor CD8+ T cell immunity, whereas ex vivo UA pre-treated T cells displayed improved anti-tumor function upon adoptive cell transfer. UA-induced TSCM formation depended on Pink1-mediated mitophagy triggering cytosolic release of the mitochondrial phosphatase Pgam5. Cytosolic Pgam5 dephosphorylated ß-catenin, which drove Wnt signaling and compensatory mitochondrial biogenesis. Collectively, we unravel a critical signaling pathway linking mitophagy to TSCM formation and suggest that the well-tolerated metabolic compound UA represents an attractive option to improve immune therapy.


Subject(s)
Coumarins , Mitophagy , Mice , Animals , Coumarins/pharmacology , Wnt Signaling Pathway , Stem Cells , Immunologic Memory
2.
Aging Cell ; 21(8): e13662, 2022 08.
Article in English | MEDLINE | ID: mdl-35778837

ABSTRACT

Osteoarthritis (OA) is the most common age-related joint disorder with no effective therapy. According to the World Health Organization, OA affects over 500 million people and is characterized by degradation of cartilage and other joint tissues, severe pain, and impaired mobility. Mitochondrial dysfunction contributes to OA pathology. However, interventions to rescue mitochondrial defects in human OA are not available. Urolithin A (Mitopure) is a natural postbiotic compound that promotes mitophagy and mitochondrial function and beneficially impacts muscle health in preclinical models of aging and in elderly and middle-aged humans. Here, we showed that Urolithin A improved mitophagy and mitochondrial respiration in primary chondrocytes from joints of both healthy donors and OA patients. Furthermore, Urolithin A reduced disease progression in a mouse model of OA, decreasing cartilage degeneration, synovial inflammation, and pain. These improvements were associated with increased mitophagy and mitochondrial content, in joints of OA mice. These findings indicate that UA promotes joint mitochondrial health, alleviates OA pathology, and supports Urolithin A's potential to improve mobility with beneficial effects on structural damage in joints.


Subject(s)
Cartilage, Articular , Osteoarthritis, Knee , Aged , Animals , Cartilage, Articular/metabolism , Chondrocytes/metabolism , Coumarins , Humans , Mice , Middle Aged , Mitochondria/metabolism , Osteoarthritis, Knee/drug therapy , Osteoarthritis, Knee/metabolism , Osteoarthritis, Knee/pathology , Pain/metabolism
3.
Cell Rep Med ; 3(5): 100633, 2022 05 17.
Article in English | MEDLINE | ID: mdl-35584623

ABSTRACT

Targeting mitophagy to activate the recycling of faulty mitochondria during aging is a strategy to mitigate muscle decline. We present results from a randomized, placebo-controlled trial in middle-aged adults where we administer a postbiotic compound Urolithin A (Mitopure), a known mitophagy activator, at two doses for 4 months (NCT03464500). The data show significant improvements in muscle strength (∼12%) with intake of Urolithin A. We observe clinically meaningful improvements with Urolithin A on aerobic endurance (peak oxygen oxygen consumption [VO2]) and physical performance (6 min walk test) but do not notice a significant improvement on peak power output (primary endpoint). Levels of plasma acylcarnitines and C-reactive proteins are significantly lower with Urolithin A, indicating higher mitochondrial efficiency and reduced inflammation. We also examine expression of proteins linked to mitophagy and mitochondrial metabolism in skeletal muscle and find a significant increase with Urolithin A administration. This study highlights the benefit of Urolithin A to improve muscle performance.


Subject(s)
Mitophagy , Muscle Strength , Biomarkers , Coumarins , Mitochondria
4.
Eur J Clin Nutr ; 76(2): 297-308, 2022 02.
Article in English | MEDLINE | ID: mdl-34117375

ABSTRACT

BACKGROUND: Urolithin A (UA) is produced by gut microflora from foods rich in ellagitannins. UA has been shown to improve mitochondrial health preclinically and in humans. Not everyone has a microbiome capable of producing UA, making supplementation with UA an appealing strategy. OBJECTIVE: This is the first detailed investigation of the prevalence of UA producers in a healthy population and the ability of direct UA supplementation to overcome both microbiome and dietary variability. Dietary intake of a glass of pomegranate juice (PJ) was used to assess UA producer status (n = 100 participants) and to characterize differences in gut microbiome between UA producers from non-producers. METHODS: Subjects were randomized (1:1) to either PJ or a food product containing UA (500 mg). Prevalence of UA producers and non-producers were determined in the PJ group. Diet questionnaires and fecal samples were collected to compare differences between UA producers and non-producers along with plasma samples at different time points to assess levels of UA and its conjugates between the interventions. RESULTS: Only 12% of subjects had detectable levels of UA at baseline. Following PJ intake ~40% of the subjects converted significantly the precursor compounds into UA. UA producers were distinguished by a significantly higher gut microbiome diversity and ratio of Firmicutes to Bacteroides. Direct supplementation with UA significantly increased plasma levels and provided a >6-fold exposure to UA vs. PJ (p < 0.0001). CONCLUSIONS: Differences in gut microbiome and diet that dictate natural exposure to UA can be overcome via direct dietary UA supplementation.


Subject(s)
Gastrointestinal Microbiome , Adult , Coumarins/pharmacology , Dietary Exposure , Dietary Supplements , Humans
5.
Trends Mol Med ; 27(7): 687-699, 2021 07.
Article in English | MEDLINE | ID: mdl-34030963

ABSTRACT

Urolithin A (UA) is a natural compound produced by gut bacteria from ingested ellagitannins (ETs) and ellagic acid (EA), complex polyphenols abundant in foods such as pomegranate, berries, and nuts. UA was discovered 40 years ago, but only recently has its impact on aging and disease been explored. UA enhances cellular health by increasing mitophagy and mitochondrial function and reducing detrimental inflammation. Several preclinical studies show how UA protects against aging and age-related conditions affecting muscle, brain, joints, and other organs. In humans, benefits of UA supplementation in the muscle are supported by recent clinical trials in elderly people. Here, we review the state of the art of UA's biology and its translational potential as a nutritional intervention in humans.


Subject(s)
Aging , Biological Products/pharmacology , Coumarins/pharmacology , Macular Degeneration/drug therapy , Mental Disorders/drug therapy , Humans
6.
Sci Transl Med ; 13(588)2021 04 07.
Article in English | MEDLINE | ID: mdl-33827972

ABSTRACT

Duchenne muscular dystrophy (DMD) is the most common muscular dystrophy, and despite advances in genetic and pharmacological disease-modifying treatments, its management remains a major challenge. Mitochondrial dysfunction contributes to DMD, yet the mechanisms by which this occurs remain elusive. Our data in experimental models and patients with DMD show that reduced expression of genes involved in mitochondrial autophagy, or mitophagy, contributes to mitochondrial dysfunction. Mitophagy markers were reduced in skeletal muscle and in muscle stem cells (MuSCs) of a mouse model of DMD. Administration of the mitophagy activator urolithin A (UA) rescued mitophagy in DMD worms and mice and in primary myoblasts from patients with DMD, increased skeletal muscle respiratory capacity, and improved MuSCs' regenerative ability, resulting in the recovery of muscle function and increased survival in DMD mouse models. These data indicate that restoration of mitophagy alleviates symptoms of DMD and suggest that UA may have potential therapeutic applications for muscular dystrophies.


Subject(s)
Mitophagy , Muscular Dystrophy, Duchenne , Animals , Coumarins , Humans , Mice , Mice, Inbred mdx , Muscle, Skeletal , Muscular Dystrophy, Duchenne/drug therapy
7.
Aging (Albany NY) ; 12(24): 24484-24503, 2020 12 30.
Article in English | MEDLINE | ID: mdl-33378272

ABSTRACT

Aging is emerging as a druggable target with growing interest from academia, industry and investors. New technologies such as artificial intelligence and advanced screening techniques, as well as a strong influence from the industry sector may lead to novel discoveries to treat age-related diseases. The present review summarizes presentations from the 7th Annual Aging Research and Drug Discovery (ARDD) meeting, held online on the 1st to 4th of September 2020. The meeting covered topics related to new methodologies to study aging, knowledge about basic mechanisms of longevity, latest interventional strategies to target the aging process as well as discussions about the impact of aging research on society and economy. More than 2000 participants and 65 speakers joined the meeting and we already look forward to an even larger meeting next year. Please mark your calendars for the 8th ARDD meeting that is scheduled for the 31st of August to 3rd of September, 2021, at Columbia University, USA.


Subject(s)
Aging , Artificial Intelligence , Biomedical Research , Longevity , Cellular Senescence , Congresses as Topic , Drug Discovery , Humans , Life Style , Pharmaceutical Preparations
8.
Sci Rep ; 9(1): 17821, 2019 Nov 25.
Article in English | MEDLINE | ID: mdl-31767924

ABSTRACT

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

9.
Nat Metab ; 1(6): 595-603, 2019 06.
Article in English | MEDLINE | ID: mdl-32694802

ABSTRACT

Urolithin A (UA) is a natural dietary, microflora-derived metabolite shown to stimulate mitophagy and improve muscle health in old animals and in preclinical models of aging1. Here, we report the results of a first-in-human clinical trial in which we administered UA, either as a single dose or as multiple doses over a 4-week period, to healthy, sedentary elderly individuals. We show that UA has a favourable safety profile (primary outcome). UA was bioavailable in plasma at all doses tested, and 4 weeks of treatment with UA at doses of 500 mg and 1,000 mg modulated plasma acylcarnitines and skeletal muscle mitochondrial gene expression in elderly individuals (secondary outcomes). These observed effects on mitochondrial biomarkers show that UA induces a molecular signature of improved mitochondrial and cellular health following regular oral consumption in humans.


Subject(s)
Coumarins/pharmacology , Mitochondria/drug effects , Mitophagy/drug effects , Aged , Coumarins/administration & dosage , Dose-Response Relationship, Drug , Humans , Sedentary Behavior
10.
J Cell Physiol ; 234(5): 6601-6610, 2019 05.
Article in English | MEDLINE | ID: mdl-30417335

ABSTRACT

Proper mitochondrial function plays a central role in cellular metabolism. Various diseases as well as aging are associated with diminished mitochondrial function. Previously, we identified 19 miRNAs putatively involved in the regulation of mitochondrial metabolism in skeletal muscle, a highly metabolically active tissue. In the current study, these 19 miRNAs were individually silenced in C2C12 myotubes using antisense oligonucleotides, followed by measurement of the expression of 27 genes known to play a major role in regulating mitochondrial metabolism. Based on the outcomes, we then focused on miR-382-5p and identified pathways affected by its silencing using microarrays, investigated protein expression, and studied cellular respiration. Silencing of miRNA-382-5p significantly increased the expression of several genes involved in mitochondrial dynamics and biogenesis. Conventional microarray analysis in C2C12 myotubes silenced for miRNA-382-5p revealed a collective downregulation of mitochondrial ribosomal proteins and respiratory chain proteins. This effect was accompanied by an imbalance between mitochondrial proteins encoded by the nuclear and mitochondrial DNA (1.35-fold, p < 0.01) and an induction of HSP60 protein (1.31-fold, p < 0.05), indicating activation of the mitochondrial unfolded protein response (mtUPR). Furthermore, silencing of miR-382-5p reduced basal oxygen consumption rate by 14% ( p < 0.05) without affecting mitochondrial content, pointing towards a more efficient mitochondrial function as a result of improved mitochondrial quality control. Taken together, silencing of miR-382-5p induces a mitonuclear protein imbalance and activates the mtUPR in skeletal muscle, a phenomenon that was previously associated with improved longevity.


Subject(s)
MicroRNAs/genetics , Mitochondria, Muscle/metabolism , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Muscle Fibers, Skeletal/metabolism , Animals , Mice , Muscle, Skeletal/metabolism , Ribosomal Proteins/metabolism , Unfolded Protein Response/genetics
11.
Sci Rep ; 8(1): 8548, 2018 06 04.
Article in English | MEDLINE | ID: mdl-29867098

ABSTRACT

Aging is accompanied by a gradual decline in both muscle mass and strength over time, which can eventually lead to pathologies, such as frailty and sarcopenia. While these two conditions are well characterized, further investigation of the early biological signs present in pre-frail elderly is still needed to help identify strategies for preventative therapeutic intervention. The goal of the present clinical study was to evaluate the level of mitochondrial (dys)function in a well-defined population of pre-frail elderly (>60 years of age). Pre-frail elderly were compared with an age-matched population of active elderly. Muscle mitochondrial function was assessed in vivo using phosphorus magnetic resonance spectroscopy (31P-MRS) and a comprehensive set of biological biomarkers were measured ex vivo in vastus lateralis muscle biopsies. In pre-frail subjects, phosphocreatine recovery was impaired and mitochondrial respiratory complex protein and activity levels were significantly lower when compared with active elderly. Analysis of microarray data showed that mitochondrial genes were also significantly down-regulated in muscle of pre-frail compared to active elderly. These results show that mitochondrial impairment is a hallmark of pre-frailty development and the onset of decline in muscle function in the elderly.


Subject(s)
Aging/metabolism , Mitochondria, Muscle/metabolism , Muscle, Skeletal/metabolism , Sarcopenia/metabolism , Aged , Aged, 80 and over , Aging/pathology , Biomarkers/metabolism , Case-Control Studies , Female , Humans , Magnetic Resonance Spectroscopy , Male , Middle Aged , Mitochondria, Muscle/pathology , Muscle, Skeletal/pathology , Sarcopenia/pathology
12.
Mol Metab ; 6(11): 1429-1442, 2017 11.
Article in English | MEDLINE | ID: mdl-29107290

ABSTRACT

OBJECTIVE: Strategies improving skeletal muscle mitochondrial capacity are commonly paralleled by improvements in (metabolic) health. We and others previously identified microRNAs regulating mitochondrial oxidative capacity, but data in skeletal muscle are limited. Therefore, the present study aimed to identify novel microRNAs regulating skeletal muscle mitochondrial metabolism. METHODS AND RESULTS: We conducted an unbiased, hypothesis-free microRNA silencing screen in C2C12 myoblasts, using >700 specific microRNA inhibitors, and investigated a broad panel of mitochondrial markers. After subsequent validation in differentiated C2C12 myotubes, and exclusion of microRNAs without a human homologue or with an adverse effect on mitochondrial metabolism, 19 candidate microRNAs remained. Human clinical relevance of these microRNAs was investigated by measuring their expression in human skeletal muscle of subject groups displaying large variation in skeletal muscle mitochondrial capacity. CONCLUSION: The results show that that microRNA-320a, microRNA-196b-3p, microRNA-150-5p, and microRNA-34c-3p are tightly related to in vivo skeletal muscle mitochondrial function in humans and identify these microRNAs as targets for improving mitochondrial metabolism.


Subject(s)
MicroRNAs/genetics , Mitochondria, Muscle/metabolism , Myoblasts, Smooth Muscle/metabolism , Animals , Cell Differentiation/genetics , Cell Line , Humans , Male , Mice , MicroRNAs/metabolism , Mitochondria, Muscle/genetics , Muscle Fibers, Skeletal/cytology , Muscle Fibers, Skeletal/metabolism , Muscle, Skeletal/cytology , Muscle, Skeletal/metabolism
13.
Food Chem Toxicol ; 108(Pt A): 289-297, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28757461

ABSTRACT

Urolithins are metabolites produced in the gut following consumption of ellagitannins and ellagic acid rich foods such as pomegranates, nuts and certain berries. Urolithin A (UA) is one of the predominant isoforms of urolithins in humans and has demonstrated compelling biological activities, suggesting potential benefits of direct consumption of UA. However, an evaluation of the safety of direct administration of UA has not yet been published. The aim of this study was to investigate for the first time the genotoxicity, toxicokinetics, and repeated dose safety of orally administered synthetic UA in rats. The battery of genotoxicity assays demonstrated that UA is not genotoxic. The ADME study showed that glucuronidated and sulfonated forms of UA are the predominant metabolites following both oral and i.v. administration. The 28-day (0, 0.175, 1.75, and 5.0% UA mixed in diet) and 90-day studies (0, 1.25, 2.5, and 5.0% UA mixed in diet) showed no alterations in clinical parameters, blood chemistry, or hematology, and did not indicate any target organs, or any specific toxic mechanisms. The NOAEL was the highest dose tested, 5% UA by weight in the diet, or 3451 mg/kg bw/day in males and 3826 mg/kg bw/day in females in the 90-day oral study.


Subject(s)
Coumarins/toxicity , Ellagic Acid/pharmacology , Hydrolyzable Tannins/pharmacology , Microbiota/drug effects , Microbiota/physiology , Animals , Coumarins/chemistry , Dose-Response Relationship, Drug , Drug Administration Schedule , Female , Humans , Male , Mice , Molecular Structure , Mutagenicity Tests , Rats , Toxicokinetics
14.
Nat Med ; 22(8): 879-88, 2016 08.
Article in English | MEDLINE | ID: mdl-27400265

ABSTRACT

The biological effects of urolithins remain poorly characterized, despite wide-spread human exposure via the dietary consumption of their metabolic precursors, the ellagitannins, which are found in the pomegranate fruit, as well as in nuts and berries. We identified urolithin A (UA) as a first-in-class natural compound that induces mitophagy both in vitro and in vivo following oral consumption. In C. elegans, UA prevented the accumulation of dysfunctional mitochondria with age and extended lifespan. Likewise, UA prolonged normal activity during aging in C. elegans, including mobility and pharyngeal pumping, while maintaining mitochondrial respiratory capacity. These effects translated to rodents, where UA improved exercise capacity in two different mouse models of age-related decline of muscle function, as well as in young rats. Our findings highlight the health benefits of urolithin A and its potential application in strategies to improve mitochondrial and muscle function.


Subject(s)
Coumarins/pharmacology , Longevity/drug effects , Mitochondria/drug effects , Mitophagy/drug effects , Muscle, Skeletal/drug effects , Myoblasts/drug effects , RNA, Messenger/drug effects , Animals , Caenorhabditis elegans , DNA, Mitochondrial/drug effects , DNA, Mitochondrial/metabolism , Fertility/drug effects , Mice , Microscopy, Confocal , Mitochondria/metabolism , Muscle, Skeletal/metabolism , Myoblasts/metabolism , Oxygen Consumption , Pharynx/drug effects , RNA, Messenger/metabolism , Rats , Real-Time Polymerase Chain Reaction
15.
PLoS Genet ; 10(9): e1004673, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25255223

ABSTRACT

The BXD genetic reference population is a recombinant inbred panel descended from crosses between the C57BL/6 (B6) and DBA/2 (D2) strains of mice, which segregate for about 5 million sequence variants. Recently, some of these variants have been established with effects on general metabolic phenotypes such as glucose response and bone strength. Here we phenotype 43 BXD strains and observe they have large variation (-5-fold) in their spontaneous activity during waking hours. QTL analyses indicate that -40% of this variance is attributable to a narrow locus containing the aryl hydrocarbon receptor (Ahr), a basic helix-loop-helix transcription factor with well-established roles in development and xenobiotic metabolism. Strains with the D2 allele of Ahr have reduced gene expression compared to those with the B6 allele, and have significantly higher spontaneous activity. This effect was also observed in B6 mice with a congenic D2 Ahr interval, and in B6 mice with a humanized AHR allele which, like the D2 allele, is expressed much less and has less enzymatic activity than the B6 allele. Ahr is highly conserved in invertebrates, and strikingly inhibition of its orthologs in D. melanogaster and C. elegans (spineless and ahr-1) leads to marked increases in basal activity. In mammals, Ahr has numerous ligands, but most are either non-selective (e.g. resveratrol) or highly toxic (e.g., 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD)). Thus, we chose to examine a major environmental influence--long term feeding with high fat diet (HFD)--to see if the effects of Ahr are dependent on major metabolic differences. Interestingly, while HFD robustly halved movement across all strains, the QTL position and effects of Ahr remained unchanged, indicating that the effects are independent. The highly consistent effects of Ahr on movement indicate that changes in its constitutive activity have a role on spontaneous movement and may influence human behavior.


Subject(s)
Motor Activity/genetics , Receptors, Aryl Hydrocarbon/genetics , Amino Acid Sequence , Animals , Conserved Sequence , Evolution, Molecular , Female , Genetic Association Studies , Humans , Male , Mice , Molecular Sequence Data , Phenotype , Phylogeny , Quantitative Trait Loci , Quantitative Trait, Heritable , Sequence Alignment
16.
Sci Rep ; 4: 5285, 2014 Jun 13.
Article in English | MEDLINE | ID: mdl-24923838

ABSTRACT

Mitochondria are semi-autonomous organelles regulated by a complex network of proteins that are vital for many cellular functions. Because mitochondrial modulators can impact many aspects of cellular homeostasis, their identification and validation has proven challenging. It requires the measurement of multiple parameters in parallel to understand the exact nature of the changes induced by such compounds. We developed a platform of assays scoring for mitochondrial function in two complementary models systems, mammalian cells and C. elegans. We first optimized cell culture conditions and established the mitochondrial signature of 1,200 FDA-approved drugs in liver cells. Using cell-based and C. elegans assays, we further defined the metabolic effects of two pharmacological classes that emerged from our hit list, i.e. imidazoles and statins. We found that these two drug classes affect respiration through different and cholesterol-independent mechanisms in both models. Our screening strategy enabled us to unequivocally identify compounds that have toxic or beneficial effects on mitochondrial activity. Furthermore, the cross-species approach provided novel mechanistic insight and allowed early validation of hits that act on mitochondrial function.


Subject(s)
Caenorhabditis elegans/drug effects , Mitochondria/drug effects , Oxidative Phosphorylation/drug effects , Oxygen Consumption/drug effects , Pharmaceutical Preparations/administration & dosage , Animals , Caenorhabditis elegans/cytology , Caenorhabditis elegans/metabolism , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Line , Cell Line, Tumor , Cluster Analysis , Drug Approval , Drug Evaluation, Preclinical/methods , Fatty Acids, Monounsaturated/pharmacology , Fluvastatin , Gene Expression/drug effects , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Imidazoles/pharmacology , Indoles/pharmacology , Lovastatin/pharmacology , MCF-7 Cells , Mice , Mitochondria/metabolism , Pharmaceutical Preparations/classification , Reproducibility of Results , Simvastatin/pharmacology , United States , United States Food and Drug Administration
17.
Hum Mol Genet ; 23(18): 4995-5008, 2014 Sep 15.
Article in English | MEDLINE | ID: mdl-24833719

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is a fatal, neurodegenerative disease that causes death of motor neurons. ALS patients and mouse models of familial ALS display organismal level metabolic dysfunction, which includes increased energy expenditure despite decreased lean mass. The pathophysiological relevance of abnormal energy homeostasis to motor neuron disease remains unclear. Leptin is an adipocyte-derived hormone that regulates whole-animal energy expenditure. Here, we report that placing mutant superoxide dismutase 1 (SOD1) mice in a leptin-deficient background improves energy homeostasis and slows disease progression. Leptin-deficient mutant SOD1 mice possess increased bodyweight and fat mass, as well as decreased energy expenditure. These observations coincide with enhanced survival, improved strength and decreased motor neuron loss. These results suggest that altering whole-body energy metabolism in mutant SOD1 mice can mitigate disease progression. We propose that manipulations that increase fat mass and reduce energy expenditure will be beneficial in the setting of motor neuron disease.


Subject(s)
Amyotrophic Lateral Sclerosis/pathology , Leptin/deficiency , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism , Amyotrophic Lateral Sclerosis/genetics , Animals , Body Weight , Disease Models, Animal , Energy Metabolism , Humans , Male , Mice , Motor Neurons/metabolism , Muscle, Skeletal/metabolism , Superoxide Dismutase-1
18.
Proc Natl Acad Sci U S A ; 110(48): 19507-12, 2013 Nov 26.
Article in English | MEDLINE | ID: mdl-24218578

ABSTRACT

Copper is an essential trace element, the imbalances of which are associated with various pathological conditions, including cancer, albeit via largely undefined molecular and cellular mechanisms. Here we provide evidence that levels of bioavailable copper modulate tumor growth. Chronic exposure to elevated levels of copper in drinking water, corresponding to the maximum allowed in public water supplies, stimulated proliferation of cancer cells and de novo pancreatic tumor growth in mice. Conversely, reducing systemic copper levels with a chelating drug, clinically used to treat copper disorders, impaired both. Under such copper limitation, tumors displayed decreased activity of the copper-binding mitochondrial enzyme cytochrome c oxidase and reduced ATP levels, despite enhanced glycolysis, which was not accompanied by increased invasiveness of tumors. The antiproliferative effect of copper chelation was enhanced when combined with inhibitors of glycolysis. Interestingly, larger tumors contained less copper than smaller tumors and exhibited comparatively lower activity of cytochrome c oxidase and increased glucose uptake. These results establish copper as a tumor promoter and reveal that varying levels of copper serves to regulate oxidative phosphorylation in rapidly proliferating cancer cells inside solid tumors. Thus, activation of glycolysis in tumors may in part reflect insufficient copper bioavailability in the tumor microenvironment.


Subject(s)
Copper/toxicity , Drinking Water/analysis , Neoplasms/physiopathology , Oxidative Phosphorylation/drug effects , Tumor Microenvironment/physiology , Water Pollutants, Chemical/toxicity , Adenosine Triphosphate/metabolism , Animals , Biological Availability , Blotting, Western , DNA Primers/genetics , Energy Metabolism/drug effects , Glycolysis/physiology , Membrane Potential, Mitochondrial/drug effects , Mice , Positron-Emission Tomography , Real-Time Polymerase Chain Reaction , Respiration
19.
PLoS One ; 8(9): e74162, 2013.
Article in English | MEDLINE | ID: mdl-24066113

ABSTRACT

Glucose is an important metabolic substrate of the retina and diabetic patients have to maintain a strict normoglycemia to avoid diabetes secondary effects, including cardiovascular disease, nephropathy, neuropathy and retinopathy. Others and we recently demonstrated the potential role of hypoglycemia in diabetic retinopathy. We showed acute hypoglycemia to induce retinal cell death both in vivo during an hyperinsulinemic/hypoglycemic clamp and in vitro in 661W photoreceptor cells cultured at low glucose concentration. In the present study, we showed low glucose to induce a decrease of BCL2 and BCL-XL anti-apoptotic proteins expression, leading to an increase of free pro-apoptotic BAX. In parallel, we showed that, in retinal cells, low glucose-induced apoptosis is involved in the process of autophagosomes formation through the AMPK/RAPTOR/mTOR pathway. Moreover, the decrease of LAMP2a expression led to a defect in the autophagosome/lysosome fusion process. Specific inhibition of autophagy, either by 3-methyladenine or by down-regulation of ATG5 or ATG7 proteins expression, increased caspase 3 activation and 661W cell death. We show that low glucose modifies the delicate equilibrium between apoptosis and autophagy. Cells struggled against low nutrient condition-induced apoptosis by starting an autophagic process, which led to cell death when inhibited. We conclude that autophagy defect is associated with low glucose-induced 661W cells death that could play a role in diabetic retinopathy. These results could modify the way of addressing negative effects of hypoglycemia. Short-term modulation of autophagy could be envisioned to treat diabetic patients in order to avoid secondary complications of the disease.


Subject(s)
Apoptosis/drug effects , Autophagy/drug effects , Glucose/pharmacology , Photoreceptor Cells/cytology , Photoreceptor Cells/drug effects , Animals , Blotting, Western , Cells, Cultured , Immunohistochemistry , Immunoprecipitation , In Situ Nick-End Labeling , Mice , Photoreceptor Cells/metabolism , Reverse Transcriptase Polymerase Chain Reaction
20.
Nat Rev Drug Discov ; 12(6): 465-83, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23666487

ABSTRACT

Mitochondrial dysfunction is not only a hallmark of rare inherited mitochondrial disorders but also implicated in age-related diseases, including those that affect the metabolic and nervous system, such as type 2 diabetes and Parkinson's disease. Numerous pathways maintain and/or restore proper mitochondrial function, including mitochondrial biogenesis, mitochondrial dynamics, mitophagy and the mitochondrial unfolded protein response. New and powerful phenotypic assays in cell-based models as well as multicellular organisms have been developed to explore these different aspects of mitochondrial function. Modulating mitochondrial function has therefore emerged as an attractive therapeutic strategy for several diseases, which has spurred active drug discovery efforts in this area.


Subject(s)
Mitochondria/drug effects , Mitochondria/physiology , Mitochondrial Diseases/drug therapy , Pharmaceutical Preparations/administration & dosage , Animals , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/metabolism , Drug Delivery Systems/trends , Drug Discovery/trends , Humans , Mitochondrial Diseases/metabolism , Parkinson Disease/drug therapy , Parkinson Disease/metabolism , Pharmaceutical Preparations/chemistry , Pharmaceutical Preparations/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL