72 results on '"Feige, JN"'
Search Results
2. A robust neuromuscular system protects rat and human skeletal muscle from sarcopenia.
- Author
-
Pannérec, A, Springer, M, Migliavacca, E, Ireland, A, Piasecki, M, Karaz, S, Jacot, G, Métairon, S, Danenberg, E, Raymond, F, Descombes, P, McPhee, JS, Feige, JN, Pannérec, A, Springer, M, Migliavacca, E, Ireland, A, Piasecki, M, Karaz, S, Jacot, G, Métairon, S, Danenberg, E, Raymond, F, Descombes, P, McPhee, JS, and Feige, JN
- Abstract
Declining muscle mass and function is one of the main drivers of loss of independence in the elderly. Sarcopenia is associated with numerous cellular and endocrine perturbations, and it remains challenging to identify those changes that play a causal role and could serve as targets for therapeutic intervention. In this study, we uncovered a remarkable differential susceptibility of certain muscles to age-related decline. Aging rats specifically lose muscle mass and function in the hindlimbs, but not in the forelimbs. By performing a comprehensive comparative analysis of these muscles, we demonstrate that regional susceptibility to sarcopenia is dependent on neuromuscular junction fragmentation, loss of motoneuron innervation, and reduced excitability. Remarkably, muscle loss in elderly humans also differs in vastus lateralis and tibialis anterior muscles in direct relation to neuromuscular dysfunction. By comparing gene expression in susceptible and non-susceptible muscles, we identified a specific transcriptomic signature of neuromuscular impairment. Importantly, differential molecular profiling of the associated peripheral nerves revealed fundamental changes in cholesterol biosynthetic pathways. Altogether our results provide compelling evidence that susceptibility to sarcopenia is tightly linked to neuromuscular decline in rats and humans, and identify dysregulation of sterol metabolism in the peripheral nervous system as an early event in this process.
- Published
- 2016
3. The pollutant diethylhexyl phthalate regulates hepatic energy metabolism via species-specific PPARalpha-dependent mechanisms.
- Author
-
Feige JN, Gerber A, Casals-Casas C, Winkler C, Bedu E, Bueno M, Gelman L, Auwerx J, Gonzalez FJ, and Desvergne B
- Abstract
Background: The modulation of energetic homeostasis by pollutants has recently emerged as a potential contributor to the onset of metabolic disorders. Diethylhexyl phthalate (DEHP) is a widely used industrial plasticizer to which humans are widely exposed. Phthalates can activate the three peroxisome proliferator-activated receptor (PPAR) isotypes on cellular models and induce peroxisome proliferation in rodents. Objectives: In this study, we aimed to evaluate the systemic and metabolic consequences of DEHP exposure that have remained so far unexplored and to characterize the underlying molecular mechanisms of action. Methods: As a proof of concept and mechanism, genetically engineered mouse models of PPARs were exposed to high doses of DEHP, followed by metabolic and molecular analyses. Results: DEHP-treated mice were protected from diet-induced obesity via PPAR[alpha]-dependent activation of hepatic fatty acid catabolism, whereas the activity of neither PPAR[beta] nor PPAR[gamma] was affected. However, the lean phenotype observed in response to DEHP in wild-type mice was surprisingly abolished in PPAR[alpha]-humanized mice. These species differences are associated with a different pattern of coregulator recruitment. Conclusion: These results demonstrate that DEHP exerts species-specific metabolic actions that rely to a large extent on PPARa signaling and highlight the metabolic importance of the species-specific activation of PPARa by xenobiotic compounds. [ABSTRACT FROM AUTHOR]
- Published
- 2010
- Full Text
- View/download PDF
4. PPAR gamma Controls Ectopic Adipogenesis and Cross-Talks with Myogenesis During Skeletal Muscle Regeneration
- Author
-
Dammone, G, Karaz, S, Lukjanenko, L, Winkler, C, Sizzano, F, Jacot, G, Migliavacca, E, Palini, A, Desvergne, B, Gilardi, F, and Feige, JN
5. Mitochondrial calcium uptake declines during aging and is directly activated by oleuropein to boost energy metabolism and skeletal muscle performance.
- Author
-
Gherardi G, Weiser A, Bermont F, Migliavacca E, Brinon B, Jacot GE, Hermant A, Sturlese M, Nogara L, Vascon F, De Mario A, Mattarei A, Garratt E, Burton M, Lillycrop K, Godfrey KM, Cendron L, Barron D, Moro S, Blaauw B, Rizzuto R, Feige JN, Mammucari C, and De Marchi U
- Abstract
Mitochondrial calcium (mtCa
2+ ) uptake via the mitochondrial calcium uniporter (MCU) couples calcium homeostasis and energy metabolism. mtCa2+ uptake via MCU is rate-limiting for mitochondrial activation during muscle contraction, but its pathophysiological role and therapeutic application remain largely uncharacterized. By profiling human muscle biopsies, patient-derived myotubes, and preclinical models, we discovered a conserved downregulation of mitochondrial calcium uniporter regulator 1 (MCUR1) during skeletal muscle aging that associates with human sarcopenia and impairs mtCa2+ uptake and mitochondrial respiration. Through a screen of 5,000 bioactive molecules, we identify the natural polyphenol oleuropein as a specific MCU activator that stimulates mitochondrial respiration via mitochondrial calcium uptake 1 (MICU1) binding. Oleuropein activates mtCa2+ uptake and energy metabolism to enhance endurance and reduce fatigue in young and aged mice but not in muscle-specific MCU knockout (KO) mice. Our work demonstrates that impaired mtCa2+ uptake contributes to mitochondrial dysfunction during aging and establishes oleuropein as a novel food-derived molecule that specifically targets MCU to stimulate mitochondrial bioenergetics and muscle performance., Competing Interests: Declaration of interests The authors A.W., F.B., B. Brinon, G.E.J., A.H., E.M., D.B., J.N.F., and U.D.M. are employees of Nestlé Research, which is part of the Société des Produits Nestlé SA (SPN). SPN has filed patents on the use of oleuropein for muscle and mitochondrial health. K.M.G. has received reimbursement for speaking at conferences sponsored by companies selling nutritional products and is part of an academic consortium that has received research funding from Bayer, Société des Produits Nestlé SA, BenevolentAI Bio Ltd., and Danone., (Copyright © 2024 The Authors. Published by Elsevier Inc. All rights reserved.)- Published
- 2024
- Full Text
- View/download PDF
6. Nicotinamide and pyridoxine stimulate muscle stem cell expansion and enhance regenerative capacity during aging.
- Author
-
Ancel S, Michaud J, Migliavacca E, Jomard C, Fessard A, Garcia P, Karaz S, Raja S, Jacot GE, Desgeorges T, Sánchez-García JL, Tauzin L, Ratinaud Y, Brinon B, Métairon S, Pinero L, Barron D, Blum S, Karagounis LG, Heshmat R, Ostovar A, Farzadfar F, Scionti I, Mounier R, Gondin J, Stuelsatz P, and Feige JN
- Subjects
- Humans, Animals, Mice, Male, Female, Aged, Middle Aged, Adult, Cell Differentiation drug effects, Niacinamide pharmacology, Pyridoxine pharmacology, Aging drug effects, Regeneration drug effects, Muscle, Skeletal drug effects, Muscle, Skeletal metabolism, Muscle, Skeletal physiology
- Abstract
Skeletal muscle relies on resident muscle stem cells (MuSCs) for growth and repair. Aging and muscle diseases impair MuSC function, leading to stem cell exhaustion and regenerative decline that contribute to the progressive loss of skeletal muscle mass and strength. In the absence of clinically available nutritional solutions specifically targeting MuSCs, we used a human myogenic progenitor high-content imaging screen of natural molecules from food to identify nicotinamide (NAM) and pyridoxine (PN) as bioactive nutrients that stimulate MuSCs and have a history of safe human use. NAM and PN synergize via CK1-mediated cytoplasmic β-catenin activation and AKT signaling to promote amplification and differentiation of MuSCs. Oral treatment with a combination of NAM and PN accelerated muscle regeneration in vivo by stimulating MuSCs, increased muscle strength during recovery, and overcame MuSC dysfunction and regenerative failure during aging. Levels of NAM and bioactive PN spontaneously declined during aging in model organisms and interindependently associated with muscle mass and walking speed in a cohort of 186 aged people. Collectively, our results establish the NAM/PN combination as a nutritional intervention that stimulates MuSCs, enhances muscle regeneration, and alleviates age-related muscle decline with a direct opportunity for clinical translation.
- Published
- 2024
- Full Text
- View/download PDF
7. A dual-color PAX7 and MYF5 in vivo reporter to investigate muscle stem cell heterogeneity in regeneration and aging.
- Author
-
Ancel S, Michaud J, Sizzano F, Tauzin L, Oliveira M, Migliavacca E, Schüler SC, Raja S, Dammone G, Karaz S, Sánchez-García JL, Metairon S, Jacot G, Bentzinger CF, Feige JN, and Stuelsatz P
- Subjects
- Animals, Mice, Stem Cells metabolism, Stem Cells cytology, Cell Proliferation, Muscle, Skeletal metabolism, Muscle, Skeletal cytology, Cell Differentiation, Mice, Transgenic, Cell Self Renewal, PAX7 Transcription Factor metabolism, PAX7 Transcription Factor genetics, Myogenic Regulatory Factor 5 metabolism, Myogenic Regulatory Factor 5 genetics, Regeneration, Aging metabolism, Genes, Reporter
- Abstract
Increasing evidence suggests that the muscle stem cell (MuSC) pool is heterogeneous. In particular, a rare subset of PAX7-positive MuSCs that has never expressed the myogenic regulatory factor MYF5 displays unique self-renewal and engraftment characteristics. However, the scarcity and limited availability of protein markers make the characterization of these cells challenging. Here, we describe the generation of StemRep reporter mice enabling the monitoring of PAX7 and MYF5 proteins based on equimolar levels of dual nuclear fluorescence. High levels of PAX7 protein and low levels of MYF5 delineate a deeply quiescent MuSC subpopulation with an increased capacity for asymmetric division and distinct dynamics of activation, proliferation, and commitment. Aging primarily reduces the MYF5
Low MuSCs and skews the stem cell pool toward MYF5High cells with lower quiescence and self-renewal potential. Altogether, we establish the StemRep model as a versatile tool to study MuSC heterogeneity and broaden our understanding of mechanisms regulating MuSC quiescence and self-renewal in homeostatic, regenerating, and aged muscles., Competing Interests: Declaration of interests Except S.C.S., all authors are or were employees of Société des Produits Nestlé SA., (Copyright © 2024 The Authors. Published by Elsevier Inc. All rights reserved.)- Published
- 2024
- Full Text
- View/download PDF
8. The Mediating Role of Kynurenine Pathway Metabolites on the Relationship Between Inflammation and Muscle Mass in Oldest-Old Men.
- Author
-
Hetherington-Rauth M, Johnson E, Migliavacca E, Langsetmo L, Hepple RT, Ryan TE, Ferrucci L, Breuillé D, Corthesy J, Lane NE, Feige JN, Napoli N, Tramontana F, Orwoll ES, and Cawthon PM
- Subjects
- Humans, Male, Aged, 80 and over, C-Reactive Protein metabolism, Tumor Necrosis Factor-alpha metabolism, Sarcopenia metabolism, 3-Hydroxyanthranilic Acid metabolism, Cytokines metabolism, Xanthurenates metabolism, Kynurenine metabolism, Kynurenine analogs & derivatives, Inflammation metabolism, Biomarkers metabolism, Tryptophan metabolism, Muscle, Skeletal metabolism
- Abstract
Tryptophan (TRP) metabolites along the kynurenine (KYN) pathway (KP) have been found to influence muscle. Proinflammatory cytokines are known to stimulate the degradation of TRP down the KP. Given that both inflammation and KP metabolites have been connected with loss of muscle, we assessed the potential mediating role of KP metabolites on inflammation and muscle mass in older men. Five hundred and five men (85.0 ± 4.2 years) from the Osteoporotic Fractures in Men cohort study with measured D3-creatine dilution (D3Cr) muscle mass, KP metabolites, and inflammation markers (C-reactive protein [CRP], alpha-1-acid glycoprotein [AGP] and a subsample [n = 305] with interleukin [IL-6, IL-1β, IL-17A] and tumor necrosis factor-α [TNF-α]) were included in the analysis. KP metabolites and inflammatory markers were measured using liquid chromatography-tandem mass spectrometry and immunoassays, respectively. 23%-92% of the inverse relationship between inflammatory markers and D3Cr muscle mass was mediated by KP metabolites (indirect effect p < .05). 3-hydroxyanthranilic acid (3-HAA), quinolinic acid (QA), TRP, xanthurenic acid (XA), KYN/TRP, 3-hydroxykynurenine (3-HK)/3-HAA, QA/3-HAA, and nicotinamide (NAM)/QA mediated the AGP relationship. 3-HAA, QA, KYN/TRP, 3-HK/XA, HKr ratio, 3-HK/3-HAA, QA/3-HAA, and NAM/QA mediated the CRP. KYN/TRP, 3-HK/XA, and NAM/QA explained the relationship for IL-6 and 3-HK/XA and QA/3-HAA for TNF-α. No mediation effect was observed for the other cytokines (indirect effect p > .05). KP metabolites, particularly higher ratios of KYN/TRP, 3-HK/XA, 3-HK/3-HAA, QA/3-HAA, and a lower ratio of NAM/QA, mediated the relationship between inflammation and low muscle mass. Our preliminary cross-sectional data suggest that interventions to alter D3Cr muscle mass may focus on KP metabolites rather than inflammation per se., (© The Author(s) 2024. Published by Oxford University Press on behalf of The Gerontological Society of America. All rights reserved. For commercial re-use, please contact reprints@oup.com for reprints and translation rights for reprints. All other permissions can be obtained through our RightsLink service via the Permissions link on the article page on our site—for further information please contact journals.permissions@oup.com.)
- Published
- 2024
- Full Text
- View/download PDF
9. Apelin stimulation of the vascular skeletal muscle stem cell niche enhances endogenous repair in dystrophic mice.
- Author
-
Le Moal E, Liu Y, Collerette-Tremblay J, Dumontier S, Fabre P, Molina T, Dort J, Orfi Z, Denault N, Boutin J, Michaud J, Giguère H, Desroches A, Trân K, Ellezam B, Vézina F, Bedard S, Raynaud C, Balg F, Sarret P, Boudreault PL, Scott MS, Denault JB, Marsault E, Feige JN, Auger-Messier M, Dumont NA, and Bentzinger CF
- Subjects
- Mice, Animals, Apelin metabolism, Muscle, Skeletal metabolism, Signal Transduction, Stem Cell Niche, Muscular Dystrophy, Duchenne metabolism
- Abstract
Impaired skeletal muscle stem cell (MuSC) function has long been suspected to contribute to the pathogenesis of muscular dystrophy (MD). Here, we showed that defects in the endothelial cell (EC) compartment of the vascular stem cell niche in mouse models of Duchenne MD, laminin α2-related MD, and collagen VI-related myopathy were associated with inefficient mobilization of MuSCs after tissue damage. Using chemoinformatic analysis, we identified the 13-amino acid form of the peptide hormone apelin (AP-13) as a candidate for systemic stimulation of skeletal muscle ECs. Systemic administration of AP-13 using osmotic pumps generated a pro-proliferative EC-rich niche that supported MuSC function through angiocrine factors and markedly improved tissue regeneration and muscle strength in all three dystrophic mouse models. Moreover, EC-specific knockout of the apelin receptor led to regenerative defects that phenocopied key pathological features of MD, including vascular defects, fibrosis, muscle fiber necrosis, impaired MuSC function, and reduced force generation. Together, these studies provide in vivo proof of concept that enhancing endogenous skeletal muscle repair by targeting the vascular niche is a viable therapeutic avenue for MD and characterized AP-13 as a candidate for further study for the systemic treatment of MuSC dysfunction.
- Published
- 2024
- Full Text
- View/download PDF
10. Trigonelline is an NAD + precursor that improves muscle function during ageing and is reduced in human sarcopenia.
- Author
-
Membrez M, Migliavacca E, Christen S, Yaku K, Trieu J, Lee AK, Morandini F, Giner MP, Stiner J, Makarov MV, Garratt ES, Vasiloglou MF, Chanvillard L, Dalbram E, Ehrlich AM, Sanchez-Garcia JL, Canto C, Karagounis LG, Treebak JT, Migaud ME, Heshmat R, Razi F, Karnani N, Ostovar A, Farzadfar F, Tay SKH, Sanders MJ, Lillycrop KA, Godfrey KM, Nakagawa T, Moco S, Koopman R, Lynch GS, Sorrentino V, and Feige JN
- Subjects
- Humans, Male, Mice, Animals, NAD metabolism, Caenorhabditis elegans, Aging, Muscle, Skeletal metabolism, Sarcopenia drug therapy, Sarcopenia prevention & control, Sarcopenia metabolism, Alkaloids pharmacology, Alkaloids therapeutic use, Alkaloids metabolism
- Abstract
Mitochondrial dysfunction and low nicotinamide adenine dinucleotide (NAD
+ ) levels are hallmarks of skeletal muscle ageing and sarcopenia1-3 , but it is unclear whether these defects result from local changes or can be mediated by systemic or dietary cues. Here we report a functional link between circulating levels of the natural alkaloid trigonelline, which is structurally related to nicotinic acid4 , NAD+ levels and muscle health in multiple species. In humans, serum trigonelline levels are reduced with sarcopenia and correlate positively with muscle strength and mitochondrial oxidative phosphorylation in skeletal muscle. Using naturally occurring and isotopically labelled trigonelline, we demonstrate that trigonelline incorporates into the NAD+ pool and increases NAD+ levels in Caenorhabditis elegans, mice and primary myotubes from healthy individuals and individuals with sarcopenia. Mechanistically, trigonelline does not activate GPR109A but is metabolized via the nicotinate phosphoribosyltransferase/Preiss-Handler pathway5,6 across models. In C. elegans, trigonelline improves mitochondrial respiration and biogenesis, reduces age-related muscle wasting and increases lifespan and mobility through an NAD+ -dependent mechanism requiring sirtuin. Dietary trigonelline supplementation in male mice enhances muscle strength and prevents fatigue during ageing. Collectively, we identify nutritional supplementation of trigonelline as an NAD+ -boosting strategy with therapeutic potential for age-associated muscle decline., (© 2024. The Author(s).)- Published
- 2024
- Full Text
- View/download PDF
11. Nutrient Metabolites Associated With Low D3Cr Muscle Mass, Strength, and Physical Performance in Older Men.
- Author
-
Hetherington-Rauth M, Johnson E, Migliavacca E, Parimi N, Langsetmo L, Hepple RT, Grzywinski Y, Corthesy J, Ryan TE, Ferrucci L, Feige JN, Orwoll ES, and Cawthon PM
- Subjects
- Male, Humans, Aged, Cohort Studies, Hand Strength physiology, Physical Functional Performance, Muscles, Nutrients, Muscle, Skeletal, Creatine, Muscle Strength physiology
- Abstract
Background: The relationship between amino acids, B vitamins, and their metabolites with D3-creatine (D3Cr) dilution muscle mass, a more direct measure of skeletal muscle mass, has not been investigated. We aimed to assess associations of plasma metabolites with D3Cr muscle mass, as well as muscle strength and physical performance in older men from the Osteoporotic Fractures in Men cohort study., Methods: Out of 1 425 men (84.2 ± 4.1 years), men with the lowest D3Cr muscle mass (n = 100), slowest walking speed (n = 100), lowest grip strength (n = 100), and a random sample (n = 200) serving as a comparison group to the low groups were included. Metabolites were analyzed using liquid chromatography-tandem mass spectrometry. Metabolite differences between the low groups and random sample and their relationships with the muscle outcomes adjusted for confounders and multiple comparisons were assessed using t-test/Mann-Whitney-Wilcoxon and partial correlations, respectively., Results: For D3Cr muscle mass, significant biomarkers (p < .001) with ≥10% fold difference and largest partial correlations were tryptophan (Trp; r = 0.31), kynurenine (Kyn)/Trp; r = -0.27), nicotinamide (Nam)/quinolinic acid (Quin; r = 0.21), and alpha-hydroxy-5-methyl-tetrahydrofolate (hm-THF; r = -0.25). For walking speed, hm-THF, Nam/Quin, and Quin had the largest significance and fold difference, whereas valine (r = 0.17), Trp (r = 0.17), HKyn/Xant (r = -0.20), neopterin (r = -0.17), 5-methyl-THF (r = -0.20), methylated folate (r = -0.21), and thiamine (r = -0.18) had the strongest correlations. Only hm-THF was correlated with grip strength (r = -0.21) and differed between the low group and the random sample., Conclusions: Future interventions focusing on how the Trp metabolic pathway or hm-THF influences D3Cr muscle mass and physical performance declines in older adults are warranted., (© The Author(s) 2023. Published by Oxford University Press on behalf of The Gerontological Society of America. All rights reserved. For permissions, please e-mail: journals.permissions@oup.com.)
- Published
- 2024
- Full Text
- View/download PDF
12. The mitochondrial calcium uniporter (MCU) activates mitochondrial respiration and enhances mobility by regulating mitochondrial redox state.
- Author
-
Weiser A, Hermant A, Bermont F, Sizzano F, Karaz S, Alvarez-Illera P, Santo-Domingo J, Sorrentino V, Feige JN, and De Marchi U
- Subjects
- Animals, Humans, Calcium metabolism, Oxidation-Reduction, Respiration, Caenorhabditis elegans metabolism, Mitochondria metabolism
- Abstract
Regulation of mitochondrial redox balance is emerging as a key event for cell signaling in both physiological and pathological conditions. However, the link between the mitochondrial redox state and the modulation of these conditions remains poorly defined. Here, we discovered that activation of the evolutionary conserved mitochondrial calcium uniporter (MCU) modulates mitochondrial redox state. By using mitochondria-targeted redox and calcium sensors and genetic MCU-ablated models, we provide evidence of the causality between MCU activation and net reduction of mitochondrial (but not cytosolic) redox state. Redox modulation of redox-sensitive groups via MCU stimulation is required for maintaining respiratory capacity in primary human myotubes and C. elegans, and boosts mobility in worms. The same benefits are obtained bypassing MCU via direct pharmacological reduction of mitochondrial proteins. Collectively, our results demonstrate that MCU regulates mitochondria redox balance and that this process is required to promote the MCU-dependent effects on mitochondrial respiration and mobility., Competing Interests: Declaration of competing interest The authors declare the following financial interests/personal relationships which may be considered as potential competing interests: A.W., A.H., F.B. , F.S., S.K., V.S., J.N.F and J. U.D.M. are or were employees of Nestlé Research, which is part of the Société des Produits Nestlé SA., (Copyright © 2023 The Authors. Published by Elsevier B.V. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
13. Evidence for inefficient contraction and abnormal mitochondrial activity in sarcopenia using magnetic resonance spectroscopy.
- Author
-
Stephenson MC, Ho JXM, Migliavacca E, Kalimeri M, Karnani N, Banerji S, Totman JJ, Feige JN, Merchant RA, and Tay SKH
- Subjects
- Male, Humans, Aged, Energy Metabolism physiology, Adenosine Triphosphate metabolism, Magnetic Resonance Spectroscopy methods, Mitochondria metabolism, Adenosine Diphosphate metabolism, Muscle, Skeletal metabolism, Sarcopenia metabolism
- Abstract
Background: Mitochondrial dysfunction has been implicated in sarcopenia.
31 P magnetic resonance spectroscopy (MRS) enables non-invasive measurement of adenosine triphosphate (ATP) synthesis rates to probe mitochondrial function. Here, we assessed muscle energetics in older sarcopenic and non-sarcopenic men and compared with muscle biopsy-derived markers of mitochondrial function., Methods: Twenty Chinese men with sarcopenia (SARC, age = 73.1 ± 4.1 years) and 19 healthy aged and sex-matched controls (CON, age = 70.3 ± 4.2 years) underwent assessment of strength, physical performance, and magnetic resonance imaging. Concentrations of phosphocreatine (PCr), ATP and inorganic phosphate (Pi) as well as muscle pH were measured at rest and during an interleaved rest-exercise protocol to probe muscle mitochondrial function. Results were compared to biopsy-derived mitochondrial complex activity and expression to understand underlying metabolic perturbations., Results: Despite matched muscle contractile power (strength/cross-sectional area), the ATP contractile cost was higher in SARC compared with CON (low-intensity exercise: 1.06 ± 0.59 vs. 0.57 ± 0.22, moderate: 0.93 ± 0.43 vs. 0.58 ± 0.68, high: 0.70 ± 0.57 vs. 0.43 ± 0.51 mmol L-1 min-1 bar-1 cm-2 , P = 0.003, <0.0001 and <0.0001, respectively). Post-exercise mitochondrial oxidative synthesis rates (a marker of mitochondrial function) tended to be longer in SARC but did not reach significance (17.3 ± 6.4 vs. 14.6 ± 6.5 mmol L-1 min-1 , P = 0.2). However, relative increases in end-exercise ADP in SARC (31.8 ± 9.9 vs. 24.0 ± 7.3 mmol L-1 , P = 0.008) may have been a compensatory mechanism. Mitochondrial complex activity was found to be associated with exercise-induced drops in PCr [citrate synthetase activity (CS), Spearman correlation rho = -0.42, P = 0.03] and end-exercise ADP (complex III, rho = -0.52, P = 0.01; CS rho = -0.45, P = 0.02; SDH rho = -0.45, P = 0.03), with CS also being strongly associated with the PCr recovery rate following low intensity exercise (rho = -0.47, P = 0.02), and the cost of contraction at high intensity (rho = -0.54, P = 0.02). Interestingly, at high intensity, the fractional contribution of oxidative phosphorylation to exercise was correlated with activity in complex II (rho = 0.5, P = 0.03), CS (rho = 0.47, P = 0.02) and SDH (rho = 0.46, P = 0.03), linking increased mitochondrial complex activity with increased ability to generate energy through oxidative pathways., Conclusions: This study used31 P MRS to assess ATP utilization and resynthesis in sarcopenic muscle and demonstrated abnormal increases in the energy cost during exercise and perturbed mitochondrial energetics in recovery. Associations between mitochondrial complex activity and the fractional contribution to energy requirement during exercise indicate increased ability to generate energy oxidatively in those with better mitochondrial complex activity., (© 2023 The Authors. Journal of Cachexia, Sarcopenia and Muscle published by John Wiley & Sons Ltd on behalf of Society on Sarcopenia, Cachexia and Wasting Disorders.)- Published
- 2023
- Full Text
- View/download PDF
14. Nicotinamide riboside kinases regulate skeletal muscle fiber-type specification and are rate-limiting for metabolic adaptations during regeneration.
- Author
-
Sonntag T, Ancel S, Karaz S, Cichosz P, Jacot G, Giner MP, Sanchez-Garcia JL, Pannérec A, Moco S, Sorrentino V, Cantó C, and Feige JN
- Abstract
Nicotinamide riboside kinases (NRKs) control the conversion of dietary Nicotinamide Riboside (NR) to NAD
+ , but little is known about their contribution to endogenous NAD+ turnover and muscle plasticity during skeletal muscle growth and remodeling. Using NRK1/2 double KO (NRKdKO) mice, we investigated the influence of NRKs on NAD+ metabolism and muscle homeostasis, and on the response to neurogenic muscle atrophy and regeneration following muscle injury. Muscles from NRKdKO animals have altered nicotinamide (NAM) salvage and a decrease in mitochondrial content. In single myonuclei RNAseq of skeletal muscle, NRK2 mRNA expression is restricted to type IIx muscle fibers, and perturbed NAD+ turnover and mitochondrial metabolism shifts the fiber type composition of NRKdKO muscle to fast glycolytic IIB fibers. NRKdKO does not influence muscle atrophy during denervation but alters muscle repair after myofiber injury. During regeneration, muscle stem cells (MuSCs) from NRKdKO animals hyper-proliferate but fail to differentiate. NRKdKO also alters the recovery of NAD+ during muscle regeneration as well as mitochondrial adaptations and extracellular matrix remodeling required for tissue repair. These metabolic perturbations result in a transient delay of muscle regeneration which normalizes during myofiber maturation at late stages of regeneration via over-compensation of anabolic IGF1-Akt signaling. Altogether, we demonstrate that NAD+ synthesis controls mitochondrial metabolism and fiber type composition via NRK1/2 and is rate-limiting for myogenic commitment and mitochondrial maturation during skeletal muscle repair., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2022 Sonntag, Ancel, Karaz, Cichosz, Jacot, Giner, Sanchez-Garcia, Pannérec, Moco, Sorrentino, Cantó and Feige.)- Published
- 2022
- Full Text
- View/download PDF
15. Association of amino acid metabolites with osteoporosis, a metabolomic approach: Bushehr elderly health program.
- Author
-
Panahi N, Fahimfar N, Roshani S, Arjmand B, Gharibzadeh S, Shafiee G, Migliavacca E, Breuille D, Feige JN, Grzywinski Y, Corthesy J, Razi F, Heshmat R, Nabipour I, Farzadfar F, Soltani A, Larijani B, and Ostovar A
- Subjects
- Aged, Amino Acids, Chromatography, Liquid, Female, Humans, Male, Metabolomics, Prospective Studies, Tandem Mass Spectrometry, Bone Density, Osteoporosis diagnosis, Osteoporosis epidemiology
- Abstract
Introduction and Objectives: Amino acids are the most frequently reported metabolites associated with low bone mineral density (BMD) in metabolomics studies. We aimed to evaluate the association between amino acid metabolic profile and bone indices in the elderly population., Methods: 400 individuals were randomly selected from 2384 elderly men and women over 60 years participating in the second stage of the Bushehr elderly health (BEH) program, a population-based prospective cohort study that is being conducted in Bushehr, a southern province of Iran. Frozen plasma samples were used to measure 29 amino acid and derivatives metabolites using the UPLC-MS/MS-based targeted metabolomics platform. We conducted Elastic net regression analysis to detect the metabolites associated with BMD of different sites and lumbar spine trabecular bone score, and also to examine the ability of the measured metabolites to differentiate osteoporosis., Results: We adjusted the analysis for possible confounders (age, BMI, diabetes, smoking, physical activity, vitamin D level, and sex). Valine, leucine, isoleucine, and alanine in women and tryptophan in men were the most important amino acids inversely associated with osteoporosis (OR range from 0.77 to 0.89). Sarcosine, followed by tyrosine, asparagine, alpha aminobutyric acid, and ADMA in women and glutamine in men and when both women and men were considered together were the most discriminating amino acids detected in individuals with osteoporosis (OR range from 1.15 to 1.31)., Conclusion: We found several amino acid metabolites associated with possible bone status in elderly individuals. Further studies are required to evaluate the utility of these metabolites as clinical biomarkers for osteoporosis prediction and their effect on bone health as dietary supplements., (© 2022. The Author(s), under exclusive licence to Springer Science+Business Media, LLC, part of Springer Nature.)
- Published
- 2022
- Full Text
- View/download PDF
16. An engineered multicellular stem cell niche for the 3D derivation of human myogenic progenitors from iPSCs.
- Author
-
Mashinchian O, De Franceschi F, Nassiri S, Michaud J, Migliavacca E, Aouad P, Metairon S, Pruvost S, Karaz S, Fabre P, Molina T, Stuelsatz P, Hegde N, Le Moal E, Dammone G, Dumont NA, Lutolf MP, Feige JN, and Bentzinger CF
- Subjects
- Animals, Cell Differentiation, Endothelial Cells, Humans, Mice, Muscle Development, Phosphatidylinositol 3-Kinases metabolism, Stem Cell Niche, Induced Pluripotent Stem Cells metabolism
- Abstract
Fate decisions in the embryo are controlled by a plethora of microenvironmental interactions in a three-dimensional niche. To investigate whether aspects of this microenvironmental complexity can be engineered to direct myogenic human-induced pluripotent stem cell (hiPSC) differentiation, we here screened murine cell types present in the developmental or adult stem cell niche in heterotypic suspension embryoids. We identified embryonic endothelial cells and fibroblasts as highly permissive for myogenic specification of hiPSCs. After two weeks of sequential Wnt and FGF pathway induction, these three-component embryoids are enriched in Pax7-positive embryonic-like myogenic progenitors that can be isolated by flow cytometry. Myogenic differentiation of hiPSCs in heterotypic embryoids relies on a specialized structural microenvironment and depends on MAPK, PI3K/AKT, and Notch signaling. After transplantation in a mouse model of Duchenne muscular dystrophy, embryonic-like myogenic progenitors repopulate the stem cell niche, reactivate after repeated injury, and, compared to adult human myoblasts, display enhanced fusion and lead to increased muscle function. Altogether, we provide a two-week protocol for efficient and scalable suspension-based 3D derivation of Pax7-positive myogenic progenitors from hiPSCs., (© 2022 The Authors.)
- Published
- 2022
- Full Text
- View/download PDF
17. A Tead1-Apelin axis directs paracrine communication from myogenic to endothelial cells in skeletal muscle.
- Author
-
Lee U, Stuelsatz P, Karaz S, McKellar DW, Russeil J, Deak M, De Vlaminck I, Lepper C, Deplancke B, Cosgrove BD, and Feige JN
- Abstract
Apelin (Apln) is a myokine that regulates skeletal muscle plasticity and metabolism and declines during aging. Through a yeast one-hybrid transcription factor binding screen, we identified the TEA domain transcription factor 1 (Tead1) as a novel regulator of the Apln promoter. Single-cell analysis of regenerating muscle revealed that the apelin receptor ( Aplnr ) is enriched in endothelial cells, whereas Tead1 is enriched in myogenic cells. Knock-down of Tead1 stimulates Apln secretion from muscle cells in vitro and myofiber-specific overexpression of Tead1 suppresses Apln secretion in vivo . Apln secretion via Tead1 knock-down in muscle cells stimulates endothelial cell expansion via endothelial Aplnr. In vivo , Apln peptide supplementation enhances endothelial cell expansion while Tead1 muscle overexpression delays endothelial remodeling following muscle injury. Our work describes a novel paracrine crosstalk in which Apln secretion is controlled by Tead1 in myogenic cells and influences endothelial remodeling during muscle repair., Competing Interests: U.L., P.S., S.K., M.D. and J.N.F. are or were employees of the Société des Produits Nestlé S.A., (© 2022 The Authors.)
- Published
- 2022
- Full Text
- View/download PDF
18. In vivo transcriptomic profiling using cell encapsulation identifies effector pathways of systemic aging.
- Author
-
Mashinchian O, Hong X, Michaud J, Migliavacca E, Lefebvre G, Boss C, De Franceschi F, Le Moal E, Collerette-Tremblay J, Isern J, Metairon S, Raymond F, Descombes P, Bouche N, Muñoz-Cánoves P, Feige JN, and Bentzinger CF
- Subjects
- Aging, Animals, Cell Differentiation, Cell Encapsulation, Mice, Muscle, Skeletal metabolism, Transcriptome, Satellite Cells, Skeletal Muscle, Stem Cells metabolism
- Abstract
Sustained exposure to a young systemic environment rejuvenates aged organisms and promotes cellular function. However, due to the intrinsic complexity of tissues it remains challenging to pinpoint niche-independent effects of circulating factors on specific cell populations. Here, we describe a method for the encapsulation of human and mouse skeletal muscle progenitors in diffusible polyethersulfone hollow fiber capsules that can be used to profile systemic aging in vivo independent of heterogeneous short-range tissue interactions. We observed that circulating long-range signaling factors in the old systemic environment lead to an activation of Myc and E2F transcription factors, induce senescence, and suppress myogenic differentiation. Importantly, in vitro profiling using young and old serum in 2D culture does not capture all pathways deregulated in encapsulated cells in aged mice. Thus, in vivo transcriptomic profiling using cell encapsulation allows for the characterization of effector pathways of systemic aging with unparalleled accuracy., Competing Interests: OM, XH, JM, GL, CB, FD, SM, FR, PD, NB, JF, CB Presently or previously employed by the Société des Produits Nestlé S.A., Switzerland, EM Presently or previously employed by the Société des Produits Nestlé; S.A., Switzerland, EL, JC, JI, PM No competing interests declared, (© 2022, Mashinchian et al.)
- Published
- 2022
- Full Text
- View/download PDF
19. Epigenome-wide association study of sarcopenia: findings from the Hertfordshire Sarcopenia Study (HSS).
- Author
-
Antoun E, Garratt ES, Taddei A, Burton MA, Barton SJ, Titcombe P, Westbury LD, Baczynska A, Migliavacca E, Feige JN, Sydall HE, Dennison E, Dodds R, Roberts HC, Richardson P, Sayer AA, Shaw S, Cooper C, Holbrook JD, Patel HP, Godfrey KM, and Lillycrop KA
- Subjects
- Aged, DNA Methylation, Epigenesis, Genetic, Hand Strength physiology, Humans, Male, Epigenome, Sarcopenia genetics
- Abstract
Background: Sarcopenia is the age-related loss of muscle mass, strength, and function. Epigenetic processes such as DNA methylation, which integrate both genetic and environmental exposures, have been suggested to contribute to the development of sarcopenia. This study aimed to determine whether differences in the muscle methylome are associated with sarcopenia and its component measures: grip strength, appendicular lean mass index (ALMi), and gait speed., Methods: Using the Infinium Human MethylationEPIC BeadChip, we measured DNA methylation in vastus lateralis muscle biopsies of 83 male participants (12 with sarcopenia) with a mean (standard deviation) age of 75.7 (3.6) years from the Hertfordshire Sarcopenia Study (HSS) and Hertfordshire Sarcopenia Study extension (HSSe) and examined associations with sarcopenia and its components. Pathway, histone mark, and transcription factor enrichment of the differentially methylated CpGs (dmCpGs) were determined, and sodium bisulfite pyrosequencing was used to validate the sarcopenia-associated dmCpGs. Human primary myoblasts (n = 6) isolated from vastus lateralis muscle biopsies from male individuals from HSSe were treated with the EZH2 inhibitor GSK343 to assess how perturbations in epigenetic processes may impact myoblast differentiation and fusion, measured by PAX7 and MYHC immunocytochemistry, and mitochondrial bioenergetics determined using the Seahorse XF96., Results: Sarcopenia was associated with differential methylation at 176 dmCpGs (false discovery rate ≤ 0.05) and 141 differentially methylated regions (Stouffer ≤ 0.05). The sarcopenia-associated dmCpGs were enriched in genes associated with myotube fusion (P = 1.40E-03), oxidative phosphorylation (P = 2.78E-02), and voltage-gated calcium channels (P = 1.59E-04). ALMi was associated with 71 dmCpGs, grip strength with 49 dmCpGs, and gait speed with 23 dmCpGs (false discovery rate ≤ 0.05). There was significant overlap between the dmCpGs associated with sarcopenia and ALMi (P = 3.4E-35), sarcopenia and gait speed (P = 4.78E-03), and sarcopenia and grip strength (P = 7.55E-06). There was also an over-representation of the sarcopenia, ALMi, grip strength, and gait speed-associated dmCpGs with sites of H3K27 trimethylation (all P ≤ 0.05) and amongst EZH2 target genes (all P ≤ 0.05). Furthermore, treatment of human primary myoblasts with the EZH2 inhibitor GSK343 inhibitor led to an increase in PAX7 expression (P ≤ 0.05), decreased myotube fusion (P = 0.043), and an increase in ATP production (P = 0.008), with alterations in the DNA methylation of genes involved in oxidative phosphorylation and myogenesis., Conclusions: These findings show that differences in the muscle methylome are associated with sarcopenia and individual measures of muscle mass, strength, and function in older individuals. This suggests that changes in the epigenetic regulation of genes may contribute to impaired muscle function in later life., (© 2021 The Authors. Journal of Cachexia, Sarcopenia and Muscle published by John Wiley & Sons Ltd on behalf of Society on Sarcopenia, Cachexia and Wasting Disorders.)
- Published
- 2022
- Full Text
- View/download PDF
20. Acute RyR1 Ca 2+ leak enhances NADH-linked mitochondrial respiratory capacity.
- Author
-
Zanou N, Dridi H, Reiken S, Imamura de Lima T, Donnelly C, De Marchi U, Ferrini M, Vidal J, Sittenfeld L, Feige JN, Garcia-Roves PM, Lopez-Mejia IC, Marks AR, Auwerx J, Kayser B, and Place N
- Subjects
- Animals, Calcium Signaling, Cell Line, Endoplasmic Reticulum metabolism, Energy Metabolism, Female, Humans, Male, Mice, Mice, Inbred C57BL, Muscle Weakness, Proteomics, Ryanodine Receptor Calcium Release Channel genetics, Sarcoplasmic Reticulum metabolism, Tacrolimus Binding Proteins, Calcium metabolism, Mitochondria metabolism, NAD metabolism, Ryanodine Receptor Calcium Release Channel metabolism
- Abstract
Sustained ryanodine receptor (RyR) Ca
2+ leak is associated with pathological conditions such as heart failure or skeletal muscle weakness. We report that a single session of sprint interval training (SIT), but not of moderate intensity continuous training (MICT), triggers RyR1 protein oxidation and nitrosylation leading to calstabin1 dissociation in healthy human muscle and in in vitro SIT models (simulated SIT or S-SIT). This is accompanied by decreased sarcoplasmic reticulum Ca2+ content, increased levels of mitochondrial oxidative phosphorylation proteins, supercomplex formation and enhanced NADH-linked mitochondrial respiratory capacity. Mechanistically, (S-)SIT increases mitochondrial Ca2+ uptake in mouse myotubes and muscle fibres, and decreases pyruvate dehydrogenase phosphorylation in human muscle and mouse myotubes. Countering Ca2+ leak or preventing mitochondrial Ca2+ uptake blunts S-SIT-induced adaptations, a result supported by proteomic analyses. Here we show that triggering acute transient Ca2+ leak through RyR1 in healthy muscle may contribute to the multiple health promoting benefits of exercise., (© 2021. The Author(s).)- Published
- 2021
- Full Text
- View/download PDF
21. Young extracellular vesicles rejuvenate aged muscle.
- Author
-
Ancel S and Feige JN
- Subjects
- Muscles, Extracellular Vesicles, Musculoskeletal System
- Published
- 2021
- Full Text
- View/download PDF
22. Muscle Stem Cell Quiescence: Controlling Stemness by Staying Asleep.
- Author
-
Ancel S, Stuelsatz P, and Feige JN
- Subjects
- Cell Division, Muscle, Skeletal, Myoblasts, Stem Cells
- Abstract
Muscle stem cells (MuSCs) are tissue-resident stem cells required for growth and repair of skeletal muscle, that are otherwise maintained in a cell-cycle-arrested state called quiescence. While quiescence was originally believed to be a state of cellular inactivity, increasing evidence suggests that quiescence is dynamically regulated and contributes to stemness, the long-term capacity to maintain regenerative functions. Here, we review the current understanding of MuSC quiescence and highlight recently discovered molecular markers, which differentiate depth of quiescence and influence self-renewal capacity. We also discuss how quiescent MuSCs integrate paracrine factors from their niche and dynamically regulate cell signaling, metabolism and proteostasis as they anticipate physiological needs, and how perturbing these cues during aging impairs muscle regeneration., Competing Interests: Declaration of Interests All authors are employees of Société des Produits Nestlé S.A., Switzerland., (Copyright © 2021 Elsevier Ltd. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
23. Nutritional Mediators of Cellular Decline and Mitochondrial Dysfunction in Older Adults.
- Author
-
Guralnik JM, Feige JN, Singh A, and Fielding RA
- Abstract
Aging is a primary risk factor for the progressive loss of function, disease onset, and increased vulnerability to negative health-related outcomes. These clinical manifestations arise in part from declines in mitochondrial, metabolic, and other processes considered to be hallmarks of aging. Collectively, these changes can be defined as age-associated cellular decline (AACD) and are often associated with fatigue, reduced strength, and low physical activity. This manuscript summarizes a recent Gerontological Society of America Annual Scientific Meeting symposium that explored mechanisms, clinical signs, and emerging cellular nutrition interventions for AACD. The session opened by highlighting results of an expert consensus that developed an initial framework to identify self-reported symptoms and observable signs of AACD in adults aged >50 years. Next, findings from the multi-ethnic molecular determinants of sarcopenia study were discussed, showing impaired mitochondrial bioenergetic capacity and NAD
+ metabolism in skeletal muscle of older adults with sarcopenia. Lastly, recent clinical evidence was presented linking urolithin A, a natural mitophagy activator, to improved mitochondrial and cellular health. The virtual panel discussed how stimulation of mitochondrial function via biological pathways, such as mitophagy and NAD+ augmentation, could improve cellular function and muscle health, potentially impacting clinical signs of AACD and overall healthy aging.- Published
- 2021
- Full Text
- View/download PDF
24. Mitochondrial Calcium Signaling in Pancreatic β-Cell.
- Author
-
Weiser A, Feige JN, and De Marchi U
- Subjects
- Animals, Diabetes Mellitus metabolism, Glucose metabolism, Humans, Calcium metabolism, Calcium Signaling physiology, Insulin-Secreting Cells metabolism, Mitochondria metabolism
- Abstract
Accumulation of calcium in energized mitochondria of pancreatic β-cells is emerging as a crucial process for pancreatic β-cell function. β-cell mitochondria sense and shape calcium signals, linking the metabolism of glucose and other secretagogues to the generation of signals that promote insulin secretion during nutrient stimulation. Here, we describe the role of mitochondrial calcium signaling in pancreatic β-cell function. We report the latest pharmacological and genetic findings, including the first mitochondrial calcium-targeted intervention strategies developed to modulate pancreatic β-cell function and their potential relevance in the context of diabetes.
- Published
- 2021
- Full Text
- View/download PDF
25. Molecular and phenotypic analysis of rodent models reveals conserved and species-specific modulators of human sarcopenia.
- Author
-
Börsch A, Ham DJ, Mittal N, Tintignac LA, Migliavacca E, Feige JN, Rüegg MA, and Zavolan M
- Subjects
- Age Factors, Aging genetics, Aging metabolism, Aging pathology, Animals, Body Composition, Disease Models, Animal, Disease Progression, Gene Expression Regulation, Humans, Male, Mice, Inbred C57BL, Muscle, Skeletal pathology, Muscle, Skeletal physiopathology, Phenotype, Rats, Sarcopenia pathology, Sarcopenia physiopathology, Signal Transduction, Species Specificity, Mice, Muscle, Skeletal metabolism, Sarcopenia genetics, Sarcopenia metabolism, Transcriptome
- Abstract
Sarcopenia, the age-related loss of skeletal muscle mass and function, affects 5-13% of individuals aged over 60 years. While rodents are widely-used model organisms, which aspects of sarcopenia are recapitulated in different animal models is unknown. Here we generated a time series of phenotypic measurements and RNA sequencing data in mouse gastrocnemius muscle and analyzed them alongside analogous data from rats and humans. We found that rodents recapitulate mitochondrial changes observed in human sarcopenia, while inflammatory responses are conserved at pathway but not gene level. Perturbations in the extracellular matrix are shared by rats, while mice recapitulate changes in RNA processing and autophagy. We inferred transcription regulators of early and late transcriptome changes, which could be targeted therapeutically. Our study demonstrates that phenotypic measurements, such as muscle mass, are better indicators of muscle health than chronological age and should be considered when analyzing aging-related molecular data.
- Published
- 2021
- Full Text
- View/download PDF
26. Targeting Mitochondrial Calcium Uptake with the Natural Flavonol Kaempferol, to Promote Metabolism/Secretion Coupling in Pancreatic β-cells.
- Author
-
Bermont F, Hermant A, Benninga R, Chabert C, Jacot G, Santo-Domingo J, Kraus MR, Feige JN, and De Marchi U
- Subjects
- Animals, Cell Culture Techniques, Humans, Mitochondria metabolism, Calcium metabolism, Calcium Channels metabolism, Insulin Secretion drug effects, Insulin-Secreting Cells drug effects, Kaempferols pharmacology
- Abstract
Pancreatic β-cells secrete insulin to lower blood glucose, following a meal. Maintenance of β-cell function is essential to preventing type 2 diabetes. In pancreatic β-cells, mitochondrial matrix calcium is an activating signal for insulin secretion. Recently, the molecular identity of the mitochondrial calcium uniporter (MCU), the transporter that mediates mitochondrial calcium uptake, was revealed. Its role in pancreatic β-cell signal transduction modulation was clarified, opening new perspectives for intervention. Here, we investigated the effects of a mitochondrial Ca
2+ -targeted nutritional intervention strategy on metabolism/secretion coupling, in a model of pancreatic insulin-secreting cells (INS-1E). Acute treatment of INS-1E cells with the natural plant flavonoid and MCU activator kaempferol, at a low micromolar range, increased mitochondrial calcium rise during glucose stimulation, without affecting the expression level of the MCU and with no cytotoxicity. Enhanced mitochondrial calcium rises potentiated glucose-induced insulin secretion. Conversely, the MCU inhibitor mitoxantrone inhibited mitochondrial Ca2+ uptake and prevented both glucose-induced insulin secretion and kaempferol-potentiated effects. The kaempferol-dependent potentiation of insulin secretion was finally validated in a model of a standardized pancreatic human islet. We conclude that the plant product kaempferol activates metabolism/secretion coupling in insulin-secreting cells by modulating mitochondrial calcium uptake., Competing Interests: The authors are employees of Nestlé Research, which is part of the Société des Produits Nestlé SA.- Published
- 2020
- Full Text
- View/download PDF
27. Mitochondrial oxidative capacity and NAD + biosynthesis are reduced in human sarcopenia across ethnicities.
- Author
-
Migliavacca E, Tay SKH, Patel HP, Sonntag T, Civiletto G, McFarlane C, Forrester T, Barton SJ, Leow MK, Antoun E, Charpagne A, Seng Chong Y, Descombes P, Feng L, Francis-Emmanuel P, Garratt ES, Giner MP, Green CO, Karaz S, Kothandaraman N, Marquis J, Metairon S, Moco S, Nelson G, Ngo S, Pleasants T, Raymond F, Sayer AA, Ming Sim C, Slater-Jefferies J, Syddall HE, Fang Tan P, Titcombe P, Vaz C, Westbury LD, Wong G, Yonghui W, Cooper C, Sheppard A, Godfrey KM, Lillycrop KA, Karnani N, and Feige JN
- Subjects
- Aged, Aged, 80 and over, Biopsy, Case-Control Studies, Energy Metabolism physiology, Humans, Jamaica, Male, Middle Aged, Mitochondria metabolism, Muscle, Skeletal cytology, Muscle, Skeletal metabolism, Oxidation-Reduction, Oxidative Phosphorylation, Oxidative Stress physiology, Proteostasis, Sarcopenia ethnology, Singapore, United Kingdom, Aging physiology, Mitochondria pathology, Muscle, Skeletal pathology, NAD biosynthesis, Sarcopenia pathology
- Abstract
The causes of impaired skeletal muscle mass and strength during aging are well-studied in healthy populations. Less is known on pathological age-related muscle wasting and weakness termed sarcopenia, which directly impacts physical autonomy and survival. Here, we compare genome-wide transcriptional changes of sarcopenia versus age-matched controls in muscle biopsies from 119 older men from Singapore, Hertfordshire UK and Jamaica. Individuals with sarcopenia reproducibly demonstrate a prominent transcriptional signature of mitochondrial bioenergetic dysfunction in skeletal muscle, with low PGC-1α/ERRα signalling, and downregulation of oxidative phosphorylation and mitochondrial proteostasis genes. These changes translate functionally into fewer mitochondria, reduced mitochondrial respiratory complex expression and activity, and low NAD
+ levels through perturbed NAD+ biosynthesis and salvage in sarcopenic muscle. We provide an integrated molecular profile of human sarcopenia across ethnicities, demonstrating a fundamental role of altered mitochondrial metabolism in the pathological loss of skeletal muscle mass and function in older people.- Published
- 2019
- Full Text
- View/download PDF
28. Latest advances in aging research and drug discovery.
- Author
-
Bakula D, Ablasser A, Aguzzi A, Antebi A, Barzilai N, Bittner MI, Jensen MB, Calkhoven CF, Chen D, Grey ADNJ, Feige JN, Georgievskaya A, Gladyshev VN, Golato T, Gudkov AV, Hoppe T, Kaeberlein M, Katajisto P, Kennedy BK, Lal U, Martin-Villalba A, Moskalev AA, Ozerov I, Petr MA, Reason, Rubinsztein DC, Tyshkovskiy A, Vanhaelen Q, Zhavoronkov A, and Scheibye-Knudsen M
- Subjects
- Drug Industry, Humans, Aging, Drug Discovery, Research
- Abstract
An increasing aging population poses a significant challenge to societies worldwide. A better understanding of the molecular, cellular, organ, tissue, physiological, psychological, and even sociological changes that occur with aging is needed in order to treat age-associated diseases. The field of aging research is rapidly expanding with multiple advances transpiring in many previously disconnected areas. Several major pharmaceutical, biotechnology, and consumer companies made aging research a priority and are building internal expertise, integrating aging research into traditional business models and exploring new go-to-market strategies. Many of these efforts are spearheaded by the latest advances in artificial intelligence, namely deep learning, including generative and reinforcement learning. To facilitate these trends, the Center for Healthy Aging at the University of Copenhagen and Insilico Medicine are building a community of Key Opinion Leaders (KOLs) in these areas and launched the annual conference series titled "Aging Research and Drug Discovery (ARDD)" held in the capital of the pharmaceutical industry, Basel, Switzerland (www.agingpharma.org). This ARDD collection contains summaries from the 6
th annual meeting that explored aging mechanisms and new interventions in age-associated diseases. The 7th annual ARDD exhibition will transpire 2nd -4th of September, 2020, in Basel.- Published
- 2019
- Full Text
- View/download PDF
29. Adipogenic progenitors keep muscle stem cells young.
- Author
-
Ancel S, Mashinchian O, and Feige JN
- Subjects
- Animals, Humans, Muscle, Skeletal cytology, Adipogenesis physiology, Aging physiology, Muscle, Skeletal physiology, Stem Cells physiology
- Published
- 2019
- Full Text
- View/download PDF
30. Aging Disrupts Muscle Stem Cell Function by Impairing Matricellular WISP1 Secretion from Fibro-Adipogenic Progenitors.
- Author
-
Lukjanenko L, Karaz S, Stuelsatz P, Gurriaran-Rodriguez U, Michaud J, Dammone G, Sizzano F, Mashinchian O, Ancel S, Migliavacca E, Liot S, Jacot G, Metairon S, Raymond F, Descombes P, Palini A, Chazaud B, Rudnicki MA, Bentzinger CF, and Feige JN
- Subjects
- Adipocytes cytology, Adipogenesis, Animals, CCN Intercellular Signaling Proteins deficiency, Cells, Cultured, Humans, Mice, Mice, Inbred C57BL, Mice, Knockout, Muscle, Skeletal cytology, Proto-Oncogene Proteins deficiency, Stem Cells cytology, Adipocytes metabolism, Aging metabolism, CCN Intercellular Signaling Proteins metabolism, Muscle, Skeletal metabolism, Proto-Oncogene Proteins metabolism, Stem Cells metabolism
- Abstract
Research on age-related regenerative failure of skeletal muscle has extensively focused on the phenotypes of muscle stem cells (MuSCs). In contrast, the impact of aging on regulatory cells in the MuSC niche remains largely unexplored. Here, we demonstrate that aging impairs the function of mouse fibro-adipogenic progenitors (FAPs) and thereby indirectly affects the myogenic potential of MuSCs. Using transcriptomic profiling, we identify WNT1 Inducible Signaling Pathway Protein 1 (WISP1) as a FAP-derived matricellular signal that is lost during aging. WISP1 is required for efficient muscle regeneration and controls the expansion and asymmetric commitment of MuSCs through Akt signaling. Transplantation of young FAPs or systemic treatment with WISP1 restores the myogenic capacity of MuSCs in aged mice and rescues skeletal muscle regeneration. Our work establishes that loss of WISP1 from FAPs contributes to MuSC dysfunction in aged skeletal muscles and demonstrates that this mechanism can be targeted to rejuvenate myogenesis., (Copyright © 2018 Elsevier Inc. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
31. Sex-Specific Associations of Blood-Based Nutrient Profiling With Body Composition in the Elderly.
- Author
-
Konz T, Santoro A, Goulet L, Bazzocchi A, Battista G, Nicoletti C, Kadi F, Ostan R, Goy M, Monnard C, Martin FP, Feige JN, Franceschi C, and Rezzi S
- Abstract
The intake of adequate amounts and types of nutrients is key for sustaining health and a good quality of life, particularly in the elderly population. There is considerable evidence suggesting that physiological changes related to age and sex modify nutritional needs, and this may be related to age-associated changes in body composition (BC), specifically in lean and fat body mass. However, there is a clear lack of understanding about the association of nutrients in blood and BC parameters in the elderly. This study investigated the relationships among blood nutrients (amino acids, fatty acids, major elements, trace-elements, and vitamins), BC and nutrient intake in a population of 176 healthy male and female Italian adults between the ages of 65 and 79 years. 89 blood markers, 77 BC parameters and dietary intake were evaluated. Multivariate data analysis was applied to infer relationships between datasets. As expected, the major variability between BC and the blood nutrient profile (BNP) observed was related to sex. Aside from clear sex-specific differences in BC, female subjects had higher BNP levels of copper, copper-to-zinc ratio, phosphorous and holotranscobalamin II and lower concentrations of branched-chain amino acids (BCAAs) and proline. Fat mass, percentage of fat mass, percentage of lean mass and the skeletal muscle index (SMI) correlated the most with BNP in both sexes. Our data showed positive correlations in male subjects among ethanolamine, glycine, albumin, and sulfur with SMI, while palmitoleic acid and oleic acid exhibited negative correlations. This differed in female subjects, where SMI was positively associated with albumin, folic acid and sulfur, while CRP, proline and cis -8,11,14-eicosatrienoic acid were negatively correlated. We investigated the influence of diet on the observed BNP and BC correlations. Intriguingly, most of the components of the BNP, except for folate, did not exhibit a correlation with nutrient intake data. An understanding of the physiological and biochemical processes underpinning the observed sex-specific correlations between BNP and BC could help in identifying nutritional strategies to manage BC-changes in aging. This would contribute to a deeper understanding of aging-associated nutritional needs with the aim of helping the elderly population to maintain metabolic health.
- Published
- 2019
- Full Text
- View/download PDF
32. The exerkine apelin reverses age-associated sarcopenia.
- Author
-
Vinel C, Lukjanenko L, Batut A, Deleruyelle S, Pradère JP, Le Gonidec S, Dortignac A, Geoffre N, Pereira O, Karaz S, Lee U, Camus M, Chaoui K, Mouisel E, Bigot A, Mouly V, Vigneau M, Pagano AF, Chopard A, Pillard F, Guyonnet S, Cesari M, Burlet-Schiltz O, Pahor M, Feige JN, Vellas B, Valet P, and Dray C
- Subjects
- Adenylate Kinase metabolism, Adolescent, Adult, Aged, Aged, 80 and over, Animals, Apelin biosynthesis, Apelin Receptors deficiency, Apelin Receptors metabolism, Body Weight, Exercise, Humans, Kinetics, Mice, Inbred C57BL, Muscle Cells metabolism, Muscle Weakness drug therapy, Muscle Weakness pathology, Muscle, Skeletal drug effects, Muscle, Skeletal metabolism, Organelle Biogenesis, Protein Biosynthesis, Proto-Oncogene Proteins c-akt metabolism, Regeneration, Ribosomal Protein S6 Kinases, 70-kDa metabolism, Sarcopenia pathology, Satellite Cells, Skeletal Muscle metabolism, Aging pathology, Apelin blood, Sarcopenia blood
- Abstract
Sarcopenia, the degenerative loss of skeletal muscle mass, quality and strength, lacks early diagnostic tools and new therapeutic strategies to prevent the frailty-to-disability transition often responsible for the medical institutionalization of elderly individuals. Herein we report that production of the endogenous peptide apelin, induced by muscle contraction, is reduced in an age-dependent manner in humans and rodents and is positively associated with the beneficial effects of exercise in older persons. Mice deficient in either apelin or its receptor (APLNR) presented dramatic alterations in muscle function with increasing age. Various strategies that restored apelin signaling during aging further demonstrated that this peptide considerably enhanced muscle function by triggering mitochondriogenesis, autophagy and anti-inflammatory pathways in myofibers as well as enhancing the regenerative capacity by targeting muscle stem cells. Taken together, these findings revealed positive regulatory feedback between physical activity, apelin and muscle function and identified apelin both as a tool for diagnosis of early sarcopenia and as the target of an innovative pharmacological strategy to prevent age-associated muscle weakness and restore physical autonomy.
- Published
- 2018
- Full Text
- View/download PDF
33. PPARγ Controls Ectopic Adipogenesis and Cross-Talks with Myogenesis During Skeletal Muscle Regeneration.
- Author
-
Dammone G, Karaz S, Lukjanenko L, Winkler C, Sizzano F, Jacot G, Migliavacca E, Palini A, Desvergne B, Gilardi F, and Feige JN
- Subjects
- Adipocytes cytology, Adipocytes metabolism, Animals, Cell Differentiation genetics, Cells, Cultured, Female, Gene Expression Regulation, Developmental, Male, Mice, Knockout, Mice, Transgenic, Muscle, Skeletal cytology, Muscle, Skeletal physiology, Myoblasts cytology, Myoblasts metabolism, PPAR gamma metabolism, Adipogenesis genetics, Muscle Development genetics, Muscle, Skeletal metabolism, PPAR gamma genetics, Regeneration genetics
- Abstract
Skeletal muscle is a regenerative tissue which can repair damaged myofibers through the activation of tissue-resident muscle stem cells (MuSCs). Many muscle diseases with impaired regeneration cause excessive adipose tissue accumulation in muscle, alter the myogenic fate of MuSCs, and deregulate the cross-talk between MuSCs and fibro/adipogenic progenitors (FAPs), a bi-potent cell population which supports myogenesis and controls intra-muscular fibrosis and adipocyte formation. In order to better characterize the interaction between adipogenesis and myogenesis, we studied muscle regeneration and MuSC function in whole body Pparg null mice generated by epiblast-specific Cre/lox deletion ( Pparg
Δ/Δ ). We demonstrate that deletion of PPARγ completely abolishes ectopic muscle adipogenesis during regeneration and impairs MuSC expansion and myogenesis after injury. Ex vivo assays revealed that perturbed myogenesis in PpargΔ/Δ mice does not primarily result from intrinsic defects of MuSCs or from perturbed myogenic support from FAPs. The immune transition from a pro- to anti-inflammatory MuSC niche during regeneration is perturbed in PpargΔ/Δ mice and suggests that PPARγ signaling in macrophages can interact with ectopic adipogenesis and influence muscle regeneration. Altogether, our study demonstrates that a PPARγ-dependent adipogenic response regulates muscle fat infiltration during regeneration and that PPARγ is required for MuSC function and efficient muscle repair.- Published
- 2018
- Full Text
- View/download PDF
34. Transcriptomic analyses reveal rhythmic and CLOCK-driven pathways in human skeletal muscle.
- Author
-
Perrin L, Loizides-Mangold U, Chanon S, Gobet C, Hulo N, Isenegger L, Weger BD, Migliavacca E, Charpagne A, Betts JA, Walhin JP, Templeman I, Stokes K, Thompson D, Tsintzas K, Robert M, Howald C, Riezman H, Feige JN, Karagounis LG, Johnston JD, Dermitzakis ET, Gachon F, Lefai E, and Dibner C
- Subjects
- Gene Expression Profiling, Glucose metabolism, Humans, Lipid Metabolism, CLOCK Proteins metabolism, Circadian Clocks, Metabolic Networks and Pathways, Muscle, Skeletal physiology
- Abstract
Circadian regulation of transcriptional processes has a broad impact on cell metabolism. Here, we compared the diurnal transcriptome of human skeletal muscle conducted on serial muscle biopsies in vivo with profiles of human skeletal myotubes synchronized in vitro. More extensive rhythmic transcription was observed in human skeletal muscle compared to in vitro cell culture as a large part of the in vivo mRNA rhythmicity was lost in vitro. siRNA-mediated clock disruption in primary myotubes significantly affected the expression of ~8% of all genes, with impact on glucose homeostasis and lipid metabolism. Genes involved in GLUT4 expression, translocation and recycling were negatively affected, whereas lipid metabolic genes were altered to promote activation of lipid utilization. Moreover, basal and insulin-stimulated glucose uptake were significantly reduced upon CLOCK depletion. Our findings suggest an essential role for the circadian coordination of skeletal muscle glucose homeostasis and lipid metabolism in humans., Competing Interests: LP, UL, SC, NH, LI, JW, IT, KS, DT, KT, MR, CH, HR, JJ, ED, FG, EL, CD No competing interests declared, CG Is a full-time employee of the Nestlé Institute of Health Sciences SA. BW Benjamin D Weger: Is a full-time employee of the Nestlé Institute of Health Sciences SA. EM Eugenia Migliavacca: Is a full-time employee of the Nestlé Institute of Health Sciences SA. AC Aline Charpagne: Is a full-time employee of the Nestlé Institute of Health Sciences SA. JB Has been a consultant for PepsiCo (Quaker) and Kellogg's. JF Jerome N Feige: Is a full-time employee of the Nestlé Institute of Health Sciences SA. LK Is an employee of Nestec Ltd., (© 2018, Perrin et al.)
- Published
- 2018
- Full Text
- View/download PDF
35. Vitamin B12 deficiency and impaired expression of amnionless during aging.
- Author
-
Pannérec A, Migliavacca E, De Castro A, Michaud J, Karaz S, Goulet L, Rezzi S, Ng TP, Bosco N, Larbi A, and Feige JN
- Subjects
- Adult, Aged, Aged, 80 and over, Aging, Animals, Female, Humans, Longitudinal Studies, Male, Membrane Proteins, Middle Aged, Rats, Rats, Wistar, Biomarkers blood, Methylmalonic Acid blood, Proteins metabolism, Vitamin B 12 blood, Vitamin B 12 Deficiency physiopathology
- Abstract
Background: Physical frailty and loss of mobility in elderly individuals lead to reduced independence, quality of life, and increased mortality. Vitamin B12 deficiency has been linked to several age-related chronic diseases, including in the musculo-skeletal system, where vitamin B12 deficiency is generally believed to be linked to poor nutritional intake. In the present study, we asked whether aging and frailty associate with altered vitamin B12 homeostasis in humans and investigated the underlying molecular mechanisms using preclinical models., Methods: We analysed a subset of the Singapore Longitudinal Aging Study and stratified 238 participants based on age and Fried frailty criteria. Levels of methyl-malonic acid (MMA), a marker for vitamin B12 deficiency, and amnionless, the vitamin B12 co-receptor that anchors the vitamin B12 transport complex to the membrane of epithelial cells, were measured in plasma. In addition, vitamin B12 levels and the molecular mechanisms of vitamin B12 uptake and excretion were analysed in ileum, kidney, liver, and blood using a rat model of natural aging where nutritional intake is fully controlled., Results: We demonstrate that aging and frailty are associated with a higher prevalence of functional vitamin B12 deficiency that can be detected by increased levels of MMA in blood (ρ = 0.25; P = 0.00013). The decline in circulating vitamin B12 levels is recapitulated in a rat model of natural aging where food composition and intake are stable. At the molecular level, these perturbations involve altered expression of amnionless in the ileum and kidney. Interestingly, we demonstrate that amnionless can be detected in serum where its levels increase during aging in both rodents and human (P = 3.3e-07 and 9.2e-07, respectively). Blood amnionless levels negatively correlate with vitamin B12 in rats (r
2 = 0.305; P = 0.0042) and positively correlate with the vitamin B12 deficiency marker MMA in humans (ρ = 0.22; P = 0.00068)., Conclusions: Our results demonstrate that aging and frailty cause intrinsic vitamin B12 deficiencies, which can occur independently of nutritional intake. Mechanistically, vitamin B12 deficiency involves the physio-pathological decline of both the intestinal uptake and the renal reabsorption system for vitamin B12. Finally, amnionless is a novel biomarker which can detect perturbed vitamin B12 bioavailability during aging and physical frailty., (© 2017 The Authors. Journal of Cachexia, Sarcopenia and Muscle published by John Wiley & Sons Ltd on behalf of the Society on Sarcopenia, Cachexia and Wasting Disorders.)- Published
- 2018
- Full Text
- View/download PDF
36. Aging and sarcopenia associate with specific interactions between gut microbes, serum biomarkers and host physiology in rats.
- Author
-
Siddharth J, Chakrabarti A, Pannérec A, Karaz S, Morin-Rivron D, Masoodi M, Feige JN, and Parkinson SJ
- Subjects
- Animals, Bacteria classification, Biomarkers blood, Genome, Bacterial, Genomics, Host-Pathogen Interactions, Rats, Aging physiology, Gastrointestinal Microbiome physiology, Sarcopenia
- Abstract
The microbiome has been demonstrated to play an integral role in the maintenance of many aspects of health that are also associated with aging. In order to identify areas of potential exploration and intervention, we simultaneously characterized age-related alterations in gut microbiome, muscle physiology and serum proteomic and lipidomic profiles in aged rats to define an integrated signature of the aging phenotype. We demonstrate that aging skews the composition of the gut microbiome, in particular by altering the Sutterella to Barneseilla ratio, and alters the metabolic potential of intestinal bacteria. Age-related changes of the gut microbiome were associated with the physiological decline of musculoskeletal function, and with molecular markers of nutrient processing/availability, and inflammatory/immune status in aged versus adult rats. Altogether, our study highlights that aging leads to a complex interplay between the microbiome and host physiology, and provides candidate microbial species to target physical and metabolic decline during aging by modulating gut microbial ecology.
- Published
- 2017
- Full Text
- View/download PDF
37. Loss of fibronectin from the aged stem cell niche affects the regenerative capacity of skeletal muscle in mice.
- Author
-
Lukjanenko L, Jung MJ, Hegde N, Perruisseau-Carrier C, Migliavacca E, Rozo M, Karaz S, Jacot G, Schmidt M, Li L, Metairon S, Raymond F, Lee U, Sizzano F, Wilson DH, Dumont NA, Palini A, Fässler R, Steiner P, Descombes P, Rudnicki MA, Fan CM, von Maltzahn J, Feige JN, and Bentzinger CF
- Subjects
- Animals, Blotting, Western, Extracellular Matrix metabolism, Fibronectins metabolism, Flow Cytometry, Integrins metabolism, Mice, Muscle, Skeletal cytology, Polymerase Chain Reaction, Aging metabolism, Fibronectins genetics, Focal Adhesion Protein-Tyrosine Kinases metabolism, Muscle, Skeletal metabolism, Regeneration genetics, Stem Cell Niche, p38 Mitogen-Activated Protein Kinases metabolism
- Abstract
Age-related changes in the niche have long been postulated to impair the function of somatic stem cells. Here we demonstrate that the aged stem cell niche in skeletal muscle contains substantially reduced levels of fibronectin (FN), leading to detrimental consequences for the function and maintenance of muscle stem cells (MuSCs). Deletion of the gene encoding FN from young regenerating muscles replicates the aging phenotype and leads to a loss of MuSC numbers. By using an extracellular matrix (ECM) library screen and pathway profiling, we characterize FN as a preferred adhesion substrate for MuSCs and demonstrate that integrin-mediated signaling through focal adhesion kinase and the p38 mitogen-activated protein kinase pathway is strongly de-regulated in MuSCs from aged mice because of insufficient attachment to the niche. Reconstitution of FN levels in the aged niche remobilizes stem cells and restores youth-like muscle regeneration. Taken together, we identify the loss of stem cell adhesion to FN in the niche ECM as a previously unknown aging mechanism.
- Published
- 2016
- Full Text
- View/download PDF
38. A robust neuromuscular system protects rat and human skeletal muscle from sarcopenia.
- Author
-
Pannérec A, Springer M, Migliavacca E, Ireland A, Piasecki M, Karaz S, Jacot G, Métairon S, Danenberg E, Raymond F, Descombes P, McPhee JS, and Feige JN
- Subjects
- Adolescent, Adult, Aged, Aged, 80 and over, Animals, Female, Gene Expression, Humans, Male, Muscle, Skeletal pathology, Muscular Atrophy pathology, Rats, Sarcopenia genetics, Sarcopenia pathology, Young Adult, Aging physiology, Muscle, Skeletal metabolism, Muscular Atrophy metabolism, Sarcopenia metabolism, Transcriptome
- Abstract
Declining muscle mass and function is one of the main drivers of loss of independence in the elderly. Sarcopenia is associated with numerous cellular and endocrine perturbations, and it remains challenging to identify those changes that play a causal role and could serve as targets for therapeutic intervention. In this study, we uncovered a remarkable differential susceptibility of certain muscles to age-related decline. Aging rats specifically lose muscle mass and function in the hindlimbs, but not in the forelimbs. By performing a comprehensive comparative analysis of these muscles, we demonstrate that regional susceptibility to sarcopenia is dependent on neuromuscular junction fragmentation, loss of motoneuron innervation, and reduced excitability. Remarkably, muscle loss in elderly humans also differs in vastus lateralis and tibialis anterior muscles in direct relation to neuromuscular dysfunction. By comparing gene expression in susceptible and non-susceptible muscles, we identified a specific transcriptomic signature of neuromuscular impairment. Importantly, differential molecular profiling of the associated peripheral nerves revealed fundamental changes in cholesterol biosynthetic pathways. Altogether our results provide compelling evidence that susceptibility to sarcopenia is tightly linked to neuromuscular decline in rats and humans, and identify dysregulation of sterol metabolism in the peripheral nervous system as an early event in this process.
- Published
- 2016
- Full Text
- View/download PDF
39. Gαi2 signaling is required for skeletal muscle growth, regeneration, and satellite cell proliferation and differentiation.
- Author
-
Minetti GC, Feige JN, Bombard F, Heier A, Morvan F, Nürnberg B, Leiss V, Birnbaumer L, Glass DJ, and Fornaro M
- Subjects
- Animals, Cell Differentiation physiology, Cells, Cultured, Humans, Mice, Mice, Inbred C57BL, Mice, Knockout, Muscle Development physiology, Muscle, Skeletal cytology, Myoblasts cytology, Myoblasts metabolism, Satellite Cells, Skeletal Muscle pathology, Signal Transduction genetics, Signal Transduction physiology, Cell Differentiation genetics, Cell Proliferation, GTP-Binding Protein alpha Subunit, Gi2 genetics, Muscle Development genetics, Muscle, Skeletal metabolism, Regeneration genetics, Satellite Cells, Skeletal Muscle metabolism
- Abstract
We have previously shown that activation of Gαi2, an α subunit of the heterotrimeric G protein complex, induces skeletal muscle hypertrophy and myoblast differentiation. To determine whether Gαi2 is required for skeletal muscle growth or regeneration, Gαi2-null mice were analyzed. Gαi2 knockout mice display decreased lean body mass, reduced muscle size, and impaired skeletal muscle regeneration after cardiotoxin-induced injury. Short hairpin RNA (shRNA)-mediated knockdown of Gαi2 in satellite cells (SCs) leads to defective satellite cell proliferation, fusion, and differentiation ex vivo. The impaired differentiation is consistent with the observation that the myogenic regulatory factors MyoD and Myf5 are downregulated upon knockdown of Gαi2. Interestingly, the expression of microRNA 1 (miR-1), miR-27b, and miR-206, three microRNAs that have been shown to regulate SC proliferation and differentiation, is increased by a constitutively active mutant of Gαi2 [Gαi2(Q205L)] and counterregulated by Gαi2 knockdown. As for the mechanism, this study demonstrates that Gαi2(Q205L) regulates satellite cell differentiation into myotubes in a protein kinase C (PKC)- and histone deacetylase (HDAC)-dependent manner.
- Published
- 2014
- Full Text
- View/download PDF
40. An antibody blocking activin type II receptors induces strong skeletal muscle hypertrophy and protects from atrophy.
- Author
-
Lach-Trifilieff E, Minetti GC, Sheppard K, Ibebunjo C, Feige JN, Hartmann S, Brachat S, Rivet H, Koelbing C, Morvan F, Hatakeyama S, and Glass DJ
- Subjects
- Activin Receptors, Type II metabolism, Animals, Antibodies, Blocking metabolism, Antibodies, Monoclonal metabolism, Antibodies, Monoclonal, Humanized, Atrophy immunology, Atrophy metabolism, Cell Differentiation physiology, Humans, Hypertrophy pathology, Mice, Muscle, Skeletal metabolism, Muscle, Skeletal pathology, Myoblasts, Skeletal immunology, Signal Transduction physiology, Smad2 Protein metabolism, Smad3 Protein metabolism, Activin Receptors, Type II immunology, Activins metabolism, Antibodies, Blocking pharmacology, Antibodies, Monoclonal pharmacology, Hypertrophy metabolism, Myoblasts, Skeletal metabolism
- Abstract
The myostatin/activin type II receptor (ActRII) pathway has been identified to be critical in regulating skeletal muscle size. Several other ligands, including GDF11 and the activins, signal through this pathway, suggesting that the ActRII receptors are major regulatory nodes in the regulation of muscle mass. We have developed a novel, human anti-ActRII antibody (bimagrumab, or BYM338) to prevent binding of ligands to the receptors and thus inhibit downstream signaling. BYM338 enhances differentiation of primary human skeletal myoblasts and counteracts the inhibition of differentiation induced by myostatin or activin A. BYM338 prevents myostatin- or activin A-induced atrophy through inhibition of Smad2/3 phosphorylation, thus sparing the myosin heavy chain from degradation. BYM338 dramatically increases skeletal muscle mass in mice, beyond sole inhibition of myostatin, detected by comparing the antibody with a myostatin inhibitor. A mouse version of the antibody induces enhanced muscle hypertrophy in myostatin mutant mice, further confirming a beneficial effect on muscle growth beyond myostatin inhibition alone through blockade of ActRII ligands. BYM338 protects muscles from glucocorticoid-induced atrophy and weakness via prevention of muscle and tetanic force losses. These data highlight the compelling therapeutic potential of BYM338 for the treatment of skeletal muscle atrophy and weakness in multiple settings.
- Published
- 2014
- Full Text
- View/download PDF
41. Genomic profiling reveals that transient adipogenic activation is a hallmark of mouse models of skeletal muscle regeneration.
- Author
-
Lukjanenko L, Brachat S, Pierrel E, Lach-Trifilieff E, and Feige JN
- Subjects
- Adipocytes pathology, Adipose Tissue pathology, Animals, Cardiotoxins, Disease Models, Animal, Fatty Acids metabolism, Gene Expression Profiling, Glycerol, Lipid Metabolism genetics, Male, Mice, Mice, Inbred C57BL, Muscle Proteins metabolism, Muscle Weakness chemically induced, Muscle Weakness pathology, Muscle, Skeletal pathology, Adipocytes metabolism, Adipogenesis genetics, Adipose Tissue metabolism, Muscle Proteins genetics, Muscle Weakness genetics, Muscle, Skeletal metabolism, Regeneration genetics
- Abstract
The marbling of skeletal muscle by ectopic adipose tissue is a hallmark of many muscle diseases, including sarcopenia and muscular dystrophies, and generally associates with impaired muscle regeneration. Although the etiology and the molecular mechanisms of ectopic adipogenesis are poorly understood, fatty regeneration can be modeled in mice using glycerol-induced muscle damage. Using comprehensive molecular and histological profiling, we compared glycerol-induced fatty regeneration to the classical cardiotoxin (CTX)-induced regeneration model previously believed to lack an adipogenic response in muscle. Surprisingly, ectopic adipogenesis was detected in both models, but was stronger and more persistent in response to glycerol. Importantly, extensive differential transcriptomic profiling demonstrated that glycerol induces a stronger inflammatory response and promotes adipogenic regulatory networks while reducing fatty acid β-oxidation. Altogether, these results provide a comprehensive mapping of gene expression changes during the time course of two muscle regeneration models, and strongly suggest that adipogenic commitment is a hallmark of muscle regeneration, which can lead to ectopic adipocyte accumulation in response to specific physio-pathological challenges.
- Published
- 2013
- Full Text
- View/download PDF
42. The corepressor NCoR1 antagonizes PGC-1α and estrogen-related receptor α in the regulation of skeletal muscle function and oxidative metabolism.
- Author
-
Pérez-Schindler J, Summermatter S, Salatino S, Zorzato F, Beer M, Balwierz PJ, van Nimwegen E, Feige JN, Auwerx J, and Handschin C
- Subjects
- Animals, Male, Mice, Mice, Knockout, Mice, Transgenic, Models, Biological, Muscle Contraction genetics, Muscle Contraction physiology, Muscle, Skeletal metabolism, Nuclear Receptor Co-Repressor 1 deficiency, Nuclear Receptor Co-Repressor 1 genetics, Oxidative Phosphorylation, Oxygen Consumption, PPAR delta metabolism, PPAR-beta metabolism, Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha, RNA, Messenger genetics, RNA, Messenger metabolism, Receptors, Estrogen antagonists & inhibitors, Receptors, Estrogen genetics, Trans-Activators antagonists & inhibitors, Trans-Activators genetics, Transcription Factors, ERRalpha Estrogen-Related Receptor, Muscle, Skeletal physiology, Nuclear Receptor Co-Repressor 1 metabolism, Receptors, Estrogen metabolism, Trans-Activators metabolism
- Abstract
Skeletal muscle exhibits a high plasticity and accordingly can quickly adapt to different physiological and pathological stimuli by changing its phenotype largely through diverse epigenetic mechanisms. The nuclear receptor corepressor 1 (NCoR1) has the ability to mediate gene repression; however, its role in regulating biological programs in skeletal muscle is still poorly understood. We therefore studied the mechanistic and functional aspects of NCoR1 function in this tissue. NCoR1 muscle-specific knockout mice exhibited a 7.2% higher peak oxygen consumption (VO(2peak)), a 11% reduction in maximal isometric force, and increased ex vivo fatigue resistance during maximal stimulation. Interestingly, global gene expression analysis revealed a high overlap between the effects of NCoR1 deletion and peroxisome proliferator-activated receptor gamma (PPARγ) coactivator 1α (PGC-1α) overexpression on oxidative metabolism in muscle. Importantly, PPARβ/δ and estrogen-related receptor α (ERRα) were identified as common targets of NCoR1 and PGC-1α with opposing effects on the transcriptional activity of these nuclear receptors. In fact, the repressive effect of NCoR1 on oxidative phosphorylation gene expression specifically antagonizes PGC-1α-mediated coactivation of ERRα. We therefore delineated the molecular mechanism by which a transcriptional network controlled by corepressor and coactivator proteins determines the metabolic properties of skeletal muscle, thus representing a potential therapeutic target for metabolic diseases.
- Published
- 2012
- Full Text
- View/download PDF
43. Blockade of the activin receptor IIb activates functional brown adipogenesis and thermogenesis by inducing mitochondrial oxidative metabolism.
- Author
-
Fournier B, Murray B, Gutzwiller S, Marcaletti S, Marcellin D, Bergling S, Brachat S, Persohn E, Pierrel E, Bombard F, Hatakeyama S, Trendelenburg AU, Morvan F, Richardson B, Glass DJ, Lach-Trifilieff E, and Feige JN
- Subjects
- Activin Receptors, Type II immunology, Activin Receptors, Type II metabolism, Adipocytes, Brown cytology, Adipocytes, Brown metabolism, Adipose Tissue, Brown ultrastructure, Animals, Antibodies, Neutralizing, Cell Differentiation, Energy Metabolism, Female, Male, Mice, Mice, Inbred C57BL, Mice, SCID, Mice, Transgenic, Microscopy, Electron, Transmission, Mitochondria metabolism, Muscle, Skeletal metabolism, Myostatin metabolism, Signal Transduction, Smad3 Protein metabolism, Transcription Factors metabolism, Activin Receptors, Type II antagonists & inhibitors, Adipogenesis physiology, Adipose Tissue, Brown metabolism, Thermogenesis physiology
- Abstract
Brown adipose tissue (BAT) is a key tissue for energy expenditure via fat and glucose oxidation for thermogenesis. In this study, we demonstrate that the myostatin/activin receptor IIB (ActRIIB) pathway, which serves as an important negative regulator of muscle growth, is also a negative regulator of brown adipocyte differentiation. In parallel to the anticipated hypertrophy of skeletal muscle, the pharmacological inhibition of ActRIIB in mice, using a neutralizing antibody, increases the amount of BAT without directly affecting white adipose tissue. Mechanistically, inhibition of ActRIIB inhibits Smad3 signaling and activates the expression of myoglobin and PGC-1 coregulators in brown adipocytes. Consequently, ActRIIB blockade in brown adipose tissue enhances mitochondrial function and uncoupled respiration, translating into beneficial functional consequences, including enhanced cold tolerance and increased energy expenditure. Importantly, ActRIIB inhibition enhanced energy expenditure only at ambient temperature or in the cold and not at thermoneutrality, where nonshivering thermogenesis is minimal, strongly suggesting that brown fat activation plays a prominent role in the metabolic actions of ActRIIB inhibition.
- Published
- 2012
- Full Text
- View/download PDF
44. TAK-1/p38/nNFκB signaling inhibits myoblast differentiation by increasing levels of Activin A.
- Author
-
Trendelenburg AU, Meyer A, Jacobi C, Feige JN, and Glass DJ
- Abstract
Background: Skeletal-muscle differentiation is required for the regeneration of myofibers after injury. The differentiation capacity of satellite cells is impaired in settings of old age, which is at least one factor in the onset of sarcopenia, the age-related loss of skeletal-muscle mass and major cause of frailty. One important cause of impaired regeneration is increased levels of transforming growth factor (TGF)-β accompanied by reduced Notch signaling. Pro-inflammatory cytokines are also upregulated in aging, which led us hypothesize that they might potentially contribute to impaired regeneration in sarcopenia. Thus, in this study, we further analyzed the muscle differentiation-inhibition pathway mediated by pro-inflammatory cytokines in human skeletal muscle cells (HuSKMCs)., Methods: We studied the modulation of HuSKMC differentiation by the pro-inflammatory cytokines interleukin (IL)-1α and tumor necrosis factor (TNF)-α The grade of differentiation was determined by either imaging (fusion index) or creatine kinase (CK) activity, a marker of muscle differentiation. Secretion of TGF-β proteins during differentiation was assessed by using a TGF-β-responsive reporter-gene assay and further identified by means of pharmacological and genetic inhibitors. In addition, signaling events were monitored by western blotting and reverse transcription PCR, both in HuSKMC cultures and in samples from a rat sarcopenia study., Results: The pro-inflammatory cytokines IL-1α and TNF-α block differentiation of human myoblasts into myotubes. This anti-differentiation effect requires activation of TGF-β-activated kinase (TAK)-1. Using pharmacological and genetic inhibitors, the TAK-1 pathway could be traced to p38 and NFκB. Surprisingly, the anti-differentiation effect of the cytokines required the transcriptional upregulation of Activin A, which in turn acted through its established signaling pathway: ActRII/ALK/SMAD. Inhibition of Activin A signaling was able to rescue human myoblasts treated with IL-1β or TNF-α, resulting in normal differentiation into myotubes. Studies in aged rats as a model of sarcopenia confirmed that this pro-inflammatory cytokine pathway identified is activated during aging., Conclusions: In this study, we found an unexpected connection between cytokine and Activin signaling, revealing a new mechanism by which cytokines affect skeletal muscle, and establishing the physiologic relevance of this pathway in the impaired regeneration seen in sarcopenia.
- Published
- 2012
- Full Text
- View/download PDF
45. Gαi2 signaling promotes skeletal muscle hypertrophy, myoblast differentiation, and muscle regeneration.
- Author
-
Minetti GC, Feige JN, Rosenstiel A, Bombard F, Meier V, Werner A, Bassilana F, Sailer AW, Kahle P, Lambert C, Glass DJ, and Fornaro M
- Subjects
- Animals, Enzyme Activation genetics, GTP-Binding Protein alpha Subunits, G12-G13 genetics, HEK293 Cells, Humans, Hypertrophy enzymology, Hypertrophy genetics, Hypertrophy pathology, Mice, Mice, Transgenic, Muscle Proteins genetics, Muscle Proteins metabolism, Muscular Atrophy enzymology, Muscular Atrophy genetics, Muscular Atrophy pathology, Mutation, Myoblasts, Skeletal pathology, NFATC Transcription Factors genetics, NFATC Transcription Factors metabolism, Protein Kinase C genetics, Protein Kinase C metabolism, Tripartite Motif Proteins, Tumor Necrosis Factor-alpha genetics, Tumor Necrosis Factor-alpha metabolism, Ubiquitin-Protein Ligases genetics, Ubiquitin-Protein Ligases metabolism, Cell Differentiation, GTP-Binding Protein alpha Subunits, G12-G13 metabolism, Myoblasts, Skeletal enzymology, Regeneration, Signal Transduction
- Abstract
Skeletal muscle atrophy results in loss of strength and an increased risk of mortality. We found that lysophosphatidic acid, which activates a G protein (heterotrimeric guanine nucleotide-binding protein)-coupled receptor, stimulated skeletal muscle hypertrophy through activation of Gα(i2). Expression of a constitutively active mutant of Gα(i2) stimulated myotube growth and differentiation, effects that required the transcription factor NFAT (nuclear factor of activated T cells) and protein kinase C. In addition, expression of the constitutively active Gα(i2) mutant inhibited atrophy caused by the cachectic cytokine TNFα (tumor necrosis factor-α) by blocking an increase in the abundance of the mRNA encoding the E3 ubiquitin ligase MuRF1 (muscle ring finger 1). Gα(i2) activation also enhanced muscle regeneration and caused a switch to oxidative fibers. Our study thus identifies a pathway that promotes skeletal muscle hypertrophy and differentiation and demonstrates that Gα(i2)-induced signaling can act as a counterbalance to MuRF1-mediated atrophy, indicating that receptors that act through Gα(i2) might represent potential targets for preventing skeletal muscle wasting.
- Published
- 2011
- Full Text
- View/download PDF
46. CREB and ChREBP oppositely regulate SIRT1 expression in response to energy availability.
- Author
-
Noriega LG, Feige JN, Canto C, Yamamoto H, Yu J, Herman MA, Mataki C, Kahn BB, and Auwerx J
- Subjects
- Animals, Basic Helix-Loop-Helix Leucine Zipper Transcription Factors, CHO Cells, Cell Line, Tumor, Cricetinae, Fasting, Hep G2 Cells, Humans, Male, Mice, Mice, Inbred C57BL, Sirtuin 1 metabolism, Transcriptional Activation, Cyclic AMP Response Element-Binding Protein metabolism, Gene Expression Regulation, Nuclear Proteins metabolism, Sirtuin 1 genetics, Transcription Factors metabolism
- Abstract
The nicotinamide adenine dinucleotide (NAD(+))-dependent deacetylase SIRT1 is a major metabolic regulator activated by energy stresses such as fasting or calorie restriction. SIRT1 activation during fasting not only relies on the increase in the NAD(+)/NADH ratio caused by energy deprivation but also involves an upregulation of SIRT1 mRNA and protein levels in various metabolic tissues. We demonstrate that SIRT1 expression is controlled systemically by the activation of the cyclic AMP response-element-binding protein upon low nutrient availability. Conversely, in the absence of energetic stress, the carbohydrate response-element-binding protein represses the expression of SIRT1. Altogether, these results demonstrate that SIRT1 expression is tightly controlled at the transcriptional level by nutrient availability and further underscore that SIRT1 is a crucial metabolic checkpoint connecting the energetic status with transcriptional programmes.
- Published
- 2011
- Full Text
- View/download PDF
47. Assessment of Spontaneous Locomotor and Running Activity in Mice.
- Author
-
Thomas C, Marcaletti S, and Feige JN
- Abstract
The locomotor activity of laboratory mice is a global behavioral trait which can be valuable for the primary phenotyping of genetically engineered mouse models as well as mouse models of pathologies affecting the central and peripheral nervous systems, the musculoskeletal system, and the control of energy homeostasis. Basal levels of mouse locomotion can be recorded using infrared monitoring of movements, and further information can be gathered by giving the animal access to a running wheel, which will greatly enhance its spontaneous physical activity. Described here are two detailed protocols to evaluate basal locomotor activity and spontaneous wheel running. Curr. Protoc. Mouse Biol. 1:185-198. © 2011 by John Wiley & Sons, Inc., (Copyright © 2011 John Wiley & Sons, Inc.)
- Published
- 2011
- Full Text
- View/download PDF
48. Exercise Performance Tests in Mice.
- Author
-
Marcaletti S, Thomas C, and Feige JN
- Abstract
Maximal exercise performance is a multifactorial process in which the cardiovascular component, the innervation of the musculature, and the contractile and metabolic properties of skeletal muscle all play key roles. Here, protocols are provided for assessment of maximal running capacity of mice on a treadmill, with a combination of short high-intensity paradigms primarily intended to test for maximal power and cardiovascular function, and longer low-intensity paradigms to assess endurance and oxidative metabolism in skeletal muscle. The coupling of treadmill running to indirect calorimetry, to correlate performance measurements to maximal oxygen consumption, is also described. Curr. Protoc. Mouse Biol. 1:141-154. © 2011 by John Wiley & Sons, Inc., (Copyright © 2011 John Wiley & Sons, Inc.)
- Published
- 2011
- Full Text
- View/download PDF
49. PPAR-mediated activity of phthalates: A link to the obesity epidemic?
- Author
-
Desvergne B, Feige JN, and Casals-Casas C
- Subjects
- Animals, Endocrine Disruptors chemistry, Endocrine System metabolism, Environmental Pollutants chemistry, Humans, Ligands, Phthalic Acids chemistry, Receptors, Cytoplasmic and Nuclear metabolism, Xenobiotics chemistry, Xenobiotics metabolism, Xenobiotics pharmacology, Endocrine Disruptors pharmacology, Endocrine System drug effects, Environmental Pollutants pharmacology, Obesity metabolism, Peroxisome Proliferator-Activated Receptors metabolism, Phthalic Acids pharmacology
- Abstract
The endocrine disruption hypothesis asserts that exposure to small amounts of some chemicals in the environment may interfere with the endocrine system and lead to harmful effects in wildlife and humans. Many of these chemicals may interact with members of the nuclear receptor superfamily. Peroxisome proliferator-activated receptors (PPARs) are such candidate members, which interact with many different endogenous and exogenous lipophilic compounds. More particularly, the roles of PPARs in lipid and carbohydrate metabolism raise the question of their activation by a sub-class of pollutants, tentatively named "metabolic disrupters". Phthalates are abundant environmental micro-pollutants in Europe and North America and may belong to this class. Mono-ethyl-hexyl-phthalate (MEHP), a metabolite of the widespread plasticizer di-ethyl-hexyl-phthalate (DEHP), has been found in exposed organisms and interacts with all three PPARs. A thorough analysis of its interactions with PPARgamma identified MEHP as a selective PPARgamma modulator, and thus a possible contributor to the obesity epidemic.
- Published
- 2009
- Full Text
- View/download PDF
50. AMPK regulates energy expenditure by modulating NAD+ metabolism and SIRT1 activity.
- Author
-
Cantó C, Gerhart-Hines Z, Feige JN, Lagouge M, Noriega L, Milne JC, Elliott PJ, Puigserver P, and Auwerx J
- Subjects
- Acetylation, Aminoimidazole Carboxamide analogs & derivatives, Animals, Cell Line, Enzyme Activation, Forkhead Box Protein O1, Forkhead Box Protein O3, Forkhead Transcription Factors genetics, Gene Expression Regulation, Genes, Mitochondrial genetics, Male, Mice, Muscle, Skeletal cytology, Muscle, Skeletal enzymology, Muscle, Skeletal metabolism, Mutation, Oxygen Consumption, Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha, Phosphorylation, Ribonucleotides, Sirtuin 1, Trans-Activators genetics, Trans-Activators metabolism, Transcription Factors, Transcription, Genetic, AMP-Activated Protein Kinases metabolism, Energy Metabolism genetics, NAD metabolism, Sirtuins metabolism
- Abstract
AMP-activated protein kinase (AMPK) is a metabolic fuel gauge conserved along the evolutionary scale in eukaryotes that senses changes in the intracellular AMP/ATP ratio. Recent evidence indicated an important role for AMPK in the therapeutic benefits of metformin, thiazolidinediones and exercise, which form the cornerstones of the clinical management of type 2 diabetes and associated metabolic disorders. In general, activation of AMPK acts to maintain cellular energy stores, switching on catabolic pathways that produce ATP, mostly by enhancing oxidative metabolism and mitochondrial biogenesis, while switching off anabolic pathways that consume ATP. This regulation can take place acutely, through the regulation of fast post-translational events, but also by transcriptionally reprogramming the cell to meet energetic needs. Here we demonstrate that AMPK controls the expression of genes involved in energy metabolism in mouse skeletal muscle by acting in coordination with another metabolic sensor, the NAD+-dependent type III deacetylase SIRT1. AMPK enhances SIRT1 activity by increasing cellular NAD+ levels, resulting in the deacetylation and modulation of the activity of downstream SIRT1 targets that include the peroxisome proliferator-activated receptor-gamma coactivator 1alpha and the forkhead box O1 (FOXO1) and O3 (FOXO3a) transcription factors. The AMPK-induced SIRT1-mediated deacetylation of these targets explains many of the convergent biological effects of AMPK and SIRT1 on energy metabolism.
- Published
- 2009
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.