Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Christelle Guillet is active.

Publication


Featured researches published by Christelle Guillet.


The FASEB Journal | 2004

Impaired anabolic response of muscle protein synthesis is associated with S6K1 dysregulation in elderly humans

Christelle Guillet; Magali Prod'homme; Michèle Balage; Pierre Gachon; Christophe Giraudet; Liliane Morin; Jean Grizard; Yves Boirie

Age‐related loss of muscle protein may involve a decreased response to anabolic factors of muscle protein synthesis through dysregulation of translation factors. To verify this hypothesis, we simultaneously investigated muscle protein synthesis and expression of some factors implicated in insulin signal transduction during hyperinsulinemia and hyperaminoacidemia in 6 young (25±1 year; mean±SEM) and 8 elderly subjects (72±2 year). Incorporation of L‐[1‐13C] leucine in muscle proteins (fractional synthesis rate, FSR) was measured in vastus lateralis, before and during a euglycemic hyperinsulinemic hyperaminoacidemic clamp, together with Western blot analysis of protein kinase B (PKB), mTOR, 4E‐BP1, and S6K1 phosphorylation. In basal state, muscle protein FSR was reduced in elderly in comparison with young subjects (0.061±0.004% per hour) vs 0.082±0.010% per hour, elderly vs. young, P<0.05). During clamp, muscle protein FSR was stimulated in young (0.119±0.006% per hour; P<0.05), but this response was significantly lower in elderly subjects (0.084±0.005% per hour, P<0.05 vs young subjects). Phosphorylation of PKB, mTOR, and 4E‐BP1 were similarly increased by insulin and amino acid in both groups, except for S6K1 phosphorylation, which was not stimulated in elderly subjects. In conclusion, 1) response of muscle protein synthesis to insulin and amino acid is impaired in elderly humans; 2) a defect in S6K1 pathway activation may be responsible for this alteration. This modification is a mechanistic basis of sarcopenia development during aging.


The Journal of Physiology | 2003

The rate of protein digestion affects protein gain differently during aging in humans

Martial Dangin; Christelle Guillet; Clara Lucia Garcia-Rodenas; Pierre Gachon; Corinne Bouteloup-Demange; Kristel Reiffers-Magnani; Jacques Fauquant; Olivier Ballevre; Bernard Beaufrere

In young men ingesting protein meals, slowly digested proteins (caseins: CAS) induce a higher protein gain than those that are rapidly digested (whey proteins: WP). Our aim was to assess whether or not this is true in elderly men receiving mixed meals. The effects of meals containing either CAS or two different amounts of WP (WP‐iN: isonitrogenous with CAS, or WP‐iL: providing the same amount of leucine as CAS) on protein metabolism (assessed by combining oral and intravenous leucine tracers) were compared in nine healthy, elderly (mean ±s.e.m. age 72 ± 1 years) and six young men (24 ± 1 years). In both age groups, WP‐iL and WP‐iN were digested faster than CAS (P < 0.001, ANOVA). Proteolysis was inhibited similarly whatever the meal and age groups (P= NS). Protein synthesis was higher with WP‐iN than with CAS or WP‐iL (P < 0.01), irrespective of age (P= NS). An age‐related effect (P < 0.05) was found with postprandial leucine balance. Leucine balance was higher with CAS than with WP‐iL (P < 0.01) in young men, but not in elderly subjects (P= NS). In isonitrogenous conditions, leucine balance was higher with WP‐iN than with CAS (P < 0.001) in both age groups, but the magnitude of the differences was higher in the elderly men (P= 0.05). In conclusion, during aging, protein gain was greater with WP (rapidly digested protein), and lower with CAS (slowly digested protein). This suggests that a ‘fast’ protein might be more beneficial than a ‘slow’ one to limit protein losses during aging.


The Journal of Clinical Endocrinology and Metabolism | 2009

Changes in Basal and Insulin and Amino Acid Response of Whole Body and Skeletal Muscle Proteins in Obese Men

Christelle Guillet; Ingrid Delcourt; Mélanie Rance; Christophe Giraudet; Stéphane Walrand; M. Bedu; Pascale Duché; Yves Boirie

CONTEXT Obesity-related insulin resistance of glucose and lipid metabolism may also affect protein kinetics, notably at the muscle level. OBJECTIVE We hypothesized that muscle protein response to insulin and amino acid is blunted during obesity. RESEARCH DESIGN AND METHODS Total (Tot) and mitochondrial (Mit) muscle proteins fractional synthesis rates (FSR) together with whole-body protein kinetics (WB) have been determined in postabsorptive state (PA) and during a hyperinsulinemic, hyperaminoacidemic, euglycemic clamp by using a continuous infusion of (13)C-leucine in six obese and eight nonobese subjects. RESULTS Responses of WB glucose disposal rate and protein breakdown to insulin and amino acid infusion were significantly lower in obese than in nonobese subjects (P < 0.05). In PA, Tot and Mit FSR were significantly lower (P < 0.05) in obese (Tot, 0.044 +/- 0.005% . h(-1); Mit, 0.064 +/- 0.008% . h(-1)) in comparison with nonobese subjects (Tot, 0.082 +/- 0.010% . h(-1); Mit, 0.140 +/- 0.006% . h(-1)). Tot FSR was similarly stimulated by insulin and amino acid in both groups (0.094 +/- 0.013 vs. 0.117 +/- 0.006% . h(-1), obese vs. nonobese; P < 0.05). Mit FSR was increased in nonobese subjects (0.179 +/- 0.007% . h(-1); P < 0.05) but not in obese subjects (0.078 +/- 0.012% . h(-1); P = not significant). CONCLUSIONS The obesity-related impairment of protein metabolism is characterized by 1) a reduced turnover rate of skeletal muscle proteins in PA; 2) a lack of stimulation of mitochondrial protein synthesis by insulin and amino acid; and 3) a lower inhibition of WB proteolysis by insulin and amino acid. Alterations of selective muscle protein kinetics may predispose obese subjects to muscle metabolic dysfunction leading to type 2 diabetes.


Clinics in Geriatric Medicine | 2011

Physiopathological Mechanism of Sarcopenia

Stéphane Walrand; Christelle Guillet; Jérôme Salles; Noël Cano; Yves Boirie

The etiology of sarcopenia is multifactorial but still poorly understood, and the sequelae of this phenomenon represent a major public health issue. Age-related loss of muscle mass can be counteracted by adequate metabolic interventions including nutritional intake and exercise training. Other strategies including changes in daily protein pattern, the speed of protein digestion, or specific amino acid supplementation may be beneficial to improve short-term muscle anabolic response in elderly people. A multimodal approach combining nutrition, exercise, hormones, and specific anabolic drugs may be an innovative treatment for limiting the development of sarcopenia with aging.


Journal of Leukocyte Biology | 2004

In vivo evidences that insulin regulates human polymorphonuclear neutrophil functions.

Stéphane Walrand; Christelle Guillet; Yves Boirie; Marie-Paule Vasson

Polymorphonuclear neutrophils (PMN) are able to destroy invasive mircoorganisms by a wide variety of functions. Whereas insulin does not stimulate hexose transport in PMN, previous reports have clearly shown that this hormone regulates glucose metabolism inside this cell, raising the question of insulin action on PMN functions in humans. It is interesting that in vitro studies established a strong relationship between specific binding of insulin to its PMN membrane receptor and the activation of the main PMN functions. Therefore, investigation in healthy subjects under strict euglycemia and physiological insulinemia was performed to understand the in vivo‐specific action of insulin on PMN functions without hyperglycemia interferences. We determined numerous PMN functions before and after hyperinsulinemia (0.5 mU/kg/min) and euglycemia (0.9 g/l) clamp for 4 h in eight adult healthy volunteers (24±6 years). The total number of PMN and the number of PMN expressing CD11b, CD15, CD62L, and CD89 were significantly increased over baseline (P<0.001), whereas the density of these receptors was down‐regulated (P<0.01) by insulin. PMN chemotaxis (+117%, P<0.05), phagocytosis (+29%, P<0.001), and bactericidal (+17–25%, P<0.001) capacities were increased during the insulin clamp (P<0.05). Therefore, insulin treatment may modulate PMN functions not only by attainment of a better metabolic control, as suggested by in vivo studies in diabetic patients, but also through a direct effect of insulin.


Aging Cell | 2014

Muscle ectopic fat deposition contributes to anabolic resistance in obese sarcopenic old rats through eIF2α activation.

Nicolas Tardif; Jérôme Salles; Christelle Guillet; Joan Tordjman; Sophie Reggio; Jean-François Landrier; Christophe Giraudet; Véronique Patrac; Justine Bertrand-Michel; Carole Migné; Marie-Laure Collin; Jean-Michel Chardigny; Yves Boirie; Stéphane Walrand

Obesity and aging are characterized by decreased insulin sensitivity (IS) and muscle protein synthesis. Intramuscular ceramide accumulation has been implicated in insulin resistance during obesity. We aimed to measure IS, muscle ceramide level, protein synthesis, and activation of intracellular signaling pathways involved in translation initiation in male Wistar young (YR, 6‐month) and old (OR, 25‐month) rats receiving a low‐ (LFD) or a high‐fat diet (HFD) for 10 weeks. A corresponding cellular approach using C2C12 myotubes treated with palmitate to induce intracellular ceramide deposition was taken. A decreased ability of adipose tissue to store lipids together with a reduced adipocyte diameter and a development of fibrosis were observed in OR after the HFD. Consequently, OR fed the HFD were insulin resistant, showed a strong increase in intramuscular ceramide level and a decrease in muscle protein synthesis associated with increased eIF2α phosphorylation. The accumulation of intramuscular lipids placed a lipid burden on mitochondria and created a disconnect between metabolic and regulating pathways in skeletal muscles of OR. In C2C12 cells, palmitate‐induced ceramide accumulation was associated with a decreased protein synthesis together with upregulated eIF2α phosphorylation. In conclusion, a reduced ability to expand adipose tissues was found in OR, reflecting a lower lipid buffering capacity. Muscle mitochondrial activity was affected in OR conferring a reduced ability to oxidize fatty acids entering the muscle cell. Hence, OR were more prone to ectopic muscle lipid accumulation than YR, leading to decreased muscle protein anabolism. This metabolic change is a potential therapeutic target to counter sarcopenic obesity.


Experimental Gerontology | 2004

Mitochondrial and sarcoplasmic proteins, but not myosin heavy chain, are sensitive to leucine supplementation in old rat skeletal muscle

Christelle Guillet; Aude Zangarelli; Anne Mishellany; Paulette Rousset; Claire Sornet; Dominique Dardevet; Yves Boirie

Leucine has a major anabolic impact on muscle protein synthesis in young as in old animals. However, myosin heavy chain (MHC), sarcoplasmic and mitochondrial proteins may differently respond to anabolic factors, especially during aging. To test this hypothesis, fractional synthesis rates (FSR) of the three muscle protein fractions were measured using a flooding dose of [1-(13)C] phenylalanine, in gastrocnemius muscle of adult (8 months) and old (22 months) rats, either in postabsorptive state (PA), or 90-120 min after ingestion of a alanine-supplemented meal (PP+A) or a leucine-supplemented meal (PP+L). In adult and old rats, in comparison with PA, leucine stimulated mitochondrial (adult: 0.260+/-0.011 vs 0.238+/-0.012%h(-1); old: 0.289+/-0.010 vs 0.250+/-0.010%h(-1); PP+L vs PA, P<0.05) and sarcoplasmic (adult: 0.182+/-0.011 vs 0.143+/-0.006%h(-1); old: 0.195+/-0.010 vs 0.149+/-0.008%h(-1); PP+L vs PA, P<0.05) protein FSR, but not MHC synthesis in old rats (0.101+/-0.009 vs 0.137+/-0.018%h(-1); PP+L vs PA, P=NS). In conclusion, synthesis of specific muscle protein is activated by leucine supplementation, but MHC may be less sensitive to anabolic factors with aging.


Obesity Reviews | 2012

Impaired protein metabolism: interlinks between obesity, insulin resistance and inflammation

Christelle Guillet; Aurélie Masgrau; Stéphane Walrand; Yves Boirie

Metabolic and structural changes in skeletal muscle that accompany obesity are often associated with the development of insulin resistance. The first events in the pathogenesis of this disorder are considered as an accumulation of lipids within skeletal muscle due to blunted muscle capacity to oxidize fatty acids. Fat infiltration is also associated with muscle fibre typology modification, decrease in muscle mass and impairments in muscle strength. Thus, as a result of obesity, mobility and quality of life are affected, and this is in part due to quantitative and qualitative impairments in skeletal muscle. In addition, the insulin resistance related to obesity results not only in defective insulin‐stimulated glucose disposal but has also detrimental consequences on protein metabolism at the skeletal muscle level and whole‐body level. This review highlights the involvement of fat accumulation and insulin resistance in metabolic disorders occurring in skeletal muscle during the development of obesity, and the impairments in the regulation of protein metabolism and protein turnover in the links between obesity, metabolic inflammation and insulin resistance.


The Journal of Physiology | 2012

Time-course changes of muscle protein synthesis associated with obesity-induced lipotoxicity

Aurélie Masgrau; Anne Mishellany-Dutour; Hitoshi Murakami; Anne-Marie Beaufrère; Stéphane Walrand; Christophe Giraudet; Carole Migné; Maude Gerbaix; Lore Metz; Daniel Courteix; Christelle Guillet; Yves Boirie

•  Prolonged obesity leads to ectopic lipid accumulation in non‐adipose tissues, particularly in skeletal muscles, inducing metabolic dysfunctions (reduced glucose uptake, mitochondria dysfunction, lipotoxicity). •  Several studies in humans and rodents have shown that obesity induces a short‐term increase in fat‐free mass but a long‐term decrease in skeletal muscle mass. •  We investigated the mechanisms potentially involved in muscle loss by measuring simultaneously protein synthesis and lipid infiltration in different types of skeletal muscles, during the development of obesity. •  Our results show that protein synthesis rate in glycolytic muscles increased together with muscle mass during the early phase of obesity development, whereas it decreased later. Reduced protein synthesis rate was associated with a high lipid accumulation in glycolytic muscles. •  These results suggest that lipid accumulation in muscles during prolonged obesity is deleterious for amino acid incorporation in skeletal muscle proteins, and thus indirectly for muscle mass.


Journal of Nutritional Biochemistry | 2012

TNFα gene knockout differentially affects lipid deposition in liver and skeletal muscle of high-fat-diet mice

Jérôme Salles; Nicolas Tardif; Jean-François Landrier; Isabelle Mothe-Satney; Christelle Guillet; Carole Boue-Vaysse; Lydie Combaret; Christophe Giraudet; Véronique Patrac; Justine Bertrand-Michel; Philippe Denis; Jean-Michel Chardigny; Yves Boirie; Stéphane Walrand

AIMS/HYPOTHESIS Inflammation and ectopic lipid deposition contribute to obesity-related insulin resistance (IR). Studies have shown that deficiency of the proinflammatory cytokine tumor necrosis factor-α (TNFα) protects against the IR induced by a high-fat diet (HFD). We aimed to evaluate the relationship between HFD-related inflammation and lipid deposition in skeletal muscle and liver. EXPERIMENTAL DESIGN Wild-type (WT) and TNFα-deficient (TNFα-KO) mice were subjected to an HFD for 12 weeks. A glucose tolerance test was performed to evaluate IR. Inflammatory status was assessed by measuring plasma and tissue transcript levels of cytokines. Lipid intermediate concentrations were measured in plasma, muscle and liver. The expression of genes involved in fatty acid transport, synthesis and oxidation was analyzed in adipose tissue, muscle and liver. RESULTS HFD induced a higher body weight gain in TNFα-KO mice than in WT mice. The weight of epididymal and abdominal adipose tissues was twofold lower in WT mice than in TNFα-KO mice, whereas liver weight was significantly heavier in WT mice. IR, systemic and adipose tissue inflammation, and plasma nonesterified fatty acid levels were reduced in TNFα-KO mice fed an HFD. TNFα deficiency improved fatty acid metabolism and had a protective effect against lipid deposition, inflammation and fibrosis associated with HFD in liver but had no impact on these markers in muscle. CONCLUSIONS Our data suggest that in an HFD context, TNFα deficiency reduced hepatic lipid accumulation through two mechanisms: an increase in adipose tissue storage capacity and a decrease in fatty acid uptake and synthesis in the liver.

Collaboration


Dive into the Christelle Guillet's collaboration.

Top Co-Authors

Avatar

Yves Boirie

Institut national de la recherche agronomique

View shared research outputs
Top Co-Authors

Avatar

Stéphane Walrand

Institut national de la recherche agronomique

View shared research outputs
Top Co-Authors

Avatar

Christophe Giraudet

Institut national de la recherche agronomique

View shared research outputs
Top Co-Authors

Avatar

Jérôme Salles

Institut national de la recherche agronomique

View shared research outputs
Top Co-Authors

Avatar

Véronique Patrac

Institut national de la recherche agronomique

View shared research outputs
Top Co-Authors

Avatar

Aurélie Masgrau

Institut national de la recherche agronomique

View shared research outputs
Top Co-Authors

Avatar

Paulette Rousset

Institut national de la recherche agronomique

View shared research outputs
Top Co-Authors

Avatar

Nicolas Tardif

Institut national de la recherche agronomique

View shared research outputs
Top Co-Authors

Avatar

Maude Gerbaix

Blaise Pascal University

View shared research outputs
Top Co-Authors

Avatar

Daniel Courteix

Australian Catholic University

View shared research outputs
Researchain Logo
Decentralizing Knowledge