Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Publication year range
1.
Cells ; 12(1)2023 01 02.
Article in English | MEDLINE | ID: mdl-36611976

ABSTRACT

Altered mitochondrial quality and function in muscle may be involved in age-related physical function decline. The role played by the autophagy-lysosome system, a major component of mitochondrial quality control (MQC), is incompletely understood. This study was undertaken to obtain initial indications on the relationship between autophagy, mitophagy, and lysosomal markers in muscle and measures of physical performance and lower extremity tissue composition in young and older adults. Twenty-three participants were enrolled, nine young (mean age: 24.3 ± 4.3 years) and 14 older adults (mean age: 77.9 ± 6.3 years). Lower extremity tissue composition was quantified volumetrically by magnetic resonance imaging and a tissue composition index was calculated as the ratio between muscle and intermuscular adipose tissue volume. Physical performance in older participants was assessed via the Short Physical Performance Battery (SPPB). Protein levels of the autophagy marker p62, the mitophagy mediator BCL2/adenovirus E1B 19 kDa protein-interacting protein 3 (BNIP3), the lysosomal markers transcription factor EB, vacuolar-type ATPase, and lysosomal-associated membrane protein 1 were measured by Western immunoblotting in vastus lateralis muscle biopsies. Older adults had smaller muscle volume and lower tissue composition index than young participants. The protein content of p62 and BNIP3 was higher in older adults. A negative correlation was detected between p62 and BNIP3 and the tissue composition index. p62 and BNIP3 were also related to the performance on the 5-time sit-to-stand test of the SPPB. Our results suggest that an altered expression of markers of the autophagy/mitophagy-lysosomal system is related to deterioration of lower extremity tissue composition and muscle dysfunction. Additional studies are needed to clarify the role of defective MQC in human muscle aging and identify novel biological targets for drug development.


Subject(s)
Mitochondria , Muscle, Skeletal , Humans , Aged , Young Adult , Adult , Aged, 80 and over , Muscle, Skeletal/metabolism , Mitochondria/metabolism , Aging/physiology , Lower Extremity , Physical Functional Performance
2.
Nutrients ; 14(21)2022 Nov 04.
Article in English | MEDLINE | ID: mdl-36364934

ABSTRACT

ß-Hydroxy-ß-methylbutyrate (HMB), a leucine metabolite, can increase skeletal muscle size and function. However, HMB may be less effective at improving muscle function in people with insufficient Vitamin D3 (25-OH-D < 30 ng/mL) which is common in middle-aged and older adults. Therefore, we tested the hypothesis that combining HMB plus Vitamin D3 (HMB + D) supplementation would improve skeletal muscle size, composition, and function in middle-aged women. In a double-blinded fashion, women (53 ± 1 yrs, 26 ± 1 kg/m2, n = 43) were randomized to take placebo or HMB + D (3 g Calcium HMB + 2000 IU D per day) during 12 weeks of sedentary behavior (SED) or resistance exercise training (RET). On average, participants entered the study Vitamin D3 insufficient while HMB + D increased 25-OH-D to sufficient levels after 8 and 12 weeks. In SED, HMB + D prevented the loss of arm lean mass observed with placebo. HMB + D increased muscle volume and decreased intermuscular adipose tissue (IMAT) volume in the thigh compared to placebo but did not change muscle function. In RET, 12-weeks of HMB + D decreased IMAT compared to placebo but did not influence the increase in skeletal muscle volume or function. In summary, HMB + D decreased IMAT independent of exercise status and may prevent the loss or increase muscle size in a small cohort of sedentary middle-aged women. These results lend support to conduct a longer duration study with greater sample size to determine the validity of the observed positive effects of HMB + D on IMAT and skeletal muscle in a small cohort of middle-aged women.


Subject(s)
Cholecalciferol , Muscle Strength , Humans , Middle Aged , Female , Aged , Cholecalciferol/pharmacology , Dietary Supplements , Muscle, Skeletal , Double-Blind Method
3.
Am J Physiol Cell Physiol ; 323(4): C1325-C1332, 2022 Oct 01.
Article in English | MEDLINE | ID: mdl-36094434

ABSTRACT

Chemotherapy is a common therapy to treat patients with breast cancer but also leads to skeletal muscle deconditioning. Skeletal muscle deconditioning is multifactorial and intermuscular adipose tissue (IMAT) accumulation is closely linked to muscle dysfunction. To date, there is no clinical study available investigating IMAT development through a longitudinal protocol and the underlying mechanisms remain unknown. Our study was dedicated to investigating IMAT content in patients with early breast cancer who were treated with chemotherapy and exploring the subsequent cellular mechanisms involved in its development. We included 13 women undergoing chemotherapy. Muscle biopsies and ultrasonography assessment were performed before and after chemotherapy completion. Histological and Western blotting analyses were conducted. We found a substantial increase in protein levels of three mature adipocyte markers (perilipin, +901%; adiponectin, +135%; FABP4, +321%; P < 0.05). These results were supported by an increase in oil red O-positive staining (+358%; P < 0.05). A substantial increase in PDGFRα protein levels was observed (+476%; P < 0.05) highlighting an increase in fibro-adipogenic progenitors (FAPs) content. The cross-sectional area of the vastus lateralis muscle fibers substantially decreased (-21%; P < 0.01), and muscle architecture was altered, as shown by a decrease in fascicle length (-15%; P < 0.05) and a decreasing trend in muscle thickness (-8%; P = 0.08). We demonstrated both IMAT development and muscle atrophy in patients with breast cancer who were treated with chemotherapy. FAPs, critical stem cells inducing both IMAT development and skeletal muscle atrophy, also increased, suggesting that FAPs likely play a critical role in the skeletal muscle deconditioning observed in patients with breast cancer who were treated with chemotherapy.


Subject(s)
Breast Neoplasms , Adiponectin/metabolism , Adipose Tissue/metabolism , Breast Neoplasms/pathology , Female , Humans , Muscle, Skeletal/metabolism , Muscular Atrophy/chemically induced , Muscular Atrophy/diagnostic imaging , Muscular Atrophy/metabolism , Perilipins/metabolism , Receptor, Platelet-Derived Growth Factor alpha/metabolism
4.
Rev Endocr Metab Disord ; 23(1): 71-85, 2022 02.
Article in English | MEDLINE | ID: mdl-34716543

ABSTRACT

In physiological conditions, the adipose organ resides in well-defined areas, where it acts providing an energy supply and as an endocrine organ involved in the control of whole-body energy metabolism. Adipose tissue adipokines connect the body's nutritional status to the regulation of energy balance. When it surrounds organs, it provides also for mechanical protection. Adipose tissue has a complex and heterogenous cellular composition that includes adipocytes, adipose tissue-derived stromal and stem cells (ASCs) which are mesenchymal stromal cells, and endothelial and immune cells, which signal to each other and to other tissues to maintain homeostasis. In obesity and in other nutrition related diseases, as well as in age-related diseases, biological and functional changes of adipose tissue give rise to several complications. Obesity triggers alterations of ASCs, impairing adipose tissue remodeling and adipose tissue function, which induces low-grade systemic inflammation, progressive insulin resistance and other metabolic disorders. Adipose tissue grows by hyperplasia recruiting new ASCs and by hypertrophy, up to its expandability limit. To overcome this limitation and to store the excess of nutrients, adipose tissue develops ectopically, involving organs such as muscle, bone marrow and the heart. The origin of ectopic adipose organ is not clearly elucidated, and a possible explanation lies in the stimulation of the adipogenic differentiation of mesenchymal precursor cells which normally differentiate toward a lineage specific for the organ in which they reside. The chronic exposition of these newly-formed adipose depots to the pathological environment, will confer to them all the phenotypic characteristics of a dysfunctional adipose tissue, perpetuating the organ alterations. Visceral fat, but also ectopic fat, either in the liver, muscle or heart, can increase the risk of developing insulin resistance, type 2 diabetes, and cardiovascular diseases. Being able to prevent and to target dysfunctional adipose tissue will avoid the progression towards the complications of obesity and other nutrition-related diseases. The aim of this review is to summarize some of the knowledge regarding the presence of adipose tissue in particular tissues (where it is not usually present), describing the composition of its adipogenic precursors, and the interactions responsible for the development of organ pathologies.


Subject(s)
Diabetes Mellitus, Type 2 , Adipocytes/metabolism , Adipogenesis , Adipokines/metabolism , Adipose Tissue/metabolism , Diabetes Mellitus, Type 2/metabolism , Humans
5.
Cell Physiol Biochem ; 53(1): 62-75, 2019.
Article in English | MEDLINE | ID: mdl-31184447

ABSTRACT

BACKGROUND/AIMS: Skeletal muscle injuries are the most common type of injury occurring in sports, and investigating skeletal muscle regeneration as well as understanding the related processes is an important aspect of the sports medicine field. The process of regeneration appears to be complex and precisely orchestrated, involving fibro-adipogenic progenitors (FAPs) which are a muscle-resident stem cell population that appears to play a major role in abnormal development of fibrotic tissue or intermuscular adipose tissue (IMAT). Our present study aims to investigate whether muscle resting or endurance exercise following muscle injury may change the behavior of FAPs and subsequently impact the development of fatty infiltrations and fibrosis, two hallmarks of regeneration failure. METHODS: We used the validated glycerol muscle injury model to mimic abnormal muscle regenerative conditions in mice. We challenged this specific regeneration model with hindlimb unloading or endurance exercise and, in a second set of experiments, we treated mice with decorin, a TGF-ß inhibitor. RESULTS: In this study, we demonstrated that: i) muscle resting just after injury leads to inhibition of IMAT development, ii) TNF-α mediated FAP apoptosis might be perturbed in this specific glycerol model of muscle injury, leading to IMAT development, and iii) treatment with the TGF-ß inhibitor decorin decreases IMAT development and might restores FAP apoptosis. CONCLUSION: In addition to the potential clinical relevance of decorin treatment in situations involving muscle plasticity and regeneration, this study also demonstrates that a period of muscle resting is necessary following muscle injury to achieve efficient muscle regeneration which is associated with a reduction in fatty infiltration. Unreasonably early resumption of exercise brings no gain to regeneration, further highlighting that this resting period is necessary.


Subject(s)
Decorin/therapeutic use , Muscle, Skeletal/injuries , Muscular Diseases/drug therapy , Transforming Growth Factor beta/antagonists & inhibitors , Adipose Tissue/drug effects , Adipose Tissue/metabolism , Animals , Apoptosis/drug effects , Decorin/pharmacology , Female , Glycerol/toxicity , Mice , Mice, Inbred C57BL , Muscle, Skeletal/pathology , Muscular Diseases/chemically induced , Muscular Diseases/pathology , Physical Conditioning, Animal , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Stem Cells/cytology , Stem Cells/drug effects , Stem Cells/metabolism , Transforming Growth Factor beta/metabolism
6.
Mol Aspects Med ; 50: 56-87, 2016 08.
Article in English | MEDLINE | ID: mdl-27106402

ABSTRACT

Identification of cost-effective interventions to maintain muscle mass, muscle strength, and physical performance during muscle wasting and aging is an important public health challenge. It requires understanding of the cellular and molecular mechanisms involved. Muscle-deconditioning processes have been deciphered by means of several experimental models, bringing together the opportunities to devise comprehensive analysis of muscle wasting. Studies have increasingly recognized the importance of fatty infiltrations or intermuscular adipose tissue for the age-mediated loss of skeletal-muscle function and emphasized that this new important factor is closely linked to inactivity. The present review aims to address three main points. We first mainly focus on available experimental models involving cell, animal, or human experiments on muscle wasting. We next point out the role of intermuscular adipose tissue in muscle wasting and aging and try to highlight new findings concerning aging and muscle-resident mesenchymal stem cells called fibro/adipogenic progenitors by linking some cellular players implicated in both FAP fate modulation and advancing age. In the last part, we review the main data on the efficiency and molecular and cellular mechanisms by which exercise, replacement hormone therapies, and ß-hydroxy-ß-methylbutyrate prevent muscle wasting and sarcopenia. Finally, we will discuss a potential therapeutic target of sarcopenia: glucose 6-phosphate dehydrogenase.


Subject(s)
Aging/physiology , Muscle, Skeletal/pathology , Muscle, Skeletal/physiopathology , Sarcopenia/pathology , Sarcopenia/physiopathology , Adipose Tissue/metabolism , Adipose Tissue/pathology , Aging/drug effects , Animals , Apoptosis , Dietary Supplements , Exercise , Glucosephosphate Dehydrogenase/metabolism , Hormone Replacement Therapy , Humans , Mitochondria/metabolism , Models, Animal , Models, Theoretical , Sarcopenia/prevention & control , Signal Transduction/drug effects , Valerates/administration & dosage , Weightlessness
7.
Article in Korean | WPRIM (Western Pacific) | ID: wpr-761601

ABSTRACT

BACKGROUND: This study investigated the effects of caloric restriction on thigh intermuscular adipose tissue (IMAT) and the associations of IMAT and metabolic risk factors. METHODS: Thirty-three obese Korean women (BMI 27.2+/-2.5 kg/m2; 32.3+/-8.7 years) were tested before and after 12 weeks of 354.7 kcal/day dietary caloric restriction: waist circumference (WC); %fat according to bioimpedence; subcutaneous adipose tissue (SAT); visceral adipose tissue (VAT); IMAT using single-slice CT scans at the levels of L4/L5 and mid-thigh (midpoint between the anterior iliac crest and patella); fasting levels of leptin, glucose, insulin, triglycerides, total-C, LDL-C, HDL-C, and HOMA-IR. Paired t-test and Spearman correlation analysis were performed. RESULTS: WC, %fat, leptin, glucose, total-C, abdominal SAT and VAT, and mid-thigh IMAT decreased (P<0.05), and %change in mid-thigh IMAT correlated with %change in HOMA-IR (P<0.05). CONCLUSION: Caloric restriction decreased the mid-thigh IMAT in obese Korean women, which may be correlated with reduction of metabolic risk.


Subject(s)
Female , Humans , Adipose Tissue , Caloric Restriction , Fasting , Glucose , Insulin , Intra-Abdominal Fat , Leptin , Obesity , Risk Factors , Subcutaneous Fat , Thigh , Tomography, X-Ray Computed , Triglycerides , Waist Circumference
SELECTION OF CITATIONS
SEARCH DETAIL
...