Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
Add more filters










Publication year range
1.
NPJ Aging ; 10(1): 11, 2024 Feb 03.
Article in English | MEDLINE | ID: mdl-38310117
2.
Biomaterials ; 275: 120982, 2021 08.
Article in English | MEDLINE | ID: mdl-34214785

ABSTRACT

Severe peripheral nerve injuries often result in permanent loss of function of the affected limb. Current treatments are limited by their efficacy in supporting nerve regeneration and behavioral recovery. Here we demonstrate that electrical stimulation through conductive nerve guides (CNGs) enhances the efficacy of human neural progenitor cells (hNPCs) in treating a sciatic nerve transection in rats. Electrical stimulation strengthened the therapeutic potential of NPCs by upregulating gene expression of neurotrophic factors which are critical in augmenting synaptic remodeling, nerve regeneration, and myelination. Electrically-stimulated hNPC-containing CNGs are significantly more effective in improving sensory and motor functions starting at 1-2 weeks after treatment than either treatment alone. Electrophysiology and muscle assessment demonstrated successful re-innervation of the affected target muscles in this group. Furthermore, histological analysis highlighted an increased number of regenerated nerve fibers with thicker myelination in electrically-stimulated hNPC-containing CNGs. The elevated expression of tyrosine kinase receptors (Trk) receptors, known to bind to neurotrophic factors, indicated the long-lasting effect from electrical stimulation on nerve regeneration and distal nerve re-innervation. These data suggest that electrically-enhanced stem cell-based therapy provides a regenerative rehabilitative approach to promote peripheral nerve regeneration and functional recovery.


Subject(s)
Neural Stem Cells , Peripheral Nerve Injuries , Animals , Axons , Electric Stimulation , Humans , Nerve Regeneration , Peripheral Nerve Injuries/therapy , Polymers , Rats , Recovery of Function , Sciatic Nerve
3.
Data Brief ; 31: 105977, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32685634

ABSTRACT

The design of aeroengine real-time control systems needs the implementation of machine learning based techniques. The lack of in-flight aeroengine performance data is a limit for the researchers interested in the development of these prediction algorithms. Dynamic aeroengine models can be used to overcome this lack. This data article presents data regarding the performance of a turbojet that were predicted by the dynamic engine model that was built using the Gas turbine Simulation Program (GSP) software. The data were also used to implement an Artificial Neural Network (ANN) that predicts the in-flight aeroengine performance, such as the Exhaust Gas Temperature (EGT). The Nonlinear AutoRegressive with eXogenous inputs (NARX) neural network was used. The neural network predictions have been also given as dataset of the present article. The data presented here are related to the article entitled "MultiGene Genetic Programming - Artificial Neural Networks approach for dynamic performance prediction of an aeroengine" [1].

4.
Nat Metab ; 2(4): 307-317, 2020 04.
Article in English | MEDLINE | ID: mdl-32601609

ABSTRACT

Aging impairs tissue repair. This is pronounced in skeletal muscle, whose regeneration by muscle stem cells (MuSCs) is robust in young adult animals but inefficient in older organisms. Despite this functional decline, old MuSCs are amenable to rejuvenation through strategies that improve the systemic milieu, such as heterochronic parabiosis. One such strategy, exercise, has long been appreciated for its benefits on healthspan, but its effects on aged stem cell function in the context of tissue regeneration are incompletely understood. Here we show that exercise in the form of voluntary wheel running accelerates muscle repair in old animals and improves old MuSC function. Through transcriptional profiling and genetic studies, we discovered that the restoration of old MuSC activation ability hinges on restoration of Cyclin D1, whose expression declines with age in MuSCs. Pharmacologic studies revealed that Cyclin D1 maintains MuSC activation capacity by repressing TGFß signaling. Taken together, these studies demonstrate that voluntary exercise is a practicable intervention for old MuSC rejuvenation. Furthermore, this work highlights the distinct role of Cyclin D1 in stem cell quiescence.


Subject(s)
Cyclin D1/metabolism , Muscle, Skeletal/cytology , Physical Conditioning, Animal , Stem Cells/cytology , Animals , Cell Separation , Cell Transplantation , Flow Cytometry , Mice , Muscle, Skeletal/metabolism , Stem Cells/metabolism
5.
Nat Commun ; 11(1): 1545, 2020 03 24.
Article in English | MEDLINE | ID: mdl-32210226

ABSTRACT

Aging is characterized by a gradual loss of function occurring at the molecular, cellular, tissue and organismal levels. At the chromatin level, aging associates with progressive accumulation of epigenetic errors that eventually lead to aberrant gene regulation, stem cell exhaustion, senescence, and deregulated cell/tissue homeostasis. Nuclear reprogramming to pluripotency can revert both the age and the identity of any cell to that of an embryonic cell. Recent evidence shows that transient reprogramming can ameliorate age-associated hallmarks and extend lifespan in progeroid mice. However, it is unknown how this form of rejuvenation would apply to naturally aged human cells. Here we show that transient expression of nuclear reprogramming factors, mediated by expression of mRNAs, promotes a rapid and broad amelioration of cellular aging, including resetting of epigenetic clock, reduction of the inflammatory profile in chondrocytes, and restoration of youthful regenerative response to aged, human muscle stem cells, in each case without abolishing cellular identity.


Subject(s)
Cell Nucleus/metabolism , Cellular Reprogramming/physiology , Cellular Senescence/physiology , RNA, Messenger/metabolism , Rejuvenation/physiology , Adolescent , Adult , Aged , Aged, 80 and over , Aging/physiology , Animals , Cells, Cultured , Chondrocytes , DNA Methylation/physiology , Endothelial Cells , Epigenesis, Genetic/physiology , Female , Fibroblasts , Gene Expression Profiling , Humans , Intravital Microscopy , Male , Mice , Middle Aged , Muscle Cells , Primary Cell Culture , Stem Cells , Young Adult
6.
J Physiol ; 598(2): 317-329, 2020 01.
Article in English | MEDLINE | ID: mdl-31784993

ABSTRACT

KEY POINTS: Our tibial fracture orthopaedic injury model in mice recapitulates the major manifestations of complex trauma, including nociceptive sensitization, bone fracture, muscle fibrosis and muscle fibre loss. Delayed exercise after complex orthopaedic trauma results in decreased muscle fibrosis and improved pain Losartan, an angiotensin-receptor blocker with anti-fibrotic abilities, recapitulates the effect of exercise on post-injury recovery and may provide an enhanced recovery option for those who are unable to exercise after injury ABSTRACT: Chronic pain and disability after limb injury are major public health problems. Early mobilization after injury improves functional outcomes for patients, although when and how to implement rehabilitation strategies remains a clinical challenge. Additionally, whether the beneficial effects of exercise can be reproduced using pharmacological tools remains unknown and may benefit patients who are unable to exercise as a result of immobilization. We developed a murine model of orthopaedic trauma combining tibia fracture and pin fixation with muscle damage. Behavioural measures included mechanical nociceptive thresholds and distances run on exercise wheels. Bone healing was quantified using microcomputed tomagraphic scanning, and muscle fibre size distribution and fibrosis were followed using immunohistochemistry. We found that the model provided robust mechanical allodynia, fibrosis and a shift to smaller average muscle fibre size lasting up to 5 weeks from injury. We also observed that allowing 'late' (weeks 1-2) rather than 'early' (weeks 0-1) exercise after injury resulted in greater overall running activity and greater reversal of allodynia. In parallel, the late running paradigm was associated with reduced muscle fibrosis, earlier increase in muscle fibre diameter and a short-term benefit in reducing callus volume. Providing the anti-fibrotic angiotensin receptor blocker losartan to mice in drinking water reduced both allodynia and muscle fibrosis. Combining losartan and late exercise provided no additional benefit. We conclude that early healing after orthopaedic trauma must be allowed prior to the initiation of exercise to achieve optimal pain, functional and physiological outcomes and that losartan is a viable candidate for translational studies.


Subject(s)
Angiotensin Receptor Antagonists/therapeutic use , Fractures, Bone/drug therapy , Losartan/therapeutic use , Muscle, Skeletal/injuries , Regeneration , Animals , Fibrosis , Hyperalgesia/drug therapy , Mice , Motor Activity , Muscle, Skeletal/pathology , Pain , Physical Conditioning, Animal , Receptors, Angiotensin , Tibia/injuries , Time Factors , Wound Healing
7.
Science ; 366(6466): 734-738, 2019 11 08.
Article in English | MEDLINE | ID: mdl-31699935

ABSTRACT

Adult stem cells are essential for tissue homeostasis. In skeletal muscle, muscle stem cells (MuSCs) reside in a quiescent state, but little is known about the mechanisms that control homeostatic turnover. Here we show that, in mice, the variation in MuSC activation rate among different muscles (for example, limb versus diaphragm muscles) is determined by the levels of the transcription factor Pax3. We further show that Pax3 levels are controlled by alternative polyadenylation of its transcript, which is regulated by the small nucleolar RNA U1. Isoforms of the Pax3 messenger RNA that differ in their 3' untranslated regions are differentially susceptible to regulation by microRNA miR206, which results in varying levels of the Pax3 protein in vivo. These findings highlight a previously unrecognized mechanism of the homeostatic regulation of stem cell fate by multiple RNA species.


Subject(s)
Muscle, Skeletal/physiology , Myoblasts, Skeletal/metabolism , PAX3 Transcription Factor/genetics , Polyadenylation , 3' Untranslated Regions , Animals , Gene Knockdown Techniques , Mice , Mice, Mutant Strains , MicroRNAs/metabolism , RNA, Messenger/metabolism , Ribonucleoprotein, U1 Small Nuclear/genetics , Ribonucleoprotein, U1 Small Nuclear/metabolism
8.
Commun Biol ; 2: 170, 2019.
Article in English | MEDLINE | ID: mdl-31098403

ABSTRACT

Traumatic skeletal muscle injuries cause irreversible tissue damage and impaired revascularization. Engineered muscle is promising for enhancing tissue revascularization and regeneration in injured muscle. Here we fabricated engineered skeletal muscle composed of myotubes interspersed with vascular endothelial cells using spatially patterned scaffolds that induce aligned cellular organization, and then assessed their therapeutic benefit for treatment of murine volumetric muscle loss. Murine skeletal myoblasts co-cultured with endothelial cells in aligned nanofibrillar scaffolds form endothelialized and aligned muscle with longer myotubes, more synchronized contractility, and more abundant secretion of angiogenic cytokines, compared to endothelialized engineered muscle formed from randomly-oriented scaffolds. Treatment of traumatically injured muscle with endothelialized and aligned skeletal muscle promotes the formation of highly organized myofibers and microvasculature, along with greater vascular perfusion, compared to treatment of muscle derived from randomly-oriented scaffolds. This work demonstrates the potential of endothelialized and aligned engineered skeletal muscle to promote vascular regeneration following transplantation.


Subject(s)
Muscle, Skeletal/blood supply , Muscle, Skeletal/injuries , Tissue Engineering/methods , Animals , Cell Line , Coculture Techniques , Cytokines/biosynthesis , Endothelial Cells/cytology , Endothelial Cells/physiology , Humans , Mice , Muscle Fibers, Skeletal/physiology , Muscle, Skeletal/physiology , Myoblasts, Skeletal/cytology , Nanofibers/ultrastructure , Regeneration/physiology , Tissue Scaffolds
9.
Nat Commun ; 9(1): 5078, 2018 12 04.
Article in English | MEDLINE | ID: mdl-30510260

ABSTRACT

Royal jelly is the queen-maker for the honey bee Apis mellifera, and has cross-species effects on longevity, fertility, and regeneration in mammals. Despite this knowledge, how royal jelly or its components exert their myriad effects has remained poorly understood. Using mouse embryonic stem cells as a platform, here we report that through its major protein component Royalactin, royal jelly can maintain pluripotency by activating a ground-state pluripotency-like gene network. We further identify Regina, a mammalian structural analog of Royalactin that also induces a naive-like state in mouse embryonic stem cells. This reveals an important innate program for stem cell self-renewal with broad implications in understanding the molecular regulation of stem cell fate across species.


Subject(s)
Fatty Acids/pharmacology , Glycoproteins/pharmacology , Insect Proteins/pharmacology , Mammals/physiology , Mouse Embryonic Stem Cells/drug effects , Pluripotent Stem Cells/drug effects , Animals , Bees/metabolism , Chromatin , Fatty Acids/chemistry , Female , Fertility , Gene Expression Regulation, Developmental/drug effects , Glycoproteins/chemistry , Insect Proteins/chemistry , Lentivirus/genetics , Lentivirus/metabolism , Longevity , Mice , Models, Molecular , Recombinant Proteins , Teratoma/pathology , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
10.
NPJ Regen Med ; 3: 18, 2018.
Article in English | MEDLINE | ID: mdl-30323949

ABSTRACT

Despite the regenerative capacity of muscle, tissue volume is not restored after volumetric muscle loss (VML), perhaps due to a loss-of-structural extracellular matrix. We recently demonstrated the structural and functional restoration of muscle tissue in a mouse model of VML using an engineered "bioconstruct," comprising an extracellular matrix scaffold (decellularized muscle), muscle stem cells (MuSCs), and muscle-resident cells (MRCs). To test the ability of the cell-based bioconstruct to restore whole-muscle biomechanics, we measured biomechanical parameters in uninjured muscles, muscles injured to produce VML lesions, and in muscles that were injured and then treated by implanting either the scaffolds alone or with bioconstructs containing the scaffolds, MuSCs, and MRCs. We measured the active and passive forces over a range of lengths, viscoelastic force relaxation, optimal length, and twitch dynamics. Injured muscles showed a narrowed length-tension curve or lower force over a narrower range of muscle lengths, and increased passive force. When treated with bioconstructs, but not with scaffolds alone, injured muscles showed active and passive length-tension relationships that were not different from uninjured muscles. Moreover, injured muscles treated with bioconstructs exhibited reduced fibrosis compared to injured muscles either untreated or treated with scaffolds alone. The cell-based bioconstruct is a promising treatment approach for future translational efforts to restore whole-muscle biomechanics in muscles with VML lesions.

11.
NPJ Regen Med ; 3: 16, 2018.
Article in English | MEDLINE | ID: mdl-30245849

ABSTRACT

Muscle regeneration can be permanently impaired by traumatic injuries, despite the high regenerative capacity of skeletal muscle. Implantation of engineered biomimetic scaffolds to the site of muscle ablation may serve as an attractive off-the-shelf therapeutic approach. The objective of the study was to histologically assess the therapeutic benefit of a three-dimensional spatially patterned collagen scaffold, in conjunction with rehabilitative exercise, for treatment of volumetric muscle loss. To mimic the physiologic organization of skeletal muscle, which is generally composed of myofibers aligned in parallel, three-dimensional parallel-aligned nanofibrillar collagen scaffolds were fabricated. When implanted into the ablated murine tibialis anterior muscle, the aligned nanofibrillar scaffolds, in conjunction with voluntary caged wheel exercise, significantly improved the density of perfused microvessels, in comparison to treatments of the randomly oriented nanofibrillar scaffold, decellularized scaffold, or in the untreated control group. The abundance of neuromuscular junctions was 19-fold higher when treated with aligned nanofibrillar scaffolds in conjunction with exercise, in comparison to treatment of aligned scaffold without exercise. Although, the density of de novo myofibers was not significantly improved by aligned scaffolds, regardless of exercise activity, the cross-sectional area of regenerating myofibers was increased by > 60% when treated with either aligned and randomly oriented scaffolds, in comparison to treatment of decellularized scaffold or untreated controls. These findings demonstrate that voluntary exercise improved the regenerative effect of aligned scaffolds by augmenting neurovascularization, and have important implications in the design of engineered biomimetic scaffolds for treatment of traumatic muscle injury.

12.
Cell Stem Cell ; 22(2): 177-190.e7, 2018 02 01.
Article in English | MEDLINE | ID: mdl-29395054

ABSTRACT

The development of cell therapy for repairing damaged or diseased skeletal muscle has been hindered by the inability to significantly expand immature, transplantable myogenic stem cells (MuSCs) in culture. To overcome this limitation, a deeper understanding of the mechanisms regulating the transition between activated, proliferating MuSCs and differentiation-primed, poorly engrafting progenitors is needed. Here, we show that methyltransferase Setd7 facilitates such transition by regulating the nuclear accumulation of ß-catenin in proliferating MuSCs. Genetic or pharmacological inhibition of Setd7 promotes in vitro expansion of MuSCs and increases the yield of primary myogenic cell cultures. Upon transplantation, both mouse and human MuSCs expanded with a Setd7 small-molecule inhibitor are better able to repopulate the satellite cell niche, and treated mouse MuSCs show enhanced therapeutic potential in preclinical models of muscular dystrophy. Thus, Setd7 inhibition may help bypass a key obstacle in the translation of cell therapy for muscle disease.


Subject(s)
Muscle Development , Protein Methyltransferases/antagonists & inhibitors , Stem Cell Transplantation , Stem Cells/cytology , Active Transport, Cell Nucleus/drug effects , Animals , Cell Differentiation/drug effects , Cell Line , Cell Lineage/drug effects , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cell Proliferation/drug effects , Cell Self Renewal/drug effects , Cells, Cultured , Gene Deletion , Histone-Lysine N-Methyltransferase , Mice , Muscle, Skeletal/physiology , MyoD Protein/metabolism , Protein Binding/drug effects , Protein Methyltransferases/metabolism , Pyrrolidines/pharmacology , Regeneration/drug effects , Stem Cells/drug effects , Stem Cells/metabolism , Sulfonamides/pharmacology , Tetrahydroisoquinolines/pharmacology , beta Catenin/metabolism
13.
Muscle Nerve ; 57(5): 705-706, 2018 05.
Article in English | MEDLINE | ID: mdl-29365348
14.
Proc Natl Acad Sci U S A ; 114(43): E8996-E9005, 2017 10 24.
Article in English | MEDLINE | ID: mdl-29073096

ABSTRACT

Tissue regeneration depends on the timely activation of adult stem cells. In skeletal muscle, the adult stem cells maintain a quiescent state and proliferate upon injury. We show that muscle stem cells (MuSCs) use direct translational repression to maintain the quiescent state. High-resolution single-molecule and single-cell analyses demonstrate that quiescent MuSCs express high levels of Myogenic Differentiation 1 (MyoD) transcript in vivo, whereas MyoD protein is absent. RNA pulldowns and costainings show that MyoD mRNA interacts with Staufen1, a potent regulator of mRNA localization, translation, and stability. Staufen1 prevents MyoD translation through its interaction with the MyoD 3'-UTR. MuSCs from Staufen1 heterozygous (Staufen1+/-) mice have increased MyoD protein expression, exit quiescence, and begin proliferating. Conversely, blocking MyoD translation maintains the quiescent phenotype. Collectively, our data show that MuSCs express MyoD mRNA and actively repress its translation to remain quiescent yet primed for activation.


Subject(s)
Gene Expression Regulation/physiology , MyoD Protein/metabolism , RNA-Binding Proteins/metabolism , Stem Cells/physiology , Animals , Cell Differentiation , Mice , Muscle Cells/physiology , MyoD Protein/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA-Binding Proteins/genetics
15.
Nat Commun ; 8: 15613, 2017 06 20.
Article in English | MEDLINE | ID: mdl-28631758

ABSTRACT

Volumetric muscle loss (VML) is associated with loss of skeletal muscle function, and current treatments show limited efficacy. Here we show that bioconstructs suffused with genetically-labelled muscle stem cells (MuSCs) and other muscle resident cells (MRCs) are effective to treat VML injuries in mice. Imaging of bioconstructs implanted in damaged muscles indicates MuSCs survival and growth, and ex vivo analyses show force restoration of treated muscles. Histological analysis highlights myofibre formation, neovascularisation, but insufficient innervation. Both innervation and in vivo force production are enhanced when implantation of bioconstructs is followed by an exercise regimen. Significant improvements are also observed when bioconstructs are used to treat chronic VML injury models. Finally, we demonstrate that bioconstructs made with human MuSCs and MRCs can generate functional muscle tissue in our VML model. These data suggest that stem cell-based therapies aimed to engineer tissue in vivo may be effective to treat acute and chronic VML.


Subject(s)
Cell- and Tissue-Based Therapy/methods , Exercise/physiology , Muscle, Skeletal/injuries , Muscle, Skeletal/transplantation , Stem Cell Transplantation/methods , Tissue Engineering/methods , Aged , Animals , Bioreactors , Female , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Middle Aged , Muscle, Skeletal/pathology , Regeneration , Tissue Scaffolds
17.
Nat Biotechnol ; 34(7): 752-9, 2016 07.
Article in English | MEDLINE | ID: mdl-27240197

ABSTRACT

A promising therapeutic strategy for diverse genetic disorders involves transplantation of autologous stem cells that have been genetically corrected ex vivo. A major challenge in such approaches is a loss of stem cell potency during culture. Here we describe an artificial niche for maintaining muscle stem cells (MuSCs) in vitro in a potent, quiescent state. Using a machine learning method, we identified a molecular signature of quiescence and used it to screen for factors that could maintain mouse MuSC quiescence, thus defining a quiescence medium (QM). We also engineered muscle fibers that mimic the native myofiber of the MuSC niche. Mouse MuSCs maintained in QM on engineered fibers showed enhanced potential for engraftment, tissue regeneration and self-renewal after transplantation in mice. An artificial niche adapted to human cells similarly extended the quiescence of human MuSCs in vitro and enhanced their potency in vivo. Our approach for maintaining quiescence may be applicable to stem cells isolated from other tissues.


Subject(s)
Batch Cell Culture Techniques/methods , Muscle Proteins/metabolism , Myoblasts, Skeletal/physiology , Myoblasts, Skeletal/transplantation , Stem Cell Niche/physiology , Tissue Preservation/methods , Animals , Cells, Cultured , Mice , Mice, Inbred C57BL , Myoblasts, Skeletal/cytology , Stem Cell Transplantation/methods , Treatment Outcome
18.
Skelet Muscle ; 5: 10, 2015.
Article in English | MEDLINE | ID: mdl-26075051

ABSTRACT

BACKGROUND: Mutations in the gene encoding ryanodine receptor type-1 (RYR1), the calcium ion (Ca (2+)) release channel in the sarcoplasmic reticulum (SR) of skeletal muscle, are linked to central core disease (CCD) and malignant hyperthermia (MH) susceptibility. We recently reported that mice lacking the skeletal isoform of calsequestrin (CASQ1-null), the primary Ca (2+) buffer in the SR of skeletal muscle and a modulator of RYR1 activity, exhibit lethal heat- and anesthetic-induced hypermetabolic episodes that resemble MH events in humans. METHODS: We compared ultrastructure, oxidative status, and contractile function in skeletal fibers of extensor digitorum longus (EDL) muscles in wild type (WT) and CASQ1-null mice at different ages (from 4 to 27 months) using structural, biochemical, and functional assays. RESULTS: About 25% of fibers in EDL muscles from CASQ1-null mice of 14 to 27 months of age exhibited large areas of structural disarray (named core-like regions), which were rarely observed in muscle from age-matched WT mice. To determine early events that may lead to the formation of cores, we analyzed EDL muscles from adult mice: at 4 to 6 months of age, CASQ1-null mice (compared to WT) displayed significantly reduced grip strength (40 ± 1 vs. 86 ± 1 mN/gr) and exhibited an increase in the percentage of damaged mitochondria (15.1% vs. 2.6%) and a decrease in average cross-sectional fiber area (approximately 37%) in EDL fibers. Finally, oxidative stress was also significantly increased (25% reduction in ratio between reduced and oxidized glutathione, or GSH/GSSG, and 35% increase in production of mitochondrial superoxide flashes). Providing ad libitum access to N-acetylcysteine in the drinking water for 2 months normalized GSH/GSSG ratio, reduced mitochondrial damage (down to 8.9%), and improved grip strength (from 46 ± 3 to 59 ± 2 mN/gr) in CASQ1-null mice. CONCLUSIONS: Our findings: 1) demonstrate that ablation of CASQ1 leads to enhanced oxidative stress, mitochondrial damage, and the formation of structural cores in skeletal muscle; 2) provide new insights in the pathogenic mechanisms that lead to damage/disappearance of mitochondria in cores; and 3) suggest that antioxidants may provide some therapeutic benefit in reducing mitochondrial damage, limiting the development of cores, and improving muscle function.

19.
Cell Res ; 25(7): 761-2, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26113258

ABSTRACT

Muscle stem cells (MuSCs) have long been considered to be potential therapeutic vehicles for diseases of muscle such as muscular dystrophies. A recent study published in Cell Research by Fu et al. reveals that recapitulating in vitro the in vivo microenvironment of MuSCs that occurs during muscle regeneration might be a major step towards translation.


Subject(s)
Inflammation/metabolism , Interferon-gamma/metabolism , Interleukin-13/metabolism , Interleukin-1alpha/metabolism , Satellite Cells, Skeletal Muscle/cytology , Satellite Cells, Skeletal Muscle/metabolism , Tumor Necrosis Factor-alpha/metabolism , Animals , Humans
20.
Diabetes ; 63(8): 2800-11, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24622799

ABSTRACT

Insulin resistance and obesity are associated with a reduction of mitochondrial content in various tissues of mammals. Moreover, a reduced nitric oxide (NO) bioavailability impairs several cellular functions, including mitochondrial biogenesis and insulin-stimulated glucose uptake, two important mechanisms of body adaptation in response to physical exercise. Although these mechanisms have been thoroughly investigated in skeletal muscle and heart, few studies have focused on the effects of exercise on mitochondria and glucose metabolism in adipose tissue. In this study, we compared the in vivo effects of chronic exercise in subcutaneous adipose tissue of wild-type (WT) and endothelial NO synthase (eNOS) knockout (eNOS(-/-)) mice after a swim training period. We then investigated the in vitro effects of NO on mouse 3T3-L1 and human subcutaneous adipose tissue-derived adipocytes after a chronic treatment with an NO donor: diethylenetriamine-NO (DETA-NO). We observed that swim training increases mitochondrial biogenesis, mitochondrial DNA content, and glucose uptake in subcutaneous adipose tissue of WT but not eNOS(-/-) mice. Furthermore, we observed that DETA-NO promotes mitochondrial biogenesis and elongation, glucose uptake, and GLUT4 translocation in cultured murine and human adipocytes. These results point to the crucial role of the eNOS-derived NO in the metabolic adaptation of subcutaneous adipose tissue to exercise training.


Subject(s)
Adipose Tissue/metabolism , Gene Expression Regulation, Enzymologic/physiology , Glucose/metabolism , Mitochondria/metabolism , Nitric Oxide Synthase Type III/metabolism , Physical Conditioning, Animal/physiology , Adipocytes/drug effects , Adipocytes/metabolism , Animals , Cell Line , Humans , Male , Mice , Mice, Knockout , Nitric Oxide/metabolism , Nitric Oxide Synthase Type III/genetics , Norepinephrine , Swimming
SELECTION OF CITATIONS
SEARCH DETAIL
...