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










Publication year range
1.
Cell Rep ; 43(3): 113881, 2024 Mar 26.
Article in English | MEDLINE | ID: mdl-38442019

ABSTRACT

An intriguing effect of short-term caloric restriction (CR) is the expansion of certain stem cell populations, including muscle stem cells (satellite cells), which facilitate an accelerated regenerative program after injury. Here, we utilized the MetRSL274G (MetRS) transgenic mouse to identify liver-secreted plasminogen as a candidate for regulating satellite cell expansion during short-term CR. Knockdown of circulating plasminogen prevents satellite cell expansion during short-term CR. Furthermore, loss of the plasminogen receptor KT (Plg-RKT) is also sufficient to prevent CR-related satellite cell expansion, consistent with direct signaling of plasminogen through the plasminogen receptor Plg-RKT/ERK kinase to promote proliferation of satellite cells. Importantly, we are able to replicate many of these findings in human participants from the CALERIE trial. Our results demonstrate that CR enhances liver protein secretion of plasminogen, which signals directly to the muscle satellite cell through Plg-RKT to promote proliferation and subsequent muscle resilience during CR.


Subject(s)
Plasminogen , Receptors, Cell Surface , Mice , Animals , Humans , Plasminogen/metabolism , Receptors, Cell Surface/metabolism , Caloric Restriction , Liver/metabolism , Mice, Transgenic , Serine Proteases , Cell Proliferation , Muscles/metabolism
2.
Clin Chem ; 70(3): 528-537, 2024 03 02.
Article in English | MEDLINE | ID: mdl-38431279

ABSTRACT

BACKGROUND: Lipid profiling is central for coronary artery disease (CAD) risk assessment. Nonadherence or unreported use of lipid-lowering drugs, particularly statins, can significantly complicate the association between lipid profile measures and CAD clinical outcomes. By combining medication history evaluation with statin analysis in plasma, we determined the effects of inaccurately reported statin use on lipid profile measures and their association with CAD risk. METHODS: We compared medication history of statin use with statin concentration measurements, by liquid chromatography-tandem mass spectrometry, in 690 participants undergoing coronary angiography (63 ± 11 years of age). Nominal logistic regression was employed to model CAD diagnosis with statin measurements, phenotypic, and lipid profile characteristics. RESULTS: Medication history of statin use was confirmed by statin assay for 81% of the patients. Surprisingly, statins were detected in 46% of patients without statin use records. Nonreported statin use was disproportionately higher among older participants. Stratifying samples by statin history resulted in underestimated LDL-lipid measures. Apolipoprotein B concentrations had a significant inverse CAD association, which became nonsignificant upon re-stratification using the statin assay data. CONCLUSIONS: Our study uncovered prominent discrepancies between medication records and actual statin use measured by mass spectrometry. We showed that inaccurate statin use assessments may lead to overestimation and underestimation of LDL levels in statin user and nonuser categories, exaggerating the reverse epidemiology association between LDL levels and CAD diagnosis. Combining medication history and quantitative statin assay data can significantly improve the design, analysis, and interpretation of clinical and epidemiological studies.


Subject(s)
Coronary Artery Disease , Hydroxymethylglutaryl-CoA Reductase Inhibitors , Humans , Coronary Angiography/methods , Coronary Artery Disease/diagnosis , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Lipids , Risk Assessment , Risk Factors , Middle Aged , Aged
4.
Nat Aging ; 3(8): 948-964, 2023 08.
Article in English | MEDLINE | ID: mdl-37500973

ABSTRACT

Heterochronic parabiosis (HPB) is known for its functional rejuvenation effects across several mouse tissues. However, its impact on biological age and long-term health is unknown. Here we performed extended (3-month) HPB, followed by a 2-month detachment period of anastomosed pairs. Old detached mice exhibited improved physiological parameters and lived longer than control isochronic mice. HPB drastically reduced the epigenetic age of blood and liver based on several clock models using two independent platforms. Remarkably, this rejuvenation effect persisted even after 2 months of detachment. Transcriptomic and epigenomic profiles of anastomosed mice showed an intermediate phenotype between old and young, suggesting a global multi-omic rejuvenation effect. In addition, old HPB mice showed gene expression changes opposite to aging but akin to several life span-extending interventions. Altogether, we reveal that long-term HPB results in lasting epigenetic and transcriptome remodeling, culminating in the extension of life span and health span.


Subject(s)
Longevity , Rejuvenation , Mice , Animals , Longevity/genetics , Multiomics , Aging/genetics
5.
Nat Commun ; 13(1): 7613, 2022 Dec 09.
Article in English | MEDLINE | ID: mdl-36494364

ABSTRACT

Pathologies associated with sarcopenia include decline in muscular strength, lean mass and regenerative capacity. Despite the substantial impact on quality of life, no pharmacological therapeutics are available to counteract the age-associated decline in functional capacity and/or, resilience. Evidence suggests immune-secreted cytokines can improve muscle regeneration, a strategy which we leverage in this study by rescuing the age-related deficiency in Meteorin-like through several in vivo add-back models. Notably, the intramuscular, peptide injection of recombinant METRNL was sufficient to improve muscle regeneration in aging. Using ex vivo media exchange and in vivo TNF inhibition, we demonstrate a mechanism of METRNL action during regeneration, showing it counteracts a pro-fibrotic gene program by triggering TNFα-induced apoptosis of fibro/adipogenic progenitor cells. These findings demonstrate therapeutic applications for METRNL to improve aged muscle, and show Fibro/Adipogenic Progenitors are viable therapeutic targets to counteract age-related loss in muscle resilience.


Subject(s)
Muscle, Skeletal , Quality of Life , Muscle, Skeletal/physiology , Adipogenesis , Stem Cells , Cytokines
6.
Exp Gerontol ; 165: 111841, 2022 08.
Article in English | MEDLINE | ID: mdl-35623538

ABSTRACT

Aging is a multifactorial process associated with progressive degradation of physiological integrity and function. One of the greatest factors contributing to the deleterious effects of aging is the decline of functional ability due to loss of muscle mass, strength, and function, a condition termed sarcopenia. Calorie restriction (CR) has consistently been shown to extend lifespan and delay the onset and progression of various age-related diseases, including sarcopenia. Additional anti-aging interventions that are receiving scientific attention are CR mimetics. Of these pharmacological compounds, rapamycin has shown similar CR-related longevity benefits without the need for diet restrictions. To investigate the potential role of rapamycin as an anti-sarcopenic alternative to CR, we conducted a study in male and female C57BL/6 J mice to assess the effects of rapamycin on age-related gene expression changes in skeletal muscle associated with loss of muscle mass, strength, and function, relative to control. We hypothesize that the effects of rapamycin will closely align with CR with respect to physical function and molecular indices associated with muscle quality. Our results indicate CR and rapamycin provide partial protection against age-related decline in muscle, while engaging uniquely different molecular pathways in skeletal muscle. Our preclinical findings of the therapeutic potential of rapamycin or a CR regimen on geroprotective benefits in muscle should be extended to translational studies towards the development of effective strategies for the prevention and management of sarcopenia.


Subject(s)
Caloric Restriction , Sarcopenia , Animals , Female , Male , Mice , Mice, Inbred C57BL , Muscle, Skeletal/physiology , Sirolimus/pharmacology
7.
Mech Ageing Dev ; 195: 111443, 2021 04.
Article in English | MEDLINE | ID: mdl-33529682

ABSTRACT

Caloric restriction (CR) can prolong aged skeletal muscle function, yet the molecular mechanisms are not completely understood. We performed phosphoproteomic analysis on muscle from young and old mice fed an ad libitum diet, and old mice fed a CR diet. CR promoted a youthful phosphoproteomic signature, suppressing several known "pro-aging" pathways including Protein kinase A (PKA). This study validates global signaling changes in skeletal muscle during CR.


Subject(s)
Aging/physiology , Caloric Restriction/methods , Cyclic AMP-Dependent Protein Kinases/metabolism , Muscle, Skeletal , Phosphoproteins/metabolism , Proteomics/methods , Rejuvenation/physiology , Animals , Mice , Mice, Inbred C57BL , Muscle, Skeletal/physiology , Muscle, Skeletal/physiopathology , Principal Component Analysis/methods , Regeneration/physiology , Signal Transduction , Time
8.
J Appl Physiol (1985) ; 130(3): 853-864, 2021 03 01.
Article in English | MEDLINE | ID: mdl-33411638

ABSTRACT

Rheumatoid arthritis (RA) is a systemic inflammatory arthritis impacting primarily joints and cardiac and skeletal muscle. RA's distinct impact on cardiac and skeletal muscle tissue is suggested by studies showing that new RA pharmacologic agents strongly improve joint inflammation, but have little impact on RA-associated mortality, cardiovascular disease, and sarcopenia. Thus, the objective is to understand the distinct effects of RA on cardiac and skeletal muscle, and to therapeutically target these tissues through endurance-based exercise as a way to improve RA mortality and morbidity. We utilize the well-characterized RA mouse model, the K/BxN mouse, to investigate cardiac and skeletal muscle pathologies, including the use of wheel-running exercise to mitigate these pathologies. Strikingly, we found that K/BxN mice, like patients with RA, also exhibit both cardiac and skeletal muscle myopathies that were correlated with circulating IL-6 levels. Three months of wheel-running exercise significantly improved K/BxN joint swelling and reduced systemic IL-6 concentrations. Importantly, there were morphological, gene expression, and functional improvements in both the skeletal muscle and cardiac myopathies with exercise. The K/BxN mouse model of RA recapitulated important RA clinical comorbidities, including altered joint, cardiac and skeletal muscle function. These morphological, molecular, and functional alterations were mitigated with regular exercise, thus suggesting exercise as a potential therapeutic intervention to lessen disease activity in the joint and the peripheral tissues, including the heart and skeletal muscle.NEW & NOTEWORTHY RA, even when controlled, is associated with skeletal muscle weakness and greater risk of cardiovascular disease (CVD). Using exercise as a therapeutic against, the progression of RA is often avoided due to fear of worsening RA pathology. We introduce the K/BxN mouse as an RA model to study both myocardial and skeletal muscle dysfunction. We show that endurance exercise can improve joint, cardiac, and skeletal muscle function in K/BxN mice, suggesting exercise may be beneficial for patients with RA.


Subject(s)
Arthritis, Rheumatoid , Muscle, Skeletal , Animals , Disease Models, Animal , Exercise Therapy , Heart , Humans , Mice
9.
Nat Metab ; 2(3): 278-289, 2020 03.
Article in English | MEDLINE | ID: mdl-32694780

ABSTRACT

The immune system plays a multifunctional role throughout the regenerative process, regulating both pro-/anti-inflammatory phases and progenitor cell function. In the present study, we identify the myokine/cytokine Meteorin-like (Metrnl) as a critical regulator of muscle regeneration. Mice genetically lacking Metrnl have impaired muscle regeneration associated with a reduction in immune cell infiltration and an inability to transition towards an anti-inflammatory phenotype. Isochronic parabiosis, joining wild-type and whole-body Metrnl knock-out (KO) mice, returns Metrnl expression in the injured muscle and improves muscle repair, providing supportive evidence for Metrnl secretion from infiltrating immune cells. Macrophage-specific Metrnl KO mice are also deficient in muscle repair. During muscle regeneration, Metrnl works, in part, through Stat3 activation in macrophages, resulting in differentiation to an anti-inflammatory phenotype. With regard to myogenesis, Metrnl induces macrophage-dependent insulin-like growth factor 1 production, which has a direct effect on primary muscle satellite cell proliferation. Perturbations in this pathway inhibit efficacy of Metrnl in the regenerative process. Together, these studies identify Metrnl as an important regulator of muscle regeneration and a potential therapeutic target to enhance tissue repair.


Subject(s)
Insulin-Like Growth Factor I/metabolism , Muscle, Skeletal/metabolism , Nerve Tissue Proteins/metabolism , STAT3 Transcription Factor/metabolism , Animals , Mice , Mice, Knockout , Nerve Tissue Proteins/genetics
10.
11.
Front Cell Dev Biol ; 7: 183, 2019.
Article in English | MEDLINE | ID: mdl-31555646

ABSTRACT

Our understanding of the molecular basis of aging has greatly increased over the past few decades. In this review, we provide an overview of the key signaling pathways associated with aging, and whose modulation has been shown to extend lifespan in a range of model organisms. We also describe how these pathways converge onto autophagy, a catabolic process that functions to recycle dysfunctional cellular material and maintains energy homeostasis. Finally, we consider various approaches of therapeutically modulating these longevity pathways, highlighting exercise as a potent geroprotector.

12.
Cells ; 8(2)2019 02 20.
Article in English | MEDLINE | ID: mdl-30791569

ABSTRACT

Skeletal muscle has remarkable regenerative capacity, relying on precise coordination between resident muscle stem cells (satellite cells) and the immune system. The age-related decline in skeletal muscle regenerative capacity contributes to the onset of sarcopenia, prolonged hospitalization, and loss of autonomy. Although several age-sensitive pathways have been identified, further investigation is needed to define targets of cellular dysfunction. Autophagy, a process of cellular catabolism, is emerging as a key regulator of muscle regeneration affecting stem cell, immune cell, and myofiber function. Muscle stem cell senescence is associated with a suppression of autophagy during key phases of the regenerative program. Macrophages, a key immune cell involved in muscle repair, also rely on autophagy to aid in tissue repair. This review will focus on the role of autophagy in various aspects of the regenerative program, including adult skeletal muscle stem cells, monocytes/macrophages, and corresponding age-associated dysfunction. Furthermore, we will highlight rejuvenation strategies that alter autophagy to improve muscle regenerative function.


Subject(s)
Autophagy , Muscle, Skeletal/physiology , Regeneration , Aging/physiology , Animals , Caloric Restriction , Humans , Models, Biological
13.
J Biol Chem ; 293(17): 6449-6459, 2018 04 27.
Article in English | MEDLINE | ID: mdl-29530981

ABSTRACT

The insulin-like growth factor 1 receptor (IGF1R) is a receptor tyrosine kinase with critical roles in various biological processes. Recent results from clinical trials targeting IGF1R indicate that IGF1R signaling pathways are more complex than previously thought. Moreover, it has become increasingly clear that the function of many proteins can be understood only in the context of a network of interactions. To that end, we sought to profile IGF1R-protein interactions with the proximity-labeling technique BioID. We applied BioID by generating a HEK293A cell line that stably expressed the BirA* biotin ligase fused to the IGF1R. Following stimulation by IGF1, biotinylated proteins were analyzed by MS. This screen identified both known and previously unknown interactors of IGF1R. One of the novel interactors was sorting nexin 6 (SNX6), a protein that forms part of the retromer complex, which is involved in intracellular protein sorting. Using co-immunoprecipitation, we confirmed that IGF1R and SNX6 physically interact. SNX6 knockdown resulted in a dramatic diminution of IGF1-mediated ERK1/2 phosphorylation, but did not affect IGF1R internalization. Bioluminescence resonance energy transfer experiments indicated that the SNX6 knockdown perturbed the association between IGF1R and the key adaptor proteins insulin receptor substrate 1 (IRS1) and SHC adaptor protein 1 (SHC1). Intriguingly, even in the absence of stimuli, SNX6 overexpression significantly increased Akt phosphorylation. Our study confirms the utility of proximity-labeling methods, such as BioID, to screen for interactors of cell-surface receptors and has uncovered a role of one of these interactors, SNX6, in the IGF1R signaling cascade.


Subject(s)
Insulin-Like Growth Factor I/metabolism , MAP Kinase Signaling System , Receptors, Somatomedin/metabolism , Sorting Nexins/metabolism , Carbon-Nitrogen Ligases/genetics , Carbon-Nitrogen Ligases/metabolism , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Gene Knockdown Techniques , HEK293 Cells , Humans , Insulin Receptor Substrate Proteins/genetics , Insulin Receptor Substrate Proteins/metabolism , Insulin-Like Growth Factor I/genetics , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/metabolism , Phosphorylation/genetics , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Receptor, IGF Type 1 , Repressor Proteins/genetics , Repressor Proteins/metabolism , Sorting Nexins/genetics , Src Homology 2 Domain-Containing, Transforming Protein 1/genetics , Src Homology 2 Domain-Containing, Transforming Protein 1/metabolism , Staining and Labeling
14.
Cell Signal ; 46: 113-119, 2018 06.
Article in English | MEDLINE | ID: mdl-29499305

ABSTRACT

The development of biased agonist drugs is widely recognized to be important for the treatment of many diseases, including cardiovascular disease. While GPCR biased agonism has been heavily characterized there is a distinct lack of information with respect to RTK biased agonism both in the identification of biased agonists as well as their attendant mechanisms. One such RTK, the Insulin-like Growth Factor 1 Receptor (IGF1R) plays an important role in a range of biological and disease processes. The micropeptide LL37 has been described as a biased agonist of the IGF1R. We were interested to further understand the mechanism by which LL37 promotes biased signaling through the IGF1R. We found that LL37 biased agonism is dependent on ß-arrestin 2. Moreover, BRET assays indicated that LL37 biased agonism is explained by the inability of LL37 to promote the recruitment of IRS1 to the IGF1R compared to IGF1. LL37 promotes an altered association of IGF1R with GRK6, which could also serve as an explanation for bias. We also demonstrated a functional consequence of this bias by showing that while LL37 can promote cell proliferation, it does not induce protein synthesis, unlike IGF1, which does both. We have recently identified HASF, a natural protein released by mesenchymal stem cells, as a novel ligand of the IGF1R. HASF is a paracrine factor with potent cardioprotective and cardio-regenerative properties which also acts via IGF1R biased signaling, preferentially activated ERK over Akt.


Subject(s)
Adaptor Proteins, Vesicular Transport/pharmacology , Antimicrobial Cationic Peptides/pharmacology , Heart Ventricles/metabolism , Membrane Proteins/pharmacology , Myoblasts, Cardiac/metabolism , Paracrine Communication/drug effects , Receptor, IGF Type 1/metabolism , beta-Arrestin 2/metabolism , Adaptor Proteins, Vesicular Transport/physiology , Animals , Antimicrobial Cationic Peptides/physiology , Cell Line , Cell Proliferation/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , G-Protein-Coupled Receptor Kinases/metabolism , HEK293 Cells , Heart Ventricles/cytology , Humans , Membrane Proteins/physiology , Mice, Knockout , Myoblasts, Cardiac/cytology , Rats , Signal Transduction/drug effects , Cathelicidins
15.
Biochem J ; 474(5): 771-780, 2017 02 20.
Article in English | MEDLINE | ID: mdl-28096202

ABSTRACT

We have recently shown that hypoxia and Akt-induced stem cell factor (HASF) protects the heart from ischemia-induced damage and promotes cardiomyocyte proliferation. While we have identified certain signaling pathways responsible for these protective effects, the receptor mediating these effects was unknown. Here, we undertook studies to identify the HASF receptor. A yeast two-hybrid screen identified a partial fragment of insulin-like growth factor 1 receptor (IGF1R) as a binding partner of HASF. Subsequent co-immunoprecipitation experiments showed that HASF bound to full-length IGF1R. Binding assays revealed a high affinity of HASF for IGF1R. The treatment of neonatal ventricular cardiomyocytes with HASF resulted in the phosphorylation of IGF1R and other proteins known to be involved in IGF1R-mediated signaling pathways. HASF-mediated ERK activation was abrogated by IGF1R pharmacological inhibitors and siRNAs that targeted IGF1R. However, siRNA-mediated knockdown of either IGF2R or the insulin receptor had no effect on HASF-induced cell signaling. Additionally, pharmacologic inhibition of IGF1R impeded HASF's ability to induce cardiomyocyte proliferation. Finally, we documented that in vivo deletion of the IGF1R completely abolished the ability of HASF to promote cardiomyocyte proliferation in an overexpression mouse model providing further evidence in vivo that the IGF1R is the functional receptor for HASF.


Subject(s)
Adaptor Proteins, Vesicular Transport/genetics , Heart Ventricles/metabolism , Membrane Proteins/genetics , Myocytes, Cardiac/metabolism , Receptor, IGF Type 1/genetics , Adaptor Proteins, Vesicular Transport/antagonists & inhibitors , Adaptor Proteins, Vesicular Transport/metabolism , Animals , Animals, Newborn , Binding Sites , Cell Proliferation/drug effects , Gene Expression Regulation , HEK293 Cells , Heart Ventricles/cytology , Heart Ventricles/drug effects , Humans , Ligands , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/metabolism , Mice , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/metabolism , Myocytes, Cardiac/cytology , Myocytes, Cardiac/drug effects , Phosphorylation , Primary Cell Culture , Protein Binding , Pyrimidines/pharmacology , Pyrroles/pharmacology , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Rats , Rats, Sprague-Dawley , Receptor, IGF Type 1/antagonists & inhibitors , Receptor, IGF Type 1/metabolism , Receptor, IGF Type 2/antagonists & inhibitors , Receptor, IGF Type 2/genetics , Receptor, IGF Type 2/metabolism , Receptor, Insulin/genetics , Receptor, Insulin/metabolism , Signal Transduction , Two-Hybrid System Techniques
16.
Circ Res ; 118(1): 95-107, 2016 Jan 08.
Article in English | MEDLINE | ID: mdl-26837742

ABSTRACT

In the past decade, substantial evidence supports the paradigm that stem cells exert their reparative and regenerative effects, in large part, through the release of biologically active molecules acting in a paracrine fashion on resident cells. The data suggest the existence of a tissue microenvironment where stem cell factors influence cell survival, inflammation, angiogenesis, repair, and regeneration in a temporal and spatial manner.


Subject(s)
Cardiology/trends , Cardiovascular Diseases/therapy , Paracrine Communication/physiology , Regenerative Medicine/trends , Animals , Cardiology/methods , Cardiovascular Diseases/diagnosis , Cell Differentiation/physiology , Humans , Myocytes, Cardiac/physiology , Myocytes, Cardiac/transplantation , Regenerative Medicine/methods , Stem Cell Transplantation/methods , Stem Cell Transplantation/trends
17.
PLoS One ; 9(2): e90398, 2014.
Article in English | MEDLINE | ID: mdl-24587351

ABSTRACT

Satellite cells are the chief contributor to skeletal muscle growth and regeneration. The study of mouse satellite cells has accelerated in recent years due to technical advancements in the isolation of these cells. The study of human satellite cells has lagged and thus little is known about how the biology of mouse and human satellite cells compare. We developed a flow cytometry-based method to prospectively isolate human skeletal muscle progenitors from the satellite cell pool using positive and negative selection markers. Results show that this pool is enriched in PAX7 expressing cells that possess robust myogenic potential including the ability to give rise to de novo muscle in vivo. We compared mouse and human satellite cells in culture and identify differences in the elaboration of the myogenic genetic program and in the sensitivity of the cells to cytokine stimulation. These results indicate that not all mechanisms regulating mouse satellite cell activation are conserved in human satellite cells and that such differences may impact the clinical translation of therapeutics validated in mouse models. Thus, the findings of this study are relevant to developing therapies to combat muscle disease.


Subject(s)
Muscle Development/genetics , Muscle, Skeletal/metabolism , Satellite Cells, Skeletal Muscle/metabolism , Adolescent , Adult , Animals , Biomarkers/metabolism , Female , Flow Cytometry , Gene Expression , Humans , Male , Mice , Middle Aged , Muscle, Skeletal/cytology , Muscle, Skeletal/growth & development , MyoD Protein/genetics , MyoD Protein/metabolism , Myogenic Regulatory Factor 5/genetics , Myogenic Regulatory Factor 5/metabolism , PAX7 Transcription Factor/genetics , PAX7 Transcription Factor/metabolism , Satellite Cells, Skeletal Muscle/cytology , Species Specificity
18.
Skelet Muscle ; 3(1): 2, 2013 Jan 31.
Article in English | MEDLINE | ID: mdl-23369649

ABSTRACT

Satellite cells are rare mononuclear skeletal muscle-resident cells that are the chief contributors to regenerative myogenesis following muscle injury. Although first identified more than 50 years ago, it is only recently that the murine satellite cell has become molecularly defined with the ability to prospectively isolate these cells from their niche. Human satellite cells are considerably less well understood with relatively few studies having been performed on them. In this review, a critical evaluation of this literature is provided along with a discussion of the practical and methodological issues involved with research on human satellite cells. The therapeutic potential of these and other cells types is also discussed, and the various challenges that face satellite cell therapy are addressed.

19.
Exp Physiol ; 96(11): 1101-13, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21804140

ABSTRACT

Duchenne muscular dystrophy is a devastating muscular dystrophy of childhood. Mutations in the dystrophin gene destroy the link between the internal muscle filaments and the extracellular matrix, resulting in severe muscle weakness and progressive muscle wasting. There is currently no cure and, whilst palliative treatment has improved, affected boys are normally confined to a wheelchair by 12 years of age and die from respiratory or cardiac complications in their twenties or thirties. Therapies currently being developed include mutation-specific treatments, DNA- and cell-based therapies, and drugs which aim to modulate cellular pathways or gene expression. This review aims to provide an overview of the different therapeutic approaches aimed at reconstructing the dystrophin-associated protein complex, including restoration of dystrophin expression and upregulation of the functional homologue, utrophin.


Subject(s)
Muscular Dystrophy, Duchenne/therapy , Animals , Codon, Nonsense/drug effects , Dystrophin/genetics , Dystrophin-Associated Protein Complex/genetics , Exons/genetics , Frameshift Mutation , Genetic Therapy , Humans , Male , Mice , Muscle, Skeletal/cytology , Muscle, Skeletal/growth & development , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/physiopathology , Myoblasts/transplantation , Sarcolemma/physiology , Up-Regulation , Utrophin/genetics
20.
J Cell Sci ; 123(Pt 12): 2008-13, 2010 Jun 15.
Article in English | MEDLINE | ID: mdl-20483958

ABSTRACT

Duchenne muscular dystrophy (DMD) is a lethal muscle disease caused by dystrophin deficiency. In normal muscle, dystrophin helps maintain sarcolemmal stability. Dystrophin also recruits neuronal nitric oxide synthase (nNOS) to the sarcolemma. Failure to anchor nNOS to the membrane leads to functional ischemia and aggravates muscle disease in DMD. Over the past two decades, a great variety of therapeutic modalities have been explored to treat DMD. A particularly attractive approach is to increase utrophin expression. Utrophin shares considerable sequence, structural and functional similarity with dystrophin. Here, we test the hypothesis that utrophin also brings nNOS to the sarcolemma. Full-length utrophin cDNA was expressed in dystrophin-deficient mdx mice by gutted adenovirus or via transgenic overexpression. Subcellular nNOS localization was determined by immunofluorescence staining, in situ nNOS activity staining and microsomal preparation western blot. Despite supra-physiological utrophin expression, we did not detect nNOS at the sarcolemma. Furthermore, transgenic utrophin overexpression failed to protect mdx muscle from exercise-associated injury. Our results suggest that full-length utrophin cannot anchor nNOS to the sarcolemma. This finding might have important implications for the development of utrophin-based DMD therapies.


Subject(s)
Dystrophin/metabolism , Muscular Dystrophy, Duchenne/metabolism , Nitric Oxide Synthase Type I/metabolism , Sarcolemma/enzymology , Utrophin/metabolism , Animals , Disease Models, Animal , Dystrophin/genetics , Female , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Muscular Dystrophy, Duchenne/enzymology , Muscular Dystrophy, Duchenne/genetics , Nitric Oxide Synthase Type I/genetics , Protein Binding , Protein Transport , Sarcolemma/genetics , Sarcolemma/metabolism , Utrophin/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...