Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
Add more filters










Publication year range
1.
Stem Cell Reports ; 18(6): 1325-1339, 2023 06 13.
Article in English | MEDLINE | ID: mdl-37315524

ABSTRACT

Skeletal muscle function and regenerative capacity decline during aging, yet factors driving these changes are incompletely understood. Muscle regeneration requires temporally coordinated transcriptional programs to drive myogenic stem cells to activate, proliferate, fuse to form myofibers, and to mature as myonuclei, restoring muscle function after injury. We assessed global changes in myogenic transcription programs distinguishing muscle regeneration in aged mice from young mice by comparing pseudotime trajectories from single-nucleus RNA sequencing of myogenic nuclei. Aging-specific differences in coordinating myogenic transcription programs necessary for restoring muscle function occur following muscle injury, likely contributing to compromised regeneration in aged mice. Differences in pseudotime alignment of myogenic nuclei when comparing aged with young mice via dynamic time warping revealed pseudotemporal differences becoming progressively more severe as regeneration proceeds. Disruptions in timing of myogenic gene expression programs may contribute to incomplete skeletal muscle regeneration and declines in muscle function as organisms age.


Subject(s)
Cell Nucleus , Stem Cells , Animals , Mice , Aging/genetics , Muscle, Skeletal , Gene Expression
2.
Nat Mater ; 22(6): 777-785, 2023 06.
Article in English | MEDLINE | ID: mdl-37217701

ABSTRACT

Hydrogels are extensively used as tunable, biomimetic three-dimensional cell culture matrices, but optically deep, high-resolution images are often difficult to obtain, limiting nanoscale quantification of cell-matrix interactions and outside-in signalling. Here we present photopolymerized hydrogels for expansion microscopy that enable optical clearance and tunable ×4.6-6.7 homogeneous expansion of not only monolayer cell cultures and tissue sections, but cells embedded within hydrogels. The photopolymerized hydrogels for expansion microscopy formulation relies on a rapid photoinitiated thiol/acrylate mixed-mode polymerization that is not inhibited by oxygen and decouples monomer diffusion from polymerization, which is particularly beneficial when expanding cells embedded within hydrogels. Using this technology, we visualize human mesenchymal stem cells and their interactions with nascently deposited proteins at <120 nm resolution when cultured in proteolytically degradable synthetic polyethylene glycol hydrogels. Results support the notion that focal adhesion maturation requires cellular fibronectin deposition; nuclear deformation precedes cellular spreading; and human mesenchymal stem cells display cell-surface metalloproteinases for matrix remodelling.


Subject(s)
Hydrogels , Microscopy , Humans , Hydrogels/pharmacology , Proteins , Cell Culture Techniques/methods , Biocompatible Materials , Polyethylene Glycols
3.
iScience ; 25(6): 104444, 2022 Jun 17.
Article in English | MEDLINE | ID: mdl-35733848

ABSTRACT

Skeletal muscle stem cells, or satellite cells (SCs), are essential to regenerate and maintain muscle. Quiescent SCs reside in an asymmetric niche between the basal lamina and myofiber membrane. To repair muscle, SCs activate, proliferate, and differentiate, fusing to repair myofibers or reacquiring quiescence to replenish the SC niche. Little is known about when SCs reacquire quiescence during regeneration or the cellular processes that direct SC fate decisions. We find that most SCs reacquire quiescence 5-10 days after muscle injury, following differentiation and fusion of most cells to regenerate myofibers. Single-cell sequencing of myogenic cells in regenerating muscle identifies SCs reacquiring quiescence and reveals that noncell autonomous signaling networks influence SC fate decisions during regeneration. SC transplantation experiments confirm that the regenerating environment influences SC fate. We define a window for SC repopulation of the niche, emphasizing the temporal contribution of the regenerative muscle environment on SC fate.

4.
Elife ; 112022 06 13.
Article in English | MEDLINE | ID: mdl-35695839

ABSTRACT

RNA-binding proteins (RBPs), essential for skeletal muscle regeneration, cause muscle degeneration and neuromuscular disease when mutated. Why mutations in these ubiquitously expressed RBPs orchestrate complex tissue regeneration and direct cell fate decisions in skeletal muscle remains poorly understood. Single-cell RNA-sequencing of regenerating Mus musculus skeletal muscle reveals that RBP expression, including the expression of many neuromuscular disease-associated RBPs, is temporally regulated in skeletal muscle stem cells and correlates with specific stages of myogenic differentiation. By combining machine learning with RBP engagement scoring, we discovered that the neuromuscular disease-associated RBP Hnrnpa2b1 is a differentiation-specifying regulator of myogenesis that controls myogenic cell fate transitions during terminal differentiation in mice. The timing of RBP expression specifies cell fate transitions by providing post-transcriptional regulation of messenger RNAs that coordinate stem cell fate decisions during tissue regeneration.


Subject(s)
Muscle Development , Muscle Fibers, Skeletal , Animals , Cell Differentiation , Mice , Muscle Development/genetics , Muscle Fibers, Skeletal/metabolism , Muscle, Skeletal/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism
5.
Biomaterials ; 277: 121097, 2021 10.
Article in English | MEDLINE | ID: mdl-34481290

ABSTRACT

Skeletal muscle tissue is mechanically dynamic with changes in stiffness influencing function, maintenance, and regeneration. We modeled skeletal muscle mechanical changes in culture with dynamically stiffening hydrogels demonstrating that the chaperone protein BAG3 transduces matrix stiffness by redistributing YAP and TAZ subcellular localization in muscle progenitor cells. BAG3 depletion increases cytoplasmic retention of YAP and TAZ, desensitizing myoblasts to changes in hydrogel elastic moduli. Upon differentiation, muscle progenitors depleted of BAG3 formed enlarged, round myotubes lacking the typical cylindrical morphology. The aberrant morphology is dependent on YAP/TAZ signaling, which was sequestered in the cytoplasm in BAG3-depleted myotubes but predominately nuclear in cylindrical myotubes of control cells. Control progenitor cells induced to differentiate on soft (E' = 4 and 12 kPa) hydrogels formed circular myotubes similar to those observed in BAG3-depleted cells. Inhibition of the Hippo pathway partially restored myotube morphologies, permitting nuclear translocation of YAP and TAZ in BAG3-depleted myogenic progenitors. Thus, BAG3 is a critical mediator of dynamic stiffness changes in muscle tissue, coupling mechanical alterations to intracellular signals and inducing changes in gene expression that influence muscle progenitor cell morphology and differentiation.


Subject(s)
Adaptor Proteins, Signal Transducing , Mechanotransduction, Cellular , Adaptor Proteins, Signal Transducing/metabolism , Muscle Fibers, Skeletal/metabolism , Myoblasts/metabolism , Trans-Activators/metabolism , Transcription Factors/metabolism
6.
Sci Adv ; 7(11)2021 03.
Article in English | MEDLINE | ID: mdl-33712460

ABSTRACT

The skeletal muscle microenvironment transiently remodels and stiffens after exercise and injury, as muscle ages, and in myopathic muscle; however, how these changes in stiffness affect resident muscle stem cells (MuSCs) remains understudied. Following muscle injury, muscle stiffness remained elevated after morphological regeneration was complete, accompanied by activated and proliferative MuSCs. To isolate the role of stiffness on MuSC behavior and determine the underlying mechanotransduction pathways, we cultured MuSCs on strain-promoted azide-alkyne cycloaddition hydrogels capable of in situ stiffening by secondary photocrosslinking of excess cyclooctynes. Using pre- to post-injury stiffness hydrogels, we found that elevated stiffness enhances migration and MuSC proliferation by localizing yes-associated protein 1 (YAP) and WW domain-containing transcription regulator 1 (WWTR1; TAZ) to the nucleus. Ablating YAP and TAZ in vivo promotes MuSC quiescence in postinjury muscle and prevents myofiber hypertrophy, demonstrating that persistent exposure to elevated stiffness activates mechanotransduction signaling maintaining activated and proliferating MuSCs.

7.
Trends Mol Med ; 27(5): 469-481, 2021 05.
Article in English | MEDLINE | ID: mdl-33384234

ABSTRACT

Skeletal muscle formation is a complex process that requires tight spatiotemporal control of key myogenic factors. Emerging evidence suggests that RNA processing is crucial for the regulation of these factors, and that multiple post-transcriptional regulatory pathways work dependently and independently of one another to enable precise control of transcripts throughout muscle development and repair. Moreover, disruption of these pathways is implicated in neuromuscular disease, and the recent development of RNA-mediated therapies shows enormous promise in the treatment of these disorders. We discuss the overlapping post-transcriptional regulatory pathways that mediate muscle development, how these pathways are disrupted in neuromuscular disorders, and advances in RNA-mediated therapies that present a novel approach to the treatment of these diseases.


Subject(s)
Muscle Development/physiology , Muscular Diseases , Neuromuscular Diseases , RNA Processing, Post-Transcriptional , Alternative Splicing , Animals , Humans , MicroRNAs , Muscle, Skeletal/embryology , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscular Diseases/etiology , Muscular Diseases/metabolism , Muscular Diseases/prevention & control , Neuromuscular Diseases/etiology , Neuromuscular Diseases/metabolism , Neuromuscular Diseases/prevention & control , Polyadenylation , RNA/metabolism
8.
Muscle Nerve ; 61(6): 740-744, 2020 06.
Article in English | MEDLINE | ID: mdl-32108365

ABSTRACT

Limb contractures are debilitating complications associated with various muscle and nervous system disorders. This report summarizes presentations at a conference at the Shirley Ryan AbilityLab in Chicago, Illinois, on April 19-20, 2018, involving researchers and physicians from diverse disciplines who convened to discuss current clinical and preclinical understanding of contractures in Duchenne muscular dystrophy, stroke, cerebral palsy, and other conditions. Presenters described changes in muscle architecture, activation, extracellular matrix, satellite cells, and muscle fiber sarcomeric structure that accompany or predispose muscles to contracture. Participants identified ongoing and future research directions that may lead to understanding of the intersecting factors that trigger contractures. These include additional studies of changes in muscle, tendon, joint, and neuronal tissues during contracture development with imaging, molecular, and physiologic approaches. Participants identified the requirement for improved biomarkers and outcome measures to identify patients likely to develop contractures and to accurately measure efficacy of treatments currently available and under development.


Subject(s)
Contracture/physiopathology , Education/trends , Musculoskeletal Diseases/physiopathology , Nervous System Diseases/physiopathology , Research Report/trends , Cerebral Palsy/diagnosis , Cerebral Palsy/physiopathology , Cerebral Palsy/therapy , Chicago , Contracture/diagnosis , Contracture/therapy , Humans , Muscular Dystrophy, Duchenne/diagnosis , Muscular Dystrophy, Duchenne/physiopathology , Muscular Dystrophy, Duchenne/therapy , Musculoskeletal Diseases/diagnosis , Musculoskeletal Diseases/therapy , Nervous System Diseases/diagnosis , Nervous System Diseases/therapy
9.
Nature ; 563(7732): 508-513, 2018 11.
Article in English | MEDLINE | ID: mdl-30464263

ABSTRACT

A dominant histopathological feature in neuromuscular diseases, including amyotrophic lateral sclerosis and inclusion body myopathy, is cytoplasmic aggregation of the RNA-binding protein TDP-43. Although rare mutations in TARDBP-the gene that encodes TDP-43-that lead to protein misfolding often cause protein aggregation, most patients do not have any mutations in TARDBP. Therefore, aggregates of wild-type TDP-43 arise in most patients by an unknown mechanism. Here we show that TDP-43 is an essential protein for normal skeletal muscle formation that unexpectedly forms cytoplasmic, amyloid-like oligomeric assemblies, which we call myo-granules, during regeneration of skeletal muscle in mice and humans. Myo-granules bind to mRNAs that encode sarcomeric proteins and are cleared as myofibres mature. Although myo-granules occur during normal skeletal-muscle regeneration, myo-granules can seed TDP-43 amyloid fibrils in vitro and are increased in a mouse model of inclusion body myopathy. Therefore, increased assembly or decreased clearance of functionally normal myo-granules could be the source of cytoplasmic TDP-43 aggregates that commonly occur in neuromuscular disease.


Subject(s)
Amyloid/metabolism , DNA-Binding Proteins/metabolism , Muscle, Skeletal/physiology , RNA, Messenger/metabolism , Regeneration , TDP-43 Proteinopathies/metabolism , Amyloid/chemistry , Amyloid/genetics , Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/pathology , Animals , Cytoplasm/metabolism , DNA-Binding Proteins/chemistry , Female , Humans , Male , Mice , Models, Biological , Muscle Fibers, Skeletal/metabolism , Muscle, Skeletal/metabolism , RNA, Messenger/genetics , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/metabolism , Sarcomeres/metabolism , TDP-43 Proteinopathies/pathology
10.
Cell Stem Cell ; 23(1): 1-2, 2018 07 05.
Article in English | MEDLINE | ID: mdl-29979984

ABSTRACT

In this issue of Cell Stem Cell, Chan et al. (2018) report that in vivo differentiation of pluripotent stem cells in induced teratomas produces functional embryonic-like muscle stem cells. These purified muscle stem cells engraft with high efficiency and regenerate serially injured muscle.


Subject(s)
Pluripotent Stem Cells , Teratoma , Cell Differentiation , Humans , Muscle Fibers, Skeletal , Myoblasts
11.
Sci Rep ; 8(1): 4309, 2018 03 09.
Article in English | MEDLINE | ID: mdl-29523805

ABSTRACT

Down syndrome, caused by trisomy 21, is characterized by a variety of medical conditions including intellectual impairments, cardiovascular defects, blood cell disorders and pre-mature aging phenotypes. Several somatic stem cell populations are dysfunctional in Down syndrome and their deficiencies may contribute to multiple Down syndrome phenotypes. Down syndrome is associated with muscle weakness but skeletal muscle stem cells or satellite cells in Down syndrome have not been investigated. We find that a failure in satellite cell expansion impairs muscle regeneration in the Ts65Dn mouse model of Down syndrome. Ts65Dn satellite cells accumulate DNA damage and over express Usp16, a histone de-ubiquitinating enzyme that regulates the DNA damage response. Impairment of satellite cell function, which further declines as Ts65Dn mice age, underscores stem cell deficiencies as an important contributor to Down syndrome pathologies.


Subject(s)
Down Syndrome/pathology , Muscle Fibers, Skeletal/physiology , Regeneration , Satellite Cells, Skeletal Muscle/physiology , Animals , Cells, Cultured , DNA Damage , Down Syndrome/metabolism , Mice , Mice, Inbred C57BL , Muscle Fibers, Skeletal/metabolism , Satellite Cells, Skeletal Muscle/metabolism , Ubiquitin Thiolesterase/metabolism
12.
PLoS One ; 13(1): e0190963, 2018.
Article in English | MEDLINE | ID: mdl-29304082

ABSTRACT

Excessive circulating triglycerides due to reduction or loss of lipoprotein lipase activity contribute to hypertriglyceridemia and increased risk for pancreatitis. The only gene therapy treatment for lipoprotein lipase deficiency decreases pancreatitis but minimally reduces hypertriglyceridemia. Synthesized in multiple tissues including striated muscle and adipose tissue, lipoprotein lipase is trafficked to blood vessel endothelial cells where it is anchored at the plasma membrane and hydrolyzes triglycerides into free fatty acids. We conditionally knocked out lipoprotein lipase in differentiated striated muscle tissue lowering striated muscle lipoprotein lipase activity causing hypertriglyceridemia. We then crossed lipoprotein lipase striated muscle knockout mice with mice possessing a conditional avian retroviral receptor gene and injected mice with either a human lipoprotein lipase retrovirus or an mCherry control retrovirus. Post-heparin plasma lipoprotein lipase activity increased for three weeks following human lipoprotein lipase retroviral infection compared to mCherry infected mice. Human lipoprotein lipase infected mice had significantly lower blood triglycerides compared to mCherry controls and were comparable to wild-type blood triglyceride levels. Thus, targeted delivery of human lipoprotein lipase into striated muscle tissue identifies a potential therapeutic target for lipoprotein lipase deficiency.


Subject(s)
Genetic Therapy , Lipoprotein Lipase/genetics , Muscle, Striated/pathology , Animals , Genetic Vectors , Humans , Hypertriglyceridemia/etiology , Mice , Mice, Knockout , Muscle, Striated/enzymology , Retroviridae/genetics
13.
Dev Dyn ; 246(5): 359-367, 2017 05.
Article in English | MEDLINE | ID: mdl-28249356

ABSTRACT

Fibroblast growth factors (FGFs) are essential for self-renewal of skeletal muscle stem cells (satellite cells) and required for maintenance and repair of skeletal muscle. Satellite cells express high levels of FGF receptors 1 and 4, low levels of FGF receptor 3, and little or no detectable FGF receptor 2. Of the multiple FGFs that influence satellite cell function in culture, FGF2 and FGF6 are the only members that regulate satellite cell function in vivo by activating ERK MAPK, p38α/ß MAPKs, PI3 kinase, PLCγ and STATs. Regulation of FGF signaling is complex in satellite cells, requiring Syndecan-4, a heparan sulfate proteoglycan, as well as ß1-integrin and fibronectin. During aging, reduced responsiveness to FGF diminishes satellite cell self-renewal, leading to impaired skeletal muscle regeneration and depletion of satellite cells. Mislocalization of ß1-integrin, reductions in fibronectin, and alterations in heparan sulfate content all contribute to reduced FGF responsiveness in satellite cells. How these cell surface proteins regulate satellite cell self-renewal is incompletely understood. Here we summarize the current knowledge, highlighting the role(s) for FGF signaling in skeletal muscle regeneration, satellite cell behavior, and age-induced muscle wasting. Developmental Dynamics 246:359-367, 2017. © 2017 Wiley Periodicals, Inc.


Subject(s)
Fibroblast Growth Factors/metabolism , Muscle, Skeletal/cytology , Satellite Cells, Skeletal Muscle/physiology , Stem Cells , Aging , Animals , Cell Self Renewal , Fibroblast Growth Factors/physiology , Humans , Signal Transduction
14.
Methods Mol Biol ; 1556: 237-244, 2017.
Article in English | MEDLINE | ID: mdl-28247353

ABSTRACT

Transplanting adult stem cells provides a stringent test for self-renewal and the assessment of comparative engraftment in competitive transplant assays. Transplantation of satellite cells into mammalian skeletal muscle provided the first critical evidence that satellite cells function as adult muscle stem cells. Transplantation of a single satellite cell confirmed and extended this hypothesis, providing proof that the satellite cell is a bona fide adult skeletal muscle stem cell as reported by Sacco et al. (Nature 456(7221):502-506). Satellite cell transplantation has been further leveraged to identify culture conditions that maintain engraftment and to identify self-renewal deficits in satellite cells from aged mice. Conversion of iPSCs (induced pluripotent stem cells) to a satellite cell-like state, followed by transplantation, demonstrated that these cells possess adult muscle stem cell properties as reported by Darabi et al. (Stem Cell Rev Rep 7(4):948-957) and Mizuno et al. (FASEB J 24(7):2245-2253). Thus, transplantation strategies involving either satellite cells derived from adult muscles or derived from iPSCs may eventually be exploited as a therapy for treating patients with diseased or failing skeletal muscle. Here, we describe methods for isolating dispersed adult mouse satellite cells and satellite cells on intact myofibers for transplantation into recipient mice to study muscle stem cell function and behavior following engraftment .


Subject(s)
Muscle, Skeletal/cytology , Stem Cell Transplantation , Stem Cells/cytology , Adult Stem Cells/cytology , Adult Stem Cells/metabolism , Biomarkers , Cell Separation/methods , Flow Cytometry/methods , Muscle Fibers, Skeletal/cytology , Muscle Fibers, Skeletal/transplantation , Regeneration , Satellite Cells, Skeletal Muscle/cytology , Stem Cell Transplantation/methods , Stem Cells/metabolism
15.
Skelet Muscle ; 6: 34, 2016.
Article in English | MEDLINE | ID: mdl-27757223

ABSTRACT

BACKGROUND: The skeletal muscle stem cell niche provides an environment that maintains quiescent satellite cells, required for skeletal muscle homeostasis and regeneration. Syndecan-3, a transmembrane proteoglycan expressed in satellite cells, supports communication with the niche, providing cell interactions and signals to maintain quiescent satellite cells. RESULTS: Syndecan-3 ablation unexpectedly improves regeneration in repeatedly injured muscle and in dystrophic mice, accompanied by the persistence of sublaminar and interstitial, proliferating myoblasts. Additionally, muscle aging is improved in syndecan-3 null mice. Since syndecan-3 null myofiber-associated satellite cells downregulate Pax7 and migrate away from the niche more readily than wild type cells, syxndecan-3 appears to regulate satellite cell homeostasis and satellite cell homing to the niche. CONCLUSIONS: Manipulating syndecan-3 provides a promising target for development of therapies to enhance muscle regeneration in muscular dystrophies and in aged muscle.


Subject(s)
Homeostasis , Muscle, Skeletal/physiology , Regeneration , Satellite Cells, Skeletal Muscle/physiology , Stem Cell Niche , Syndecan-3/physiology , Animals , Female , Male , Mice , Mice, Knockout , Muscle, Skeletal/injuries , Muscle, Skeletal/pathology , Satellite Cells, Skeletal Muscle/pathology , Syndecan-3/genetics
16.
Methods Mol Biol ; 1460: 141-62, 2016.
Article in English | MEDLINE | ID: mdl-27492171

ABSTRACT

Adult skeletal muscle stem cells, termed satellite cells, regenerate and repair the functional contractile cells in adult skeletal muscle called myofibers. Satellite cells reside in a niche between the basal lamina and sarcolemma of myofibers. Isolating single myofibers and their associated satellite cells provides a culture system that partially mimics the in vivo environment. We describe methods for isolating and culturing intact individual myofibers and their associated satellite cells from the mouse extensor digitorum longus muscle. Following dissection and isolation of individual myofibers we provide protocols for myofiber transplantation, satellite cell transfection, immune detection of satellite cell antigens, and assays to examine satellite cell self-renewal and proliferation.


Subject(s)
Cell Culture Techniques , Cell Separation/methods , Fluorescent Antibody Technique , Muscle, Skeletal/cytology , Satellite Cells, Skeletal Muscle/cytology , Satellite Cells, Skeletal Muscle/metabolism , Animals , Cell Differentiation , Cell Proliferation , Mice , Satellite Cells, Skeletal Muscle/transplantation , Transfection
17.
Cell Rep ; 16(5): 1379-1390, 2016 08 02.
Article in English | MEDLINE | ID: mdl-27452471

ABSTRACT

Following skeletal muscle injury, muscle stem cells (satellite cells) are activated, proliferate, and differentiate to form myofibers. We show that mRNA-decay protein AUF1 regulates satellite cell function through targeted degradation of specific mRNAs containing 3' AU-rich elements (AREs). auf1(-/-) mice undergo accelerated skeletal muscle wasting with age and impaired skeletal muscle repair following injury. Satellite cell mRNA analysis and regeneration studies demonstrate that auf1(-/-) satellite cell self-renewal is impaired due to increased stability and overexpression of ARE-mRNAs, including cell-autonomous overexpression of matrix metalloprotease MMP9. Secreted MMP9 degrades the skeletal muscle matrix, preventing satellite-cell-mediated regeneration and return to quiescence. Blocking MMP9 activity in auf1(-/-) mice restores skeletal muscle repair and maintenance of the satellite cell population. Control of ARE-mRNA decay by AUF1 represents a mechanism for adult stem cell regulation and is implicated in human skeletal muscle wasting diseases.


Subject(s)
Adult Stem Cells/metabolism , Heterogeneous-Nuclear Ribonucleoprotein D/metabolism , Muscle, Skeletal/metabolism , Myoblasts/metabolism , RNA Stability/physiology , RNA, Messenger/metabolism , RNA-Binding Proteins/metabolism , 3' Untranslated Regions/genetics , Animals , Female , Heterogeneous Nuclear Ribonucleoprotein D0 , Male , Matrix Metalloproteinase 9/metabolism , Mice , Regeneration/physiology
18.
Circ Res ; 118(7): 1143-50; discussion 1150, 2016 Apr 01.
Article in English | MEDLINE | ID: mdl-27034276

ABSTRACT

This "Controversies in Cardiovascular Research" article evaluates the evidence for and against the hypothesis that the circulating blood level of growth differentiation factor 11 (GDF11) decreases in old age and that restoring normal GDF11 levels in old animals rejuvenates their skeletal muscle and reverses pathological cardiac hypertrophy and cardiac dysfunction. Studies supporting the original GDF11 hypothesis in skeletal and cardiac muscle have not been validated by several independent groups. These new studies have either found no effects of restoring normal GDF11 levels on cardiac structure and function or have shown that increasing GDF11 or its closely related family member growth differentiation factor 8 actually impairs skeletal muscle repair in old animals. One possible explanation for what seems to be mutually exclusive findings is that the original reagent used to measure GDF11 levels also detected many other molecules so that age-dependent changes in GDF11 are still not well known. The more important issue is whether increasing blood [GDF11] repairs old skeletal muscle and reverses age-related cardiac pathologies. There are substantial new and existing data showing that GDF8/11 can exacerbate rather than rejuvenate skeletal muscle injury in old animals. There is also new evidence disputing the idea that there is pathological hypertrophy in old C57bl6 mice and that GDF11 therapy can reverse cardiac pathologies. Finally, high [GDF11] causes reductions in body and heart weight in both young and old animals, suggestive of a cachexia effect. Our conclusion is that elevating blood levels of GDF11 in the aged might cause more harm than good.


Subject(s)
Aging/pathology , Bone Morphogenetic Proteins/therapeutic use , Growth Differentiation Factors/therapeutic use , Muscular Diseases/drug therapy , Aging/blood , Animals , Bone Morphogenetic Proteins/blood , Bone Morphogenetic Proteins/deficiency , Bone Morphogenetic Proteins/pharmacology , Bone Morphogenetic Proteins/toxicity , Cachexia/chemically induced , Cells, Cultured , Drug Evaluation, Preclinical , Growth Differentiation Factors/blood , Growth Differentiation Factors/deficiency , Growth Differentiation Factors/pharmacology , Growth Differentiation Factors/toxicity , Heart/drug effects , Humans , Hypertrophy , Mice, Inbred C57BL , Models, Animal , Muscle, Skeletal/injuries , Muscle, Skeletal/physiology , Muscles/pathology , Muscular Diseases/physiopathology , Myocardium/pathology , Myostatin/physiology , Myostatin/therapeutic use , Myostatin/toxicity , Parabiosis , Recombinant Proteins/therapeutic use , Recombinant Proteins/toxicity , Regeneration/drug effects , Reproducibility of Results , Signal Transduction , Single-Blind Method , Smad2 Protein/physiology , Smad3 Protein/physiology
19.
Skelet Muscle ; 5: 42, 2015.
Article in English | MEDLINE | ID: mdl-26668715

ABSTRACT

BACKGROUND: Adult skeletal muscle adapts to functional needs, maintaining consistent numbers of myonuclei and stem cells. Although resident muscle stem cells or satellite cells are required for muscle growth and repair, in uninjured muscle, these cells appear quiescent and metabolically inactive. To investigate the satellite cell contribution to myofibers in adult uninjured skeletal muscle, we labeled satellite cells by inducing a recombination of LSL-tdTomato in Pax7(CreER) mice and scoring tdTomato+ myofibers as an indicator of satellite cell fusion. RESULTS: Satellite cell fusion into myofibers plateaus postnatally between 8 and 12 weeks of age, reaching a steady state in hindlimb muscles, but in extra ocular or diaphragm muscles, satellite cell fusion is maintained at postnatal levels irrespective of the age assayed. Upon recombination and following a 2-week chase in 6-month-old mice, tdTomato-labeled satellite cells fused into myofibers as 20, 50, and 80 % of hindlimb, extra ocular, and diaphragm myofibers, respectively, were tdTomato+. Satellite cells contribute to uninjured myofibers either following a cell division or directly without an intervening cell division. CONCLUSIONS: The frequency of satellite cell fusion into the skeletal muscle fibers is greater than previously estimated, suggesting an important functional role for satellite cell fusion into adult myofibers and a requirement for active maintenance of satellite cell numbers in uninjured skeletal muscle.

20.
Stem Cells ; 33(10): 3138-51, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26304770

ABSTRACT

The transcription factor Pax7 regulates skeletal muscle stem cell (satellite cells) specification and maintenance through various mechanisms, including repressing the activity of the muscle regulatory factor MyoD. Hence, Pax7-to-MyoD protein ratios can determine maintenance of the committed-undifferentiated state or activation of the differentiation program. Pax7 expression decreases sharply in differentiating myoblasts but is maintained in cells (re)acquiring quiescence, yet the mechanisms regulating Pax7 levels based on differentiation status are not well understood. Here we show that Pax7 levels are directly regulated by the ubiquitin-ligase Nedd4. Our results indicate that Nedd4 is expressed in quiescent and activated satellite cells, that Nedd4 and Pax7 physically interact during early muscle differentiation-correlating with Pax7 ubiquitination and decline-and that Nedd4 loss of function prevented this effect. Furthermore, even transient nuclear accumulation of Nedd4 induced a drop in Pax7 levels and precocious muscle differentiation. Consequently, we propose that Nedd4 functions as a novel Pax7 regulator, which activity is temporally and spatially controlled to modulate the Pax7 protein levels and therefore satellite cell fate.


Subject(s)
Cell Differentiation/genetics , Endosomal Sorting Complexes Required for Transport/biosynthesis , Muscle Development , PAX7 Transcription Factor/biosynthesis , Satellite Cells, Skeletal Muscle/metabolism , Ubiquitin-Protein Ligases/biosynthesis , Animals , Cell Proliferation/genetics , Endosomal Sorting Complexes Required for Transport/genetics , Gene Expression Regulation, Developmental , Humans , Mice , Muscle, Skeletal/growth & development , Muscle, Skeletal/metabolism , MyoD Protein/biosynthesis , Nedd4 Ubiquitin Protein Ligases , PAX7 Transcription Factor/genetics , Proteasome Endopeptidase Complex/genetics , Satellite Cells, Skeletal Muscle/cytology , Ubiquitin-Protein Ligases/genetics , Ubiquitination
SELECTION OF CITATIONS
SEARCH DETAIL
...