Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
1.
Metabolism ; 103: 154025, 2020 02.
Article in English | MEDLINE | ID: mdl-31765667

ABSTRACT

BACKGROUND: Metabolic complications are highly prevalent in cancer survivors treated with irradiation but the underlying mechanisms remain unknown. METHODS: Chow or high fat-fed C57Bl/6J mice were irradiated (6Gy) before investigating the impact on whole-body or skeletal muscle metabolism and profiling their lipidomic signature. Using a transgenic mouse model (Tg:Pax7-nGFP), we isolated muscle progenitor cells (satellite cells) and characterised their metabolic functions. We recruited childhood cancer survivors, grouped them based on the use of total body irradiation during their treatment and established their lipidomic profile. RESULTS: In mice, irradiation delayed body weight gain and impaired fat pads and muscle weights. These changes were associated with impaired whole-body fat oxidation in chow-fed mice and altered ex vivo skeletal muscle fatty acid oxidation, potentially due to a reduction in oxidative fibres and reduced mitochondrial enzyme activity. Irradiation led to fasting hyperglycaemia and impaired glucose uptake in isolated skeletal muscles. Cultured satellite cells from irradiated mice showed decreased fatty acid oxidation and reduced glucose uptake, recapitulating the host metabolic phenotype. Irradiation resulted in a remodelling of lipid species in skeletal muscles, with the extensor digitorum longus muscle being particularly affected. A large number of lipid species were reduced, with several of these species showing a positive correlation with mitochondrial enzymes activity. In cancer survivors exposed to irradiation, we found a similar decrease in systemic levels of most lipid species, and lipid species that increased were positively correlated with insulin resistance (HOMA-IR). CONCLUSION: Irradiation leads to long-term alterations in body composition, and lipid and carbohydrate metabolism in skeletal muscle, and affects muscle progenitor cells. Such changes result in persistent impairment of metabolic functions, providing a new mechanism for the increased prevalence of metabolic diseases reported in irradiated individuals. In this context, changes in the lipidomic signature in response to irradiation could be of diagnostic value.


Subject(s)
Cancer Survivors , Metabolic Diseases/etiology , Mitochondria/radiation effects , Muscle, Skeletal/radiation effects , Neoplasms/radiotherapy , Whole-Body Irradiation/adverse effects , Adolescent , Adult , Animals , Child , Child, Preschool , Energy Metabolism/radiation effects , Female , Follow-Up Studies , Humans , Male , Metabolic Diseases/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mitochondria/physiology , Muscle, Skeletal/metabolism , Neoplasms/metabolism , Oxidation-Reduction/radiation effects , Radiation Injuries/metabolism , Radiation Injuries/pathology , Whole-Body Irradiation/veterinary , X-Ray Therapy , X-Rays/adverse effects , Young Adult
2.
Sci Rep ; 8(1): 4604, 2018 03 15.
Article in English | MEDLINE | ID: mdl-29545590

ABSTRACT

The development of novel small molecule inhibitors of the cancer-associated tropomyosin 3.1 (Tpm3.1) provides the ability to examine the metabolic function of specific actin filament populations. We have determined the ability of these anti-Tpm (ATM) compounds to regulate glucose metabolism in mice. Acute treatment (1 h) of wild-type (WT) mice with the compounds (TR100 and ATM1001) led to a decrease in glucose clearance due mainly to suppression of glucose-stimulated insulin secretion (GSIS) from the pancreatic islets. The impact of the drugs on GSIS was significantly less in Tpm3.1 knock out (KO) mice indicating that the drug action is on-target. Experiments in MIN6 ß-cells indicated that the inhibition of GSIS by the drugs was due to disruption to the cortical actin cytoskeleton. The impact of the drugs on insulin-stimulated glucose uptake (ISGU) was also examined in skeletal muscle ex vivo. In the absence of drug, ISGU was decreased in KO compared to WT muscle, confirming a role of Tpm3.1 in glucose uptake. Both compounds suppressed ISGU in WT muscle, but in the KO muscle there was little impact of the drugs. Collectively, this data indicates that the ATM drugs affect glucose metabolism in vivo by inhibiting Tpm3.1's function with few off-target effects.


Subject(s)
Actin Cytoskeleton/metabolism , Glucose/metabolism , Insulin-Secreting Cells/metabolism , Insulin/metabolism , Tropomyosin/antagonists & inhibitors , Actin Cytoskeleton/drug effects , Animals , Glucose/administration & dosage , Insulin-Secreting Cells/drug effects , Male , Mice , Mice, Knockout , Tropomyosin/physiology
3.
J Cell Sci ; 131(6)2018 03 19.
Article in English | MEDLINE | ID: mdl-29487177

ABSTRACT

Many actin filaments in animal cells are co-polymers of actin and tropomyosin. In many cases, non-muscle myosin II associates with these co-polymers to establish a contractile network. However, the temporal relationship of these three proteins in the de novo assembly of actin filaments is not known. Intravital subcellular microscopy of secretory granule exocytosis allows the visualisation and quantification of the formation of an actin scaffold in real time, with the added advantage that it occurs in a living mammal under physiological conditions. We used this model system to investigate the de novo assembly of actin, tropomyosin Tpm3.1 (a short isoform of TPM3) and myosin IIA (the form of non-muscle myosin II with its heavy chain encoded by Myh9) on secretory granules in mouse salivary glands. Blocking actin polymerization with cytochalasin D revealed that Tpm3.1 assembly is dependent on actin assembly. We used time-lapse imaging to determine the timing of the appearance of the actin filament reporter LifeAct-RFP and of Tpm3.1-mNeonGreen on secretory granules in LifeAct-RFP transgenic, Tpm3.1-mNeonGreen and myosin IIA-GFP (GFP-tagged MYH9) knock-in mice. Our findings are consistent with the addition of tropomyosin to actin filaments shortly after the initiation of actin filament nucleation, followed by myosin IIA recruitment.


Subject(s)
Actin Cytoskeleton/metabolism , Actins/metabolism , Nonmuscle Myosin Type IIA/metabolism , Tropomyosin/metabolism , Actin Cytoskeleton/genetics , Actins/genetics , Animals , Female , Male , Mice , Mice, Inbred C57BL , Myosin Heavy Chains , Nonmuscle Myosin Type IIA/genetics , Protein Binding , Secretory Vesicles/genetics , Secretory Vesicles/metabolism , Tropomyosin/genetics
4.
Cytoskeleton (Hoboken) ; 74(10): 379-389, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28834398

ABSTRACT

We have identified novel actin filaments defined by tropomyosin Tpm4.2 at the ER. EM analysis of mouse embryo fibroblasts (MEFs) isolated from mice expressing a mutant Tpm4.2 (Tpm4Plt53/Plt53 ), incapable of incorporating into actin filaments, revealed swollen ER structures compared with wild-type (WT) MEFs (Tpm4+/+ ). ER-to-Golgi, but not Golgi-to-ER trafficking was altered in the Tpm4Plt53/Plt53 MEFs following the transfection of the temperature sensitive ER-associated ts045-VSVg construct. Exogenous Tpm4.2 was able to rescue the ER-to-Golgi trafficking defect in the Tpm4Plt53/Plt53 cells. The treatment of WT MEFs with the myosin II inhibitor, blebbistatin, blocked the Tpm4.2-dependent ER-to-Golgi trafficking. The lack of an effect on ER-to-Golgi trafficking following treatment of MEFs with CK666 indicates that branched Arp2/3-containing actin filaments are not involved in anterograde vesicle trafficking. We propose that unbranched, Tpm4.2-containing filaments have an important role in maintaining ER/Golgi structure and that these structures, in conjunction with myosin II motors, mediate ER-to-Golgi trafficking.


Subject(s)
Actin Cytoskeleton/metabolism , Endoplasmic Reticulum/metabolism , Golgi Apparatus/metabolism , Myosin Type II/metabolism , Tropomyosin/metabolism , Actin Cytoskeleton/drug effects , Actin Cytoskeleton/genetics , Actins/genetics , Actins/metabolism , Animals , Brefeldin A/pharmacology , Cell Movement/genetics , Cell Movement/physiology , Cells, Cultured , Cytoskeletal Proteins/metabolism , Cytoskeleton/drug effects , Cytoskeleton/metabolism , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/genetics , Fibroblasts/metabolism , Golgi Apparatus/drug effects , Golgi Apparatus/genetics , Humans , Mice , Protein Transport/drug effects , Protein Transport/genetics , Tropomyosin/genetics
5.
Sci Rep ; 6: 23538, 2016 Mar 23.
Article in English | MEDLINE | ID: mdl-27004919

ABSTRACT

Sexual reproduction in animals requires close interactions with the opposite sex. These interactions may generate costs of reproduction, because mates can induce detrimental physiological or physical effects on one another, due to their interest in maximising their own fitness. To understand how a male's presence influences aspects of female physiology implicated in reproductive costs in mice, independent of offspring production, we paired females with vasectomised, castrated or intact males, or other females. Being paired with a male, irrespective of his gonadal status, increased female weight. This effect was transient in females paired with castrated males but more persistent in those with vasectomised males. Those paired with males also showed an increase in corticosterone, suggesting an increased stress response. However, this was dependent on the gonadal status of the male housing partner, since those housed with vasectomised males had lower corticosterone than those with castrated males. Altered energy metabolism was only detectable in pregnant females, and oxidative stress was not consistently affected by a female's housing partner. These results suggest that a male's presence alters female weight, and stresses associated with reproduction could be induced by simply the presence of a male, but reduced by mating and/or being solicited to mate.


Subject(s)
Corticosterone/analysis , Sexual Behavior, Animal/physiology , Stress, Physiological , Animals , Body Mass Index , Female , Male , Mice , Pregnancy
7.
Mol Biol Cell ; 26(13): 2475-90, 2015 Jul 01.
Article in English | MEDLINE | ID: mdl-25971798

ABSTRACT

ERK-regulated cell proliferation requires multiple phosphorylation events catalyzed first by MEK and then by casein kinase 2 (CK2), followed by interaction with importin7 and subsequent nuclear translocation of pERK. We report that genetic manipulation of a core component of the actin filaments of cancer cells, the tropomyosin Tm5NM1, regulates the proliferation of normal cells both in vitro and in vivo. Mouse embryo fibroblasts (MEFs) lacking Tm5NM1, which have reduced proliferative capacity, are insensitive to inhibition of ERK by peptide and small-molecule inhibitors, indicating that ERK is unable to regulate proliferation of these knockout (KO) cells. Treatment of wild-type MEFs with a CK2 inhibitor to block phosphorylation of the nuclear translocation signal in pERK resulted in greatly decreased cell proliferation and a significant reduction in the nuclear translocation of pERK. In contrast, Tm5NM1 KO MEFs, which show reduced nuclear translocation of pERK, were unaffected by inhibition of CK2. This suggested that it is nuclear translocation of CK2-phosphorylated pERK that regulates cell proliferation and this capacity is absent in Tm5NM1 KO cells. Proximity ligation assays confirmed a growth factor-stimulated interaction of pERK with Tm5NM1 and that the interaction of pERK with importin7 is greatly reduced in the Tm5NM1 KO cells.


Subject(s)
Actin Cytoskeleton/physiology , MAP Kinase Signaling System/physiology , Tropomyosin/physiology , Actin Cytoskeleton/genetics , Actin Cytoskeleton/metabolism , Active Transport, Cell Nucleus , Animals , Casein Kinase II/metabolism , Cell Line, Tumor , Cell Proliferation/physiology , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Mitogen-Activated Protein Kinase Kinases/metabolism , Phosphorylation , Tropomyosin/genetics , Tropomyosin/metabolism
8.
Traffic ; 16(7): 691-711, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25783006

ABSTRACT

Actin has an ill-defined role in the trafficking of GLUT4 glucose transporter vesicles to the plasma membrane (PM). We have identified novel actin filaments defined by the tropomyosin Tpm3.1 at glucose uptake sites in white adipose tissue (WAT) and skeletal muscle. In Tpm 3.1-overexpressing mice, insulin-stimulated glucose uptake was increased; while Tpm3.1-null mice they were more sensitive to the impact of high-fat diet on glucose uptake. Inhibition of Tpm3.1 function in 3T3-L1 adipocytes abrogates insulin-stimulated GLUT4 translocation and glucose uptake. In WAT, the amount of filamentous actin is determined by Tpm3.1 levels and is paralleled by changes in exocyst component (sec8) and Myo1c levels. In adipocytes, Tpm3.1 localizes with MyoIIA, but not Myo1c, and it inhibits Myo1c binding to actin. We propose that Tpm3.1 determines the amount of cortical actin that can engage MyoIIA and generate contractile force, and in parallel limits the interaction of Myo1c with actin filaments. The balance between these actin filament populations may determine the efficiency of movement and/or fusion of GLUT4 vesicles with the PM.


Subject(s)
Actin Cytoskeleton/metabolism , Glucose/metabolism , Tropomyosin/metabolism , 3T3 Cells , Adipocytes/metabolism , Animals , Glucose Transporter Type 4/metabolism , Humans , Mice , Mice, Inbred C57BL , Myosin Type I/metabolism , Protein Binding , Protein Transport , Tropomyosin/genetics
9.
Cytoskeleton (Hoboken) ; 71(12): 663-76, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25545457

ABSTRACT

Epithelial cells generate contractile forces at their cell-cell contacts. These are concentrated at the specialized apical junction of the zonula adherens (ZA), where a ring of stabilized E-cadherin lies adjacent to prominent actomyosin bundles. Coupling of adhesion and actomyosin contractility yields tension in the junction. The biogenesis of junctional contractility requires actin assembly at the ZA as well as the recruitment of nonmuscle myosin II, but the molecular regulators of these processes are not yet fully understood. We now report a role for tropomyosins 5NM1 (Tm5NM1) and 5NM2 (Tm5NM2) in their generation. Both these tropomyosin isoforms were found at the ZA and their depletion by RNAi or pharmacological inhibition reduced both F-actin and myosin II content at the junction. Photoactivation analysis revealed that the loss of F-actin was attributable to a decrease in filament stability. These changes were accompanied by a decrease in E-cadherin content at junctions. Ultimately, both long-term depletion of Tm5NM1/2 and acute inhibition with drugs caused junctional tension to be reduced. Thus these tropomyosin isoforms are novel contributors to junctional contractility and integrity.


Subject(s)
Actin Cytoskeleton/metabolism , Actomyosin/metabolism , Adherens Junctions/metabolism , Epithelial Cells/metabolism , Tropomyosin/metabolism , Animals , Caco-2 Cells , Cell Communication/physiology , Fluorescent Antibody Technique , Gene Knockdown Techniques , Humans , Immunohistochemistry , Mice , Mice, Knockout , Protein Isoforms/metabolism , RNA, Small Interfering , Transfection
10.
Proc Natl Acad Sci U S A ; 111(22): 8119-24, 2014 Jun 03.
Article in English | MEDLINE | ID: mdl-24843175

ABSTRACT

Oxidative stress (an overproduction of reactive oxygen species in relation to defense mechanisms) may restrict investment in life history traits, such as growth, reproduction, lifespan, and the production of sexual signals to attract mates. The constraint on sexual signaling by oxidative stress is of particular interest because it has been proposed as a mechanism ensuring that only good-quality males produce the most attractive sexual signals. Despite these predictions, evidence supporting this theory is, at best, equivocal. We used a superoxide dismutase knockout mouse to demonstrate that oxidative stress directly impairs investment in morphological (preputial glands) and molecular (major urinary proteins) components of olfactory signaling essential for mate attraction. By maintaining males in a much more competitive environment than usual for mouse laboratory experiments, we also revealed a range of phenotypes of superoxide dismutase deficiency not observed in previous studies of this mouse model. This range included impaired bioenergetic function, which was undetectable in the control environment of this study. We urge further examination of model organisms in seminatural conditions and more competitive laboratory environments, as important phenotypes can be exposed under these more demanding conditions.


Subject(s)
Oxidative Stress/physiology , Sex Attractants/physiology , Sexual Behavior, Animal/physiology , Smell/physiology , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism , Animals , Competitive Behavior/physiology , Energy Metabolism/physiology , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Phenols/metabolism , Plant Extracts/metabolism , Signal Transduction/physiology , Social Environment , Superoxide Dismutase-1 , Territoriality
11.
Bioarchitecture ; 3(2): 25-37, 2013.
Article in English | MEDLINE | ID: mdl-23807088

ABSTRACT

While the general understanding of muscle regenerative capacity is that it declines with increasing age due to impairments in the number of muscle progenitor cells and interaction with their niche, studies vary in their model of choice, indices of myogenic repair, muscle of interest and duration of studies. We focused on the net outcome of regeneration, functional architecture, compared across three models of acute muscle injury to test the hypothesis that satellite cells maintain their capacity for effective myogenic regeneration with age. Muscle regeneration in extensor digitorum longus muscle (EDL) of young (3 mo-old), old (22 mo-old) and senescent female mice (28 mo-old) was evaluated for architectural features, fiber number and central nucleation, weight, collagen and fat deposition. The 3 injury paradigms were: a myotoxin (notexin) which leaves the blood vessels and nerves intact, freezing (FI) that damages local muscle, nerve and blood vessels and denervation-devascularization (DD) which dissociates the nerves and blood vessels from the whole muscle. Histological analyses revealed successful architectural regeneration following notexin injury with negligible fibrosis and fully restored function, regardless of age. In comparison, the regenerative response to injuries that damaged the neurovascular supply (FI and DD) was less effective, but similar across the ages. The focus on net regenerative outcome demonstrated that old and senescent muscle has a robust capacity to regenerate functional architecture.


Subject(s)
Aging/physiology , Muscle, Skeletal/physiology , Regeneration/physiology , Animals , Female , Immunohistochemistry , Mice , Mice, Inbred C57BL , Muscle, Skeletal/injuries , Muscle, Skeletal/pathology , Recovery of Function
12.
J Invest Dermatol ; 133(5): 1330-9, 2013 May.
Article in English | MEDLINE | ID: mdl-23303457

ABSTRACT

Precise orchestration of actin polymer into filaments with distinct characteristics of stability, bundling, and branching underpins cell migration. A key regulator of actin filament specialization is the tropomyosin family of actin-associating proteins. This multi-isoform family of proteins assemble into polymers that lie in the major groove of polymerized actin filaments, which in turn determine the association of molecules that control actin filament organization. This suggests that tropomyosins may be important regulators of actin function during physiological processes dependent on cell migration, such as wound healing. We have therefore analyzed the requirement for tropomyosin isoform expression in a mouse model of cutaneous wound healing. We find that mice in which the 9D exon from the TPM3/γTm tropomyosin gene is deleted (γ9D -/-) exhibit a more rapid wound-healing response 7 days after wounding compared with wild-type mice. Accelerated wound healing was not associated with increased cell proliferation, matrix remodeling, or epidermal abnormalities, but with increased cell migration. Rac GTPase activity and paxillin phosphorylation are elevated in cells from γ9D -/- mice, suggesting the activation of paxillin/Rac signaling. Collectively, our data reveal that tropomyosin isoform expression has an important role in temporal regulation of cell migration during wound healing.


Subject(s)
Cell Movement/physiology , Skin/injuries , Skin/physiopathology , Tropomyosin/metabolism , Wound Healing/physiology , Animals , Cell Line, Tumor , Cell Proliferation , Cells, Cultured , Extracellular Matrix/metabolism , Female , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Models, Animal , Paxillin/metabolism , Phosphorylation , Signal Transduction/physiology , Tropomyosin/deficiency , Tropomyosin/genetics , rac GTP-Binding Proteins/metabolism
13.
Brain ; 134(Pt 12): 3516-29, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22067542

ABSTRACT

Nemaline myopathy, the most common congenital myopathy, is caused by mutations in genes encoding thin filament and thin filament-associated proteins in skeletal muscles. Severely affected patients fail to survive beyond the first year of life due to severe muscle weakness. There are no specific therapies to combat this muscle weakness. We have generated the first knock-in mouse model for severe nemaline myopathy by replacing a normal allele of the α-skeletal actin gene with a mutated form (H40Y), which causes severe nemaline myopathy in humans. The Acta1(H40Y) mouse has severe muscle weakness manifested as shortened lifespan, significant forearm and isolated muscle weakness and decreased mobility. Muscle pathologies present in the human patients (e.g. nemaline rods, fibre atrophy and increase in slow fibres) were detected in the Acta1(H40Y) mouse, indicating that it is an excellent model for severe nemaline myopathy. Mating of the Acta1(H40Y) mouse with hypertrophic four and a half LIM domains protein 1 and insulin-like growth factor-1 transgenic mice models increased forearm strength and mobility, and decreased nemaline pathologies. Dietary L-tyrosine supplements also alleviated the mobility deficit and decreased the chronic repair and nemaline rod pathologies. These results suggest that L-tyrosine may be an effective treatment for muscle weakness and immobility in nemaline myopathy.


Subject(s)
Muscle Weakness/genetics , Muscle, Skeletal/pathology , Myopathies, Nemaline/drug therapy , Myopathies, Nemaline/genetics , Tyrosine/therapeutic use , Animals , Disease Models, Animal , Hand Strength , Hypertrophy/genetics , Hypertrophy/pathology , Mice , Mice, Transgenic , Muscle Contraction/genetics , Muscle Weakness/drug therapy , Muscle Weakness/pathology , Mutation , Myopathies, Nemaline/pathology , Phenotype
14.
J Muscle Res Cell Motil ; 30(5-6): 187-97, 2009.
Article in English | MEDLINE | ID: mdl-19997772

ABSTRACT

In addition to the highly specialized contractile apparatus, it is becoming increasingly clear that there is an extensive actin cytoskeleton which underpins a wide range of functions in striated muscle. Isoforms of cytoskeletal actin and actin-associated proteins (non-muscle myosins, cytoskeletal tropomyosins, and cytoskeletal alpha-actinins) have been detected in a number of regions of striated muscle: the sub-sarcolemmal costamere, the Z-disc and the T-tubule/sarcoplasmic reticulum membranes. As the only known function of these proteins is through association with actin filaments, their presence in striated muscles indicates that there are spatially and functionally distinct cytoskeletal actin filament systems in these tissues. These filaments are likely to have important roles in mechanical support, ion channel function, myofibrillogenenous and vesicle trafficking.


Subject(s)
Actin Cytoskeleton/physiology , Actins/physiology , Cytoskeleton/physiology , Muscle, Striated/physiology , Actin Cytoskeleton/ultrastructure , Actins/ultrastructure , Cytoskeleton/ultrastructure , Muscle Contraction/physiology , Muscle Proteins/physiology , Muscle, Striated/ultrastructure , Sarcolemma/physiology , Sarcolemma/ultrastructure , Sarcoplasmic Reticulum/physiology , Sarcoplasmic Reticulum/ultrastructure
15.
Cell Motil Cytoskeleton ; 66(9): 710-20, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19530183

ABSTRACT

We have identified a number of extra-sarcomeric actin filaments defined by cytoskeletal tropomyosin (Tm) isoforms. Expression of a cytoskeletal Tm (Tm3) not normally present in skeletal muscle in a transgenic mouse resulted in muscular dystrophy. In the present report we show that muscle pathology in this mouse is late onset (between 2 and 6 months of age) and is predominately in the back and paraspinal muscles. In the Tm3 mice, Evans blue dye uptake in muscle and serum levels of creatine kinase were markedly increased following downhill exercise, and the force drop following a series of lengthening contractions in isolated muscles (extensor digitorum longus) was also significantly increased in these mice. These results demonstrate that expression of an inappropriate Tm in skeletal muscle results in increased susceptibility to contraction-induced damage. The extra-sarcomeric actin cytoskeleton therefore may have an important role in protecting the muscle from contractile stress.


Subject(s)
Actin Cytoskeleton/metabolism , Cytoskeleton/metabolism , Muscle Contraction , Muscle, Skeletal/metabolism , Muscular Dystrophies/physiopathology , Tropomyosin/metabolism , Animals , Creatine Kinase/blood , Male , Mice , Mice, Transgenic , Muscle, Skeletal/pathology , Muscular Dystrophies/metabolism , Muscular Dystrophies/pathology , Protein Isoforms/metabolism , Tropomyosin/genetics
16.
Mol Biol Cell ; 20(1): 400-9, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19005216

ABSTRACT

The functional diversity of the actin microfilaments relies in part on the actin binding protein tropomyosin (Tm). The muscle-specific Tms regulate actin-myosin interactions and hence contraction. However, there is less known about the roles of the numerous cytoskeletal isoforms. We have shown previously that a cytoskeletal Tm, Tm5NM1, defines a Z-line adjacent cytoskeleton in skeletal muscle. Recently, we identified a second cytoskeletal Tm in this region, Tm4. Here we show that Tm4 and Tm5NM1 define separate actin filaments; the former associated with the terminal sarcoplasmic reticulum (SR) and other tubulovesicular structures. In skeletal muscles of Tm5NM1 knockout (KO) mice, Tm4 localization was unchanged, demonstrating the specificity of the membrane association. Tm5NM1 KO muscles exhibit potentiation of T-system depolarization and decreased force rundown with repeated T-tubule depolarizations consistent with altered T-tubule function. These results indicate that a Tm5NM1-defined actin cytoskeleton is required for the normal excitation-contraction coupling in skeletal muscle.


Subject(s)
Muscle Contraction/physiology , Muscle, Skeletal/physiology , Protein Isoforms/metabolism , Tropomyosin/metabolism , Actins/metabolism , Animals , Immunohistochemistry , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle, Skeletal/pathology , Muscle, Skeletal/ultrastructure , Protein Isoforms/genetics , Tropomyosin/genetics
17.
J Cell Biol ; 183(6): 1033-48, 2008 Dec 15.
Article in English | MEDLINE | ID: mdl-19075112

ABSTRACT

Regulators of skeletal muscle mass are of interest, given the morbidity and mortality of muscle atrophy and myopathy. Four-and-a-half LIM protein 1 (FHL1) is mutated in several human myopathies, including reducing-body myopathy (RBM). The normal function of FHL1 in muscle and how it causes myopathy remains unknown. We find that FHL1 transgenic expression in mouse skeletal muscle promotes hypertrophy and an oxidative fiber-type switch, leading to increased whole-body strength and fatigue resistance. Additionally, FHL1 overexpression enhances myoblast fusion, resulting in hypertrophic myotubes in C2C12 cells, (a phenotype rescued by calcineurin inhibition). In FHL1-RBM C2C12 cells, there are no hypertrophic myotubes. FHL1 binds with the calcineurin-regulated transcription factor NFATc1 (nuclear factor of activated T cells, cytoplasmic, calcineurin-dependent 1), enhancing NFATc1 transcriptional activity. Mutant RBM-FHL1 forms aggregate bodies in C2C12 cells, sequestering NFATc1 and resulting in reduced NFAT nuclear translocation and transcriptional activity. NFATc1 also colocalizes with mutant FHL1 to reducing bodies in RBM-afflicted skeletal muscle. Therefore, via NFATc1 signaling regulation, FHL1 appears to modulate muscle mass and strength enhancement.


Subject(s)
Intracellular Signaling Peptides and Proteins/metabolism , Muscle Proteins/metabolism , Muscle, Skeletal/pathology , Muscular Diseases/pathology , Animals , Calcineurin/metabolism , Cell Fusion , GATA2 Transcription Factor/metabolism , Humans , Hypertrophy , LIM Domain Proteins , Mice , Muscle Fibers, Skeletal/metabolism , Muscle Fibers, Skeletal/pathology , Muscle, Skeletal/metabolism , Muscular Diseases/metabolism , Mutation/genetics , Myoblasts/metabolism , Myoblasts/pathology , NFATC Transcription Factors/metabolism , Organ Size , Protein Binding , Signal Transduction , Transcription, Genetic , Transcriptional Activation
18.
Hum Mol Genet ; 17(8): 1076-86, 2008 Apr 15.
Article in English | MEDLINE | ID: mdl-18178581

ABSTRACT

A common nonsense polymorphism (R577X) in the ACTN3 gene results in complete deficiency of the fast skeletal muscle fiber protein alpha-actinin-3 in an estimated one billion humans worldwide. The XX null genotype is under-represented in elite sprint athletes, associated with reduced muscle strength and sprint performance in non-athletes, and is over-represented in endurance athletes, suggesting that alpha-actinin-3 deficiency increases muscle endurance at the cost of power generation. Here we report that muscle from Actn3 knockout mice displays reduced force generation, consistent with results from human association studies. Detailed analysis of knockout mouse muscle reveals reduced fast fiber diameter, increased activity of multiple enzymes in the aerobic metabolic pathway, altered contractile properties, and enhanced recovery from fatigue, suggesting a shift in the properties of fast fibers towards those characteristic of slow fibers. These findings provide the first mechanistic explanation for the reported associations between R577X and human athletic performance and muscle function.


Subject(s)
Actinin/genetics , Actinin/metabolism , Muscle, Skeletal/physiology , Physical Endurance/genetics , Animals , Body Weight/genetics , Female , Humans , Male , Mice , Mice, Knockout , Muscle Fibers, Fast-Twitch/physiology , Muscle Fibers, Slow-Twitch/physiology , Muscle Strength/genetics , Muscle, Skeletal/pathology
19.
Adv Exp Med Biol ; 644: 143-57, 2008.
Article in English | MEDLINE | ID: mdl-19209820

ABSTRACT

A number of congenital muscle diseases and disorders are caused by mutations in genes that encode the proteins present in or associated with the thin filaments of the muscle sarcomere. These genes include alpha-skeletal actin (ACTA1), beta-tropomyosin (TPM2), alpha-tropomyosin slow (TPM3), nebulin (NEB), troponin I fast (TNNI2), troponin T slow (TNNT1), troponin T fast (TNNT3) and cofilin (CFL2). Mutations in two of the four tropomyosin (Tm) genes, TPM2 and TPM3, result in at least three different skeletal muscle diseases and one disorder as distinguished by the presence of specific clinical features and/or structural abnormalities--nemaline myopathy (TPM2 and TPM3), distal arthrogryposis (TPM2), cap disease (TPM2) and congenital fiber type disproportion (TPM3). These diseases have overlapping clinical features and pathologies and there are cases of family members who have the same mutation, but different diseases (Table 1). The relatively recent discovery of nonmuscle or cytoskeletal Tms in skeletal muscle adds to this complexity since it is now possible that a disease-causing mutation could be in a striated isoform and a cytoskeletal isoform both present in muscle.


Subject(s)
Muscle, Skeletal/metabolism , Muscular Diseases/metabolism , Tropomyosin/chemistry , Tropomyosin/physiology , Actins/metabolism , Animals , Cytoskeleton/metabolism , Genotype , Humans , Mice , Models, Biological , Models, Genetic , Muscles/metabolism , Mutation , Protein Isoforms , Sarcomeres/metabolism , Tropomyosin/metabolism
20.
Cell Motil Cytoskeleton ; 65(1): 73-85, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17968984

ABSTRACT

The organisation of structural proteins in muscle into highly ordered sarcomeres occurs during development, regeneration and focal repair of skeletal muscle fibers. The involvement of cytoskeletal proteins in this process has been documented, with nonmuscle gamma-actin found to play a role in sarcomere assembly during muscle differentiation and also shown to be up-regulated in dystrophic muscles which undergo regeneration and repair [Lloyd et al.,2004; Hanft et al.,2006]. Here, we show that a cytoskeletal tropomyosin (Tm), Tm4, defines actin filaments in two novel compartments in muscle fibers: a Z-line associated cytoskeleton (Z-LAC), similar to a structure we have reported previously [Kee et al.,2004], and longitudinal filaments that are orientated parallel to the sarcomeric apparatus, present during myofiber growth and repair/regeneration. Tm4 is upregulated in paradigms of muscle repair including induced regeneration and focal repair and in muscle diseases with repair/regeneration features, muscular dystrophy and nemaline myopathy. Longitudinal Tm4-defined filaments also are present in diseased muscle. Transition of the Tm4-defined filaments from a longitudinal to a Z-LAC orientation is observed during the course of muscle regeneration. This Tm4-defined cytoskeleton is a marker of growth and repair/regeneration in response to injury, disease state and stress in skeletal muscle.


Subject(s)
Muscle, Skeletal/metabolism , Regeneration/physiology , Tropomyosin/physiology , Adult , Animals , Biomarkers , Child , Child, Preschool , Cytoskeleton/metabolism , Disease Models, Animal , Humans , Infant , Infant, Newborn , Mice , Mice, Inbred mdx , Middle Aged , Muscle, Skeletal/pathology , Muscle, Skeletal/physiopathology , Muscular Dystrophies/metabolism , Muscular Dystrophies/physiopathology , Myopathies, Nemaline/metabolism , Myopathies, Nemaline/physiopathology , Sarcomeres/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...