Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
J Cell Sci ; 2024 Sep 23.
Article in English | MEDLINE | ID: mdl-39308425

ABSTRACT

Unambiguous targeting of cellular structures for in situ cryo-electron microscopy in the heterogeneous, dense, and compacted environment of the cytoplasm remains challenging. Here we have developed a cryogenic correlative light and electron microscopy (cryo-CLEM) workflow which combines thin cells grown on a mechanically defined substratum to rapidly analyse organelles and macromolecular complexes by cryo-electron tomography (cryo-ET). We coupled these advancements with optogenetics to redistribute perinuclear-localised organelles to the cell periphery, allowing visualisation of organelles otherwise positioned in cellular regions too thick for cryo-ET. This reliable and robust workflow allows for fast in situ analyses without the requirement for cryo-focused ion beam milling. Using this protocol, cells can be frozen, imaged by cryo-fluorescence microscopy and be ready for batch cryo-ET within a day.

2.
J Muscle Res Cell Motil ; 41(1): 11-22, 2020 03.
Article in English | MEDLINE | ID: mdl-31054005

ABSTRACT

The interactions of cytoskeletal actin filaments with myosin family motors are essential for the integrity and function of eukaryotic cells. They support a wide range of force-dependent functions. These include mechano-transduction, directed transcellular transport processes, barrier functions, cytokinesis, and cell migration. Despite the indispensable role of tropomyosins in the generation and maintenance of discrete actomyosin-based structures, the contribution of individual cytoskeletal tropomyosin isoforms to the structural and functional diversification of the actin cytoskeleton remains a work in progress. Here, we review processes that contribute to the dynamic sorting and targeted distribution of tropomyosin isoforms in the formation of discrete actomyosin-based structures in animal cells and their effects on actin-based motility and contractility.


Subject(s)
Actins/metabolism , Tropomyosin/metabolism , Humans
4.
Exp Physiol ; 99(4): 664-74, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24414176

ABSTRACT

Muscles of mdx mice are known to be more susceptible to contraction-induced damage than wild-type muscle. However, it is not clear whether this is because of dystrophin deficiency or because of the abnormal branching morphology of dystrophic muscle fibres. This distinction has an important bearing on our traditional understanding of the function of dystrophin as a mechanical stabilizer of the sarcolemma. In this study, we address the question: 'Does dystrophin-positive, regenerated muscle containing branched fibres also show an increased susceptibility to contraction-induced damage?' We produced a model of fibre branching by injecting dystrophin-positive extensor digitorum longus muscles with notexin. The regenerated muscle was examined at 21 days postinjection. Notexin-injected muscle contained 29% branched fibres and was not more susceptible to damage from mild eccentric contractions than contralateral saline-injected control muscle. Regenerated muscles also had greater mass, greater cross-sectional area and lower specific force than control muscles. We conclude that the number of branched fibres in this regenerated muscle is below the threshold needed to increase susceptibility to damage. However, it would serve as an ideal control for muscles of young mdx mice, allowing for clearer differentiation of the effects of dystrophin deficiency from the effects of fibre regeneration and morphology.


Subject(s)
Elapid Venoms , Muscle Development , Muscle, Skeletal/physiopathology , Muscular Diseases/physiopathology , Regeneration , Animals , Disease Models, Animal , Dystrophin/metabolism , Male , Mice, Inbred C57BL , Muscle Contraction , Muscle Fibers, Skeletal/metabolism , Muscle Fibers, Skeletal/pathology , Muscle Strength , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscular Diseases/chemically induced , Muscular Diseases/metabolism , Muscular Diseases/pathology , Recovery of Function , Time Factors
5.
Int J Obes (Lond) ; 34(3): 524-36, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20029376

ABSTRACT

OBJECTIVE: The Ski gene regulates skeletal muscle differentiation in vitro and and in vivo. In the c-Ski overexpression mouse model there occurs marked skeletal muscle hypertrophy with decreased adipose tissue mass. In this study, we have investigated the underlying molecular mechanisms responsible for the increased skeletal muscle and decreased adipose tissue mass in the c-Ski mouse. APPROACH: Growth and body composition analysis (tissue weights and dual energy X-ray absorptiometry) coupled with skeletal muscle and white adipose gene expression and metabolic phenotyping in c-Ski mice and wild-type (WT) littermate controls was performed. RESULTS: The growth and body composition studies confirmed the early onset of accelerated body growth, with increased lean mass and decreased fat mass in the c-Ski mice. Gene expression analysis in skeletal muscle from c-Ski mice compared with WT mice showed significant differences in myogenic and lipogenic gene expressions that are consistent with the body composition phenotype. Skeletal muscle of c-Ski mice had significantly repressed Smad1, 4, 7 and myostatin gene expression and elevated myogenin, myocyte enhancer factor 2, insulin-like growth factor-1 receptor and insulin-like growth factor-2 expression. Strikingly, expression of the mRNAs encoding the master lipogenic regulators, sterol-regulatory enhancer binding protein 1c (SREBP1c), and the nuclear receptor liver X-receptor-alpha, and their downstream target genes, SCD-1 and FAS, were suppressed in skeletal muscle of c-Ski mice, as were the expressions of other nuclear receptors involved in adipogenesis and metabolism, such as peroxisome proliferator-activated receptor-gamma, glucocorticoid receptor and retinoic acid receptor-related orphan receptor-alpha. Transfection analysis demonstrated Ski repressed the SREBP1c promoter. Moreover, palmitate oxidation and oxidative enzyme activity was increased in skeletal muscle of c-Ski mice. These results suggest that the Ski phenotype involves attenuated lipogenesis, decreased myostatin signalling, coupled to increased myogenesis and fatty acid oxidation. CONCLUSION: Ski regulates several genetic programs and signalling pathways that regulate skeletal muscle and adipose mass to influence body composition development, suggesting that Ski may have a role in risk for obesity and metabolic disease.


Subject(s)
Body Composition/genetics , DNA-Binding Proteins/genetics , Lipogenesis/genetics , Muscle, Skeletal/physiology , Proto-Oncogene Proteins/genetics , Animals , Body Composition/physiology , DNA-Binding Proteins/physiology , Fatty Acids/metabolism , Gene Silencing , Growth/physiology , Mice , Mice, Transgenic , Myostatin/metabolism , Proto-Oncogene Proteins/physiology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sterol Regulatory Element Binding Protein 1/metabolism , Thinness/genetics , Thinness/metabolism
6.
Cell Motil Cytoskeleton ; 58(3): 200-11, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15146538

ABSTRACT

We describe a simple culture method for obtaining highly differentiated clonal C2C12 myotubes using a feeder layer of confluent fibroblasts, and document the expression of contractile protein expression and aspects of myofibre morphology using this system. Traditional culture methods using collagen- or laminin-coated tissue-culture plastic typically results in a cyclic pattern of detachment and reformation of myotubes, rarely producing myotubes of a mature adult phenotype. C2C12 co-culture on a fibroblast substratum facilitates the sustained culture of contractile myotubes, resulting in a mature sarcomeric register with evidence for peripherally migrating nuclei. Immunoblot analysis demonstrates that desmin, tropomyosin, sarcomeric actin, alpha-actinin-2 and slow myosin are detected throughout myogenic differentiation, whereas adult fast myosin heavy chain isoforms, members of the dystrophin-associated complex, and alpha-actinin-3 are not expressed at significant levels until >6 days of differentiation, coincident with the onset of contractile activity. Electrical stimulation of mature myotubes reveals typical and reproducible calcium transients, demonstrating functional maturation with respect to calcium handling proteins. Immunocytochemical staining demonstrates a well-defined sarcomeric register throughout the majority of myotubes (70-80%) and a striated staining pattern is observed for desmin, indicating alignment of the intermediate filament network with the sarcomeric register. We report that culture volume affects the fusion index and rate of sarcomeric development in developing myotubes and propose that a fibroblast feeder layer provides an elastic substratum to support contractile activity and likely secretes growth factors and extracellular matrix proteins that assist myotube development.


Subject(s)
Cell Nucleus/physiology , Fibroblasts/metabolism , Muscle Fibers, Fast-Twitch/metabolism , Muscle Fibers, Skeletal/physiology , Myosins/biosynthesis , Adult , Animals , Calcium Signaling/physiology , Cell Communication/physiology , Cell Differentiation/physiology , Cell Nucleus/ultrastructure , Cell Survival/physiology , Cells, Cultured , Coculture Techniques/methods , Elasticity , Extracellular Matrix Proteins/metabolism , Growth Substances/metabolism , Humans , Infant , Infant, Newborn , Intermediate Filaments/physiology , Intermediate Filaments/ultrastructure , Mice , Microscopy, Electron, Transmission , Muscle Contraction/physiology , Muscle Fibers, Fast-Twitch/ultrastructure , Muscle Fibers, Skeletal/ultrastructure , Muscle Proteins/metabolism , Myosin Heavy Chains/metabolism , Sarcomeres/metabolism , Sarcomeres/ultrastructure , Time Factors
7.
Neuromuscul Disord ; 12(10): 952-7, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12467751

ABSTRACT

Skeletal muscle function was measured in anaesthetised transgenic mice having a mutation in the TPM3 gene (slow alpha-tropomyosin), a similar mutation as found in some patients with nemaline myopathy, and was compared with control muscles. Measurements of isometric and dynamic muscle performance were done with electrical nerve stimulation at physiological temperatures. No muscle weakness was found in the transgenic muscles when performance was measured at muscle optimum length. This was true not only with full activation but also at lower activation levels, indicating that calcium sensitivity was not affected at this length. Also, fatigability was not affected in these conditions. However, isometric force of the muscles with the mutation in TPM3 was lower at lengths below optimum, with more impairment at decreasing length. As the muscles are active over a large range of different muscle lengths during daily activities, this finding may explain, at least in part, the muscle weakness experienced by patients with nemaline myopathy.


Subject(s)
Isometric Contraction/physiology , Muscle Weakness/genetics , Muscle, Skeletal/physiopathology , Mutation , Tropomyosin/genetics , Animals , Electric Stimulation , Female , In Vitro Techniques , Isometric Contraction/genetics , Mice , Mice, Transgenic , Muscle Fatigue/genetics , Muscle Fatigue/physiology , Muscle Weakness/physiopathology
8.
Am J Hum Genet ; 68(6): 1333-43, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11333380

ABSTRACT

Nemaline myopathy (NM) is a clinically and genetically heterogeneous disorder characterized by muscle weakness and the presence of nemaline bodies (rods) in skeletal muscle. Disease-causing mutations have been reported in five genes, each encoding a protein component of the sarcomeric thin filament. Recently, we identified mutations in the muscle alpha-skeletal-actin gene (ACTA1) in a subset of patients with NM. In the present study, we evaluated a new series of 35 patients with NM. We identified five novel missense mutations in ACTA1, which suggested that mutations in muscle alpha-skeletal actin account for the disease in approximately 15% of patients with NM. The mutations appeared de novo and represent new dominant mutations. One proband subsequently had two affected children, a result consistent with autosomal dominant transmission. The seven patients exhibited marked clinical variability, ranging from severe congenital-onset weakness, with death from respiratory failure during the 1st year of life, to a mild childhood-onset myopathy, with survival into adulthood. There was marked variation in both age at onset and clinical severity in the three affected members of one family. Common pathological features included abnormal fiber type differentiation, glycogen accumulation, myofibrillar disruption, and "whorling" of actin thin filaments. The percentage of fibers with rods did not correlate with clinical severity; however, the severe, lethal phenotype was associated with both severe, generalized disorganization of sarcomeric structure and abnormal localization of sarcomeric actin. The marked variability, in clinical phenotype, among patients with different mutations in ACTA1 suggests that both the site of the mutation and the nature of the amino acid change have differential effects on thin-filament formation and protein-protein interactions. The intrafamilial variability suggests that alpha-actin genotype is not the sole determinant of phenotype.


Subject(s)
Actins/genetics , Muscle, Skeletal/metabolism , Mutation, Missense/genetics , Myopathies, Nemaline/genetics , Actins/chemistry , Adolescent , Adult , Amino Acid Sequence , Australia , Child , Child, Preschool , Cohort Studies , DNA Mutational Analysis , Female , Humans , Immunohistochemistry , Infant , Infant, Newborn , Male , Microscopy, Electron , Middle Aged , Models, Molecular , Molecular Sequence Data , Muscle, Skeletal/pathology , Muscle, Skeletal/physiopathology , Muscle, Skeletal/ultrastructure , Myopathies, Nemaline/metabolism , Myopathies, Nemaline/pathology , Myopathies, Nemaline/physiopathology , Phenotype , Protein Conformation , Protein Isoforms/chemistry , Protein Isoforms/genetics , RNA, Messenger/analysis , RNA, Messenger/genetics
9.
J Cell Sci ; 114(Pt 3): 513-24, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11171321

ABSTRACT

Muscle differentiation is characterized by the induction of genes encoding contractile structural proteins and the repression of nonmuscle isoforms from these gene families. We have examined the importance of this regulated order of gene expression by expressing the two sarcomeric muscle actins characteristic of the differentiated state, i.e. alpha-skeletal and alpha-cardiac actin, in C2 mouse myoblasts. Precocious accumulation of transcripts and proteins for a group of differentiation-specific genes was elicited by alpha-skeletal actin only: four muscle tropomyosins, two muscle actins, desmin and MyoD. The nonmuscle isoforms of tropomyosin and actin characteristic of the undifferentiated state continued to be expressed, and no myosin heavy or light chain or troponin transcripts characteristic of muscle differentiation were induced. Stable transfectants displayed a substantial reduction in cell surface area and in the levels of nonmuscle tropomyosins and beta-actin, consistent with a relationship between the composition of the actin cytoskeleton and cell surface area. The transfectants displayed normal cell cycle progression. We propose that alpha-skeletal actin can activate a regulatory pathway linking a subset of muscle genes that operates independently of normal differentiation and withdrawal from the cell cycle.


Subject(s)
Actins/physiology , Cell Cycle , Cell Differentiation , Muscle, Skeletal/metabolism , Protein Isoforms/physiology , Actins/genetics , Animals , Base Sequence , Cell Movement/physiology , DNA Primers , Fluorescent Antibody Technique, Indirect , Gene Expression Regulation , Humans , Mice , Microscopy, Fluorescence , Muscle, Skeletal/cytology , MyoD Protein/genetics , Protein Isoforms/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Transfection
10.
Hum Mol Genet ; 10(4): 317-28, 2001 Feb 15.
Article in English | MEDLINE | ID: mdl-11157795

ABSTRACT

Nemaline myopathy is a hereditary disease of skeletal muscle defined by a distinct pathology of electron-dense accumulations within the sarcomeric units called rods, muscle weakness and, in most cases, a slow oxidative (type 1) fiber predominance. We generated a transgenic mouse model to study this disorder by expressing an autosomal dominant mutant of alpha-tropomyosin(slow) previously identified in a human cohort. Rods were found in all muscles, but to varying extents which did not correlate with the amount of mutant protein present. In addition, a pathological feature not commonly associated with this disorder, cytoplasmic bodies, was found in the mouse and subsequently identified in human samples. Muscle weakness is a major feature of this disease and was examined with respect to fiber composition, degree of rod-containing fibers, fiber mechanics and fiber diameter. Hypertrophy of fast, glycolytic (type 2B) fibers was apparent at 2 months of age. Muscle weakness was apparent in mice at 5-6 months of age, mimicking the late onset observed in humans with this mutation. The late onset did not correlate with observed changes in fiber type and rod pathology. Rather, the onset of muscle weakness correlates with an age-related decrease in fiber diameter and suggests that early onset is prevented by hypertrophy of fast, glycolytic fibers. We suggest that the clinical phenotype is precipitated by a failure of the hypertrophy to persist and therefore compensate for muscle weakness.


Subject(s)
Muscle Development , Muscle Fibers, Slow-Twitch/pathology , Muscle Weakness/genetics , Muscle, Skeletal/growth & development , Muscle, Skeletal/pathology , Myopathies, Nemaline/genetics , Point Mutation , Tropomyosin/genetics , Amino Acid Substitution/genetics , Animals , Arginine/genetics , Disease Models, Animal , Dissection , Female , Glycolysis/genetics , Humans , Hypertrophy , Inclusion Bodies/pathology , Inclusion Bodies/ultrastructure , Methionine/genetics , Mice , Mice, Inbred Strains , Mice, Transgenic , Microtubules/pathology , Microtubules/ultrastructure , Muscle Fibers, Fast-Twitch/drug effects , Muscle Fibers, Fast-Twitch/metabolism , Muscle Fibers, Fast-Twitch/pathology , Muscle Fibers, Fast-Twitch/ultrastructure , Muscle Fibers, Slow-Twitch/drug effects , Muscle Fibers, Slow-Twitch/metabolism , Muscle Fibers, Slow-Twitch/ultrastructure , Muscle Weakness/pathology , Muscle Weakness/physiopathology , Muscle, Skeletal/physiopathology , Muscle, Skeletal/ultrastructure , Myopathies, Nemaline/pathology , Myopathies, Nemaline/physiopathology , Oxidation-Reduction , RNA, Messenger/biosynthesis , Sarcoplasmic Reticulum/pathology , Sarcoplasmic Reticulum/ultrastructure , Strontium/pharmacology , Tropomyosin/biosynthesis
11.
J Biol Chem ; 274(21): 14948-55, 1999 May 21.
Article in English | MEDLINE | ID: mdl-10329696

ABSTRACT

Expression analysis of a novel cDNA isolated from immortal murine fibroblasts revealed a single transcript of 3.0 kilobase pairs that was highly expressed in mouse and human striated muscle and in mouse heart. The gene has therefore been named striamin. Its expression was confined to skeletal muscle types with a fast glycolytic (2B) contractile phenotype. It was also detected in C2C12 mouse myoblasts and was down-regulated during in vitro myogenesis. The cDNA has a single open reading frame encoding a predicted 16.8-kDa protein of 149 amino acids with no homology to known proteins. Microinjection and transfection of green fluorescence protein-tagged striamin demonstrated that it localizes to the nucleus. Coimmunoprecipitations revealed that it can interact with p53 (a positive marker for myoblast differentiation) in vivo and in vitro. Furthermore, it repressed p53 activity in p53-mediated reporter assays. Fluorescence in situ hybridization with a mouse P1 genomic clone localized the gene to chromosome 12C3, which is syntenic to human chromosome 14q21-22.


Subject(s)
Carrier Proteins/genetics , Carrier Proteins/isolation & purification , Muscle Proteins , Muscle, Skeletal , Tumor Suppressor Protein p53/physiology , Amino Acid Sequence , Animals , Base Sequence , Carrier Proteins/physiology , Chromosome Mapping , Cloning, Molecular , DNA, Complementary , Humans , Mice , Molecular Sequence Data , Nuclear Proteins
12.
Mol Cell Biol ; 18(11): 6641-52, 1998 Nov.
Article in English | MEDLINE | ID: mdl-9774679

ABSTRACT

The molecular mechanisms which are responsible for restricting skeletal muscle gene expression to specific fiber types, either slow or fast twitch, are unknown. As a first step toward defining the components which direct slow-fiber-specific gene expression, we identified the sequence elements of the human troponin I slow upstream enhancer (USE) that bind muscle nuclear proteins. These include an E-box, a MEF2 element, and two other elements, USE B1 and USE C1. In vivo analysis of a mutation that disrupts USE B1 binding activity suggested that the USE B1 element is essential for high-level expression in slow-twitch muscles. This mutation does not, however, abolish slow-fiber specificity. A similar analysis indicated that the USE C1 element may play only a minor role. We report the cloning of a novel human USE B1 binding protein, MusTRD1 (muscle TFII-I repeat domain-containing protein 1), which is expressed predominantly in skeletal muscle. Significantly, MusTRD1 contains two repeat domains which show remarkable homology to the six repeat domains of the recently cloned transcription factor TFII-I. Furthermore, both TFII-I and MusTRD1 bind to similar but distinct sequences, which happen to conform with the initiator (Inr) consensus sequence. Given the roles of MEF2 and basic helix-loop-helix (bHLH) proteins in muscle gene expression, the similarity of TFII-I and MusTRD1 is intriguing, as TFII-I is believed to coordinate the interaction of MADS-box proteins, bHLH proteins, and the general transcription machinery.


Subject(s)
DNA-Binding Proteins/chemistry , DNA-Binding Proteins/physiology , Enhancer Elements, Genetic/genetics , Muscle Proteins/chemistry , Trans-Activators , Transcription Factors/chemistry , Troponin I/genetics , Amino Acid Sequence , Animals , Base Sequence , Binding Sites/genetics , Cloning, Molecular , Helix-Loop-Helix Motifs/genetics , Humans , Immunohistochemistry , Mice , Mice, Transgenic , Molecular Sequence Data , Muscle, Skeletal/cytology , Muscle, Skeletal/metabolism , Nuclear Proteins/metabolism , Protein Biosynthesis/genetics , Rats , Sequence Analysis, DNA , Sequence Deletion/genetics
13.
Genes Dev ; 12(17): 2748-58, 1998 Sep 01.
Article in English | MEDLINE | ID: mdl-9732272

ABSTRACT

We report a novel mechanism of gene regulation in skeletal muscle fibers. Within an individual myofiber nucleus, not all muscle loci are transcriptionally active at a given time and loci are regulated independently. This phenomenon is particularly remarkable because the nuclei within a myofiber share a common cytoplasm. Both endogenous muscle-specific and housekeeping genes and transgenes are regulated in this manner. Therefore, despite the uniform protein composition of the contractile apparatus along the length of the fiber, the loci that encode this structure are not transcribed continuously. The total number of active loci for a particular gene is dynamic, changing during fetal development, regeneration, and in the adult, and potentially reflects the growth status of the fiber. The data reveal that transcription in particular stages of muscle fiber maturation occurs in pulses and is defined by a stochastic mechanism.


Subject(s)
Gene Expression Regulation , Muscle Fibers, Skeletal/metabolism , Muscle, Skeletal/metabolism , Aging , Animals , Cell Nucleus/metabolism , Embryonic and Fetal Development , Gene Expression Regulation, Developmental , Mice , Mice, Transgenic , Muscle Contraction , Muscle Fibers, Skeletal/physiology , Muscle, Skeletal/physiology , Regeneration , Transcription, Genetic , beta-Galactosidase/biosynthesis , beta-Galactosidase/genetics
14.
J Appl Physiol (1985) ; 84(3): 1083-7, 1998 Mar.
Article in English | MEDLINE | ID: mdl-9480972

ABSTRACT

We examined the regulation of the troponin I slow (TnIs) promoter during skeletal muscle unloading-induced protein isoform transition, by using a transgenic mouse line harboring the -4,200 to +12 base pairs region of the human TnIs promoter. Eighteen female transgenic mice ( approximately 30 g body mass) were randomly divided into two groups: weight-bearing (WB) controls (n = 9) and hindlimb unloaded (HU; n = 9). The HU mice were tail suspended for 7 days. Body mass was unchanged in the WB group but was reduced (-6%; P < 0.05) after the HU treatment. Absolute soleus muscle mass (-25%) and soleus mass relative to body mass (-16%) were both lower (P < 0.05) in the HU group compared with the WB mice. Northern blot analyses indicate that 7 days of HU result in a 64% decrease (P < 0.05) in the abundance of endogenous TnIs mRNA (microg/mg muscle) in the mouse soleus. Furthermore, there is a trend for the abundance of the fast troponin I mRNA to be increased (+34%). Analysis of transgenic chloramphenicol acetyltransferase activity in the soleus muscle revealed no difference (P > 0.05) between WB and HU groups. We conclude that additional elements are necessary for the TnIs gene to respond to an unloading-induced, slow-to-fast isoform transition stimulus.


Subject(s)
Hindlimb Suspension/physiology , Muscle, Skeletal/physiology , Troponin I/physiology , Animals , Blotting, Northern , Body Weight/physiology , Chloramphenicol O-Acetyltransferase/biosynthesis , Chloramphenicol O-Acetyltransferase/genetics , Chloramphenicol O-Acetyltransferase/metabolism , DNA Probes , Female , Humans , Mice , Mice, Transgenic , Muscle, Skeletal/enzymology , Organ Size/physiology , RNA/biosynthesis , Transcription, Genetic , Troponin I/genetics
15.
Am J Physiol ; 273(6): C1801-10, 1997 12.
Article in English | MEDLINE | ID: mdl-9435483

ABSTRACT

The postnatal expression profiles of alpha-sarcomeric actin transcripts and protein are quantified in mouse striated muscles from birth to postnatal day 56 by Northern and Western blot analyses. alpha-Cardiac actin (alpha-CA) transcripts transiently increase between 12 and 21 days after birth in the quadriceps muscle, reaching approximately 90% that found in the adult mouse heart. Although alpha-CA is the alpha-sarcomeric actin isoform expressed in the immature fiber, the expression profiles of other contractile protein isoforms indicate that this postnatal period is not reflective of an immature phenotype. alpha-Skeletal actin (alpha-SA) transcripts accumulate to approximately 32% of the total alpha-sarcomeric actin transcripts in the adult heart. Our study shows that 1) there is a simultaneous reappearance of alpha-CA and alpha-SA in postnatal skeletal and heart muscles, respectively, and 2) the contractile protein gene expression profile characteristic of adult skeletal muscle is not achieved until after 42 days postnatal in the mouse. We propose there is a previously uncharacterized period of postnatal striated muscle maturation marked by the reappearance of the minor alpha-sarcomeric actins.


Subject(s)
Actins/biosynthesis , Aging/metabolism , Gene Expression Regulation, Developmental , Muscle, Skeletal/metabolism , Myocardium/metabolism , Sarcomeres/metabolism , Transcription, Genetic , Animals , Animals, Newborn , Crosses, Genetic , Female , Heart/growth & development , Male , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Muscle Development , Muscle, Skeletal/growth & development , Myosin Light Chains/biosynthesis , RNA, Messenger/biosynthesis , Troponin/biosynthesis , Troponin T
16.
DNA Cell Biol ; 14(7): 599-607, 1995 Jul.
Article in English | MEDLINE | ID: mdl-7626219

ABSTRACT

Troponin I (TnI) is a muscle-specific protein involved in the calcium-mediated contraction of striated muscle. Three TnI isoforms have been identified, each encoded by a separate gene and expressed in specific striated muscles in the adult. The slow isoform gene (TnIs) is transcriptionally regulated during skeletal muscle development such that its expression in the adult is restricted to muscle fibers innervated by a slow nerve. To delineate regions of this gene that are responsive to information imparted by the slow nerve, we generated transgenic mice carrying -4,200 to +12 bp of the human TnIs gene linked to the bacterial chloramphenicol acetyltransferase (CAT) coding region. By Northern blot analysis, we detected transgene transcripts only in muscles containing slow-twitch fibers. CAT histochemical analysis revealed that expression of the transgene is restricted solely to slow-twitch fibers as characterized by type I myosin heavy-chain (MyHC) expression. Using regeneration as a model for neural influenced expression, we show that this gene construct also contains sequences necessary to respond to cues from the central nervous system.


Subject(s)
Muscle Fibers, Slow-Twitch/metabolism , Muscle, Skeletal/metabolism , Promoter Regions, Genetic , Troponin/genetics , Animals , Chloramphenicol O-Acetyltransferase/biosynthesis , DNA Probes , Humans , Mice , Mice, Transgenic , Muscle Fibers, Slow-Twitch/cytology , Muscle, Skeletal/cytology , Organ Specificity , Recombinant Fusion Proteins/biosynthesis , Troponin/biosynthesis , Troponin I
17.
Dev Biol ; 169(2): 487-503, 1995 Jun.
Article in English | MEDLINE | ID: mdl-7781893

ABSTRACT

The differentiation and diversification of striated muscle is a complex process involving numerous temporal and spatial alterations in the pattern of contractile protein isoform gene expression. In order to gain insight into the regulation of contractile protein isoform changes during skeletal and cardiac muscle formation, the expression of a transgene comprising a chloramphenicol acetyltransferase (CAT) reporter gene linked with sequences from -4200 to +12 of the human slow skeletal troponin I (TnIs) gene, and all three endogenous mouse troponin I (TnI) isoform genes, was investigated in embryonic, neonatal, and postnatal mice. The -4200 TnIsCAT transgene was properly activated in the limb and trunk skeletal muscle primordia and the early embryonic atrium and ventricle of the heart. Along with the endogenous mouse TnIs gene, expression of the CAT transgene began to segregate into the presumptive slow-twitch myofibers at late fetal stages and expression declined in the neonatal and postnatal heart except for the conductive tissues, in which expression persisted into adulthood. However, expression of the CAT transgene during development did not completely follow the endogenous mouse TnIs gene. The expression of the CAT transgene was aberrantly low in the embryonic cardiac outflow tract and the ventricles of the fetal heart. In addition to its expression in striated muscles, the transgene was expressed aberrantly in the primordial axial skeleton. We conclude that the upstream sequences from the human TnIs gene contain sufficient regulatory information to confer appropriate transgene expression during the early differentiation of skeletal muscles and during the establishment of fiber type upon the maturation of myofibers. However, additional regulatory elements are likely to be required for correct temporal and spatial regulation in the heart and somitic mesoderm during development. In vitro DNA transfection of cultured skeletal and cardiac muscle cells identified a cell type-specific enhancer element within the first intron of the TnIs gene whose absence in the transgene may account for the aberrant expression observed in vivo. In addition, we provide the first evidence that the fast-twitch skeletal muscle isoform of troponin I, TnIf, is transiently expressed during early cardiac muscle development.


Subject(s)
Gene Expression Regulation, Developmental , Muscle, Skeletal/metabolism , Troponin/genetics , Animals , Animals, Newborn , Base Sequence , Chloramphenicol O-Acetyltransferase/genetics , Enhancer Elements, Genetic , Female , Fetus/metabolism , Heart/embryology , Heart/growth & development , Humans , Male , Mice , Mice, Transgenic , Molecular Sequence Data , Muscle Development , Muscle, Skeletal/embryology , Muscle, Skeletal/growth & development , Myocardium/metabolism , Troponin I
18.
Proc Natl Acad Sci U S A ; 92(13): 6185-9, 1995 Jun 20.
Article in English | MEDLINE | ID: mdl-7597099

ABSTRACT

Contractile proteins are encoded by multigene families, most of whose members are differentially expressed in fast- versus slow-twitch myofibers. This fiber-type-specific gene regulation occurs by unknown mechanisms and does not occur within cultured myocytes. We have developed a transient, whole-animal assay using somatic gene transfer to study this phenomenon and have identified a fiber-type-specific regulatory element within the promoter region of a slow myofiber-specific gene. A plasmid-borne luciferase reporter gene fused to various muscle-specific contractile gene promoters was differentially expressed when injected into slow- versus fast-twitch rat muscle: the luciferase gene was preferentially expressed in slow muscle when fused to a slow troponin I promoter, and conversely, was preferentially expressed in fast muscle when fused to a fast troponin C promoter. In contrast, the luciferase gene was equally well expressed by both muscle types when fused to a nonfiber-type-specific skeletal actin promoter. Deletion analysis of the troponin I promoter region revealed that a 157-bp enhancer conferred slow-muscle-preferential activity upon a minimal thymidine kinase promoter. Transgenic analysis confirmed the role of this enhancer in restricting gene expression to slow-twitch myofibers. Hence, somatic gene transfer may be used to rapidly define elements that direct myofiber-type-specific gene expression prior to the generation of transgenic mice.


Subject(s)
Chloramphenicol O-Acetyltransferase/analysis , Gene Expression Regulation, Enzymologic , Luciferases/analysis , Muscle Fibers, Skeletal/physiology , Muscle, Skeletal/physiology , Transcription, Genetic , Transfection , Animals , Cells, Cultured , Chloramphenicol O-Acetyltransferase/biosynthesis , DNA Transposable Elements , Female , Luciferases/biosynthesis , Mice , Mice, Transgenic , Muscle Fibers, Skeletal/metabolism , Muscle, Skeletal/metabolism , Organ Specificity , Plasmids , Promoter Regions, Genetic , Rats , Rats, Sprague-Dawley
19.
Am J Physiol ; 268(2 Pt 1): C466-71, 1995 Feb.
Article in English | MEDLINE | ID: mdl-7864086

ABSTRACT

Ten rats were exposed to 9 days of zero gravity aboard the National Aeronautics and Space Administration SLS-1 space mission (June 1991). Levels of fast and slow isoform mRNAs from six contractile protein gene families were quantified in the flight soleus and extensor digitorum longus (EDL) muscles. The gene families studied were myosin light chain-1 (MLC-1), myosin light chain-2 (MLC-2), troponin (Tn) T, TnI, TnC, and tropomyosin. In the EDL muscle there was no change in slow mRNA levels with a general increase in fast mRNA levels from 23 to 232%. Changes in slow mRNA levels were seen in the flight soleus muscle with TnCslow and TnTslow levels increasing slightly, and MLC-1slow a, MLC-1slow b, TnIslow, alpha-Tmslow, and MLC-2slow levels decreasing. All fast mRNA levels increased in the flight soleus muscle from 170 to 1,100%. We can conclude that exposure to zero gravity results in 1) a general increase in fast mRNA levels in both fast and slow muscles and 2) differing directional changes in slow mRNA accumulation in the soleus muscle.


Subject(s)
Contractile Proteins/genetics , RNA, Messenger/metabolism , Space Flight , Weightlessness , Animals , Contractile Proteins/chemistry , Hindlimb , Isomerism , Muscles/metabolism , Rats , Rats, Sprague-Dawley , Time Factors , Toes
20.
J Biol Chem ; 269(16): 12212-9, 1994 Apr 22.
Article in English | MEDLINE | ID: mdl-8163527

ABSTRACT

In order to elucidate mechanisms involved in striated muscle contractile protein isoform expression, we have defined regulatory elements in the cardiac actin gene necessary for postnatal expression at the level of transcript accumulation in the heart and hindlimb muscles of transgenic mice. During this developmental period in the rodent, cardiac actin expression essentially remains constant in the heart, but declines significantly in skeletal muscle. We determined that a 13-kilobase human cardiac actin gene fragment contains sufficient information to direct this maturation-based developmental expression, as well as striated muscle-specific and high level expression. We localized an element responsible for maturation-based down-regulation in the 3' flank of the gene between approximately 950 and 2120 base pairs downstream of the polyadenylation site. Furthermore, we determined that -800 base pairs of 5'-flanking DNA, which contains multiple MyoD1 binding sites, as well as serum response element and AP1 binding sites, can account for striated muscle-specific expression, but not high level expression. Findings indicate that sequence(s) responsible for high level expression of the gene must be located within the body of the gene. We conclude that the human cardiac actin gene contains distinct sequences which confer developmental, tissue-specific, and high level expression.


Subject(s)
Actins/biosynthesis , Actins/genetics , Aging/metabolism , Gene Expression , Muscles/metabolism , Myocardium/metabolism , Animals , Heart/growth & development , Humans , Mice , Mice, Transgenic , Muscle Development , Plasmids , RNA, Messenger/biosynthesis , RNA, Messenger/metabolism , Restriction Mapping , Transcription, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL