Open Access

Mass spectrometry-based proteomic analysis of middle-aged vs. aged vastus lateralis reveals increased levels of carbonic anhydrase isoform 3 in senescent human skeletal muscle

  • Authors:
    • Lisa Staunton
    • Margit Zweyer
    • Dieter Swandulla
    • Kay Ohlendieck
  • View Affiliations

  • Published online on: July 6, 2012     https://doi.org/10.3892/ijmm.2012.1056
  • Pages: 723-733
  • Copyright: © Staunton et al. This is an open access article distributed under the terms of Creative Commons Attribution License [CC BY_NC 3.0].

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The age-related loss of skeletal muscle mass and associated progressive decline in contractile strength is a serious pathophysiological issue in the elderly. In order to investigate global changes in the skeletal muscle proteome after the fifth decade of life, this study analysed total extracts from human vastus lateralis muscle by fluorescence difference in-gel electrophoresis. Tissue specimens were derived from middle-aged (47-62 years) vs. aged (76-82 years) individuals and potential changes in the protein expression profiles were compared between these two age groups by a comprehensive gel electrophoresis-based survey. Age-dependent alterations in the concentration of 19 protein spots were revealed and mass spectrometry identified these components as being involved in the excitation-contraction-relaxation cycle, muscle metabolism, ion handling and the cellular stress response. This indicates a generally perturbed protein expression pattern in senescent human muscle. Increased levels of mitochondrial enzymes and isoform switching of the key contractile protein, actin, support the idea of glycolytic-to-oxidative and fast-to-slow transition processes during muscle aging. Importantly, the carbonic anhydrase (CA)3 isoform displayed an increased abundance during muscle aging, which was independently verified by immunoblotting of differently aged human skeletal muscle samples. Since the CA3 isoform is relatively muscle-specific and exhibits a fibre type-specific expression pattern, this enzyme may represent an interesting new biomarker of sarcopenia. Increased levels of CA are indicative of an increased demand of CO2-removal in senescent muscle, and also suggest age-related fibre type shifting to slower-contracting muscles during human aging.

Introduction

Although natural aging of the body may be considered a fundamental biological process (1), it is not clear which molecular and evolutionary genetic mechanisms trigger the progressive decline in physiological functions and decrease the rate of reproduction (2). Aging is most likely not due to active gene programming (3), but more likely based on evolved limitations in somatic maintenance (4). In humans, one of the most striking features of aging is a gradual reduction in skeletal muscle mass and a concomitant decline in contractile strength (5). This age-related progressive loss of muscle mass and function has been termed sarcopenia and is believed to be due to a multifactorial etiology (68). As reviewed by Berger and Doherty (9), epidemiological studies of age-related skeletal muscle wasting indicate that nearly half the population over 75 years of age is suffering from some form of sarcopenia leading in severe cases to loss of independence. Although the findings from a large number of longitudinal and cross-sectional studies of skeletal muscle aging (1019) do not concur on the exact rate and extent of contractile tissue loss (20), these investigations agree that human aging is associated with a general impairment of structural and functional elements of the musculoskeletal system (21). Since individual skeletal muscles are differentially affected during the natural aging process (22), one should be careful about extrapolating age-related changes in a particular muscle group to the aging process of the entire neuromuscular system.

Muscle wasting occurs in all aged individuals to a varying degree (23), but this degenerative process may be accelerated by a variety of exacerbating factors, such as lack of physical activity, improper nutritional intake, extensive pharmacotherapy and/or chronic illness (24). For example, extended periods of bed rest in elderly patients are clearly related to inactivity-induced insulin resistance and further complicate the sarcopenic syndrome (25). Advancing age is associated with the loss of spinal motor neurons due to apoptosis (26), an impaired capacity for axonal re-innervation of deinnervated muscle fibers (27) and age-induced low-grade inflammation (28). Histological hallmarks of sarcopenia are a reduction in fibre numbers, a decline in fibre size and potential loss of entire motor units, whereby preferential atrophy of type II fibres has been established as a consequence of aging (21,27,29). The diagnosis of sarcopenia is based on the combined presence of a low muscle mass and a low gait speed (3032). The clinical cut-off point for sarcopenia is considered to be a percentage of muscle mass two standard deviations below the mean measured in young adults of the same gender and ethnic background, as well as a walking speed below 0.8 m/sec in the 4 m walking test (31).

Since a reduction in skeletal muscle mass is pronounced in aged lower limb muscle groups, the human vastus lateralis muscle has been the center of attention in numerous studies on old age (33). Between the ages of 20 and 80 years, the cross-sectional area of the vastus lateralis muscle may be reduced by up to 40% (34,35). Epidemiological studies have indicated accelerated muscle wasting after the fifth decade with an approximately 2% reduction in muscle mass per year (9). We carried out a comparative survey of total extracts from middle-aged vs. aged vastus lateralis muscle tissue. Fluorescence difference in-gel electrophoresis (DIGE) in combination with mass spectrometry (MS) was employed to study human muscle aging, since this advanced method of modern protein biochemistry is capable of swiftly evaluating potential changes in large numbers of protein species (36). Over the last decade, MS-based proteomics has identified numerous novel protein factors involved in myogenesis, muscle differentiation, fibre transitions and various neuromuscular pathologies, as summarised in recent reviews (3739). With respect to the natural aging process, proteomics technology has been applied to study altered protein expression levels associated with cellular changes in skeletal muscle tissues (40), whereby most studies have focused on animal models of aging, such as the senescent Wistar rat (4151).

A previous proteomic survey of human muscle aging has investigated differences in protein expression between young adults (20–25 years) and aged (70–76 years) individuals (52). In order to build on these findings and to determine potential changes after the fifth decade of life, we used fluorescent tagging of the vastus lateralis muscle proteome from middle-aged (47–62 years) vs. aged (76–82 years) individuals and conducted a comprehensive gel electrophoresis-based survey of human skeletal muscle aging. Densitometric scanning revealed a differential expression pattern for 19 2-dimensional (2D) protein spots and the subsequent mass spectrometric analysis identified these muscle-associated proteins as being involved in contraction, relaxation, ion homeostasis, cellular stress response and fibre metabolism. Alterations in key muscle proteins from vastus lateralis muscle support the idea of generally perturbed protein expression patterns during human aging and agree with the notion that sarcopenia is associated with the fast-to-slow and glycolytic-to-oxidative transition processes (5254). Of note, the muscle-specific carbonic anhydrase (CA)3 isoform (EC 4.2.1.1) (55), which has not been previously identified by proteomic surveys of human muscle aging (38,52), showed an increase in senescent skeletal muscle by both MS-based proteomics and independent immunoblot analysis. Elevated levels of CA3 may be indicative of an increased demand for efficient CO2-removal during fibre aging and/or age-related fibre type shifting to slower-contracting muscle populations (54). The establishment of a novel biomarker of muscle aging after the fifth decade of life may be exploitable for the future development of a more reliable assay to diagnose sarcopenia in old age.

Materials and methods

Materials

For the fluorescence 2D-DIGE analysis of aged human skeletal muscles, immobilised pH gradient (IPG) drystrips pH 3–11, CyDye DIGE Fluor minimal dyes Cy3 and Cy5, Coomassie Brilliant Blue, ampholytes, acetonitrile, cover fluid, and the 2D-Clean-Up kit were purchased from Amersham Biosciences/GE Healthcare (Little Chalfont, UK). Ultrapure protogel acrylamide stock solutions were from National Diagnostics (Atlanta, GA, USA). Sequencing grade-modified trypsin for peptide generation was obtained from Promega (Madison, WI, USA). LC-MS Formic acid and Chromasolv water were purchased from Fluka Chemical Corp. (Milwaukee, WI, USA) and spin filters were from Fisher Scientific (Loughborough, UK). Chemiluminescence substrate, protease inhibitors and nitrocellulose sheets were from Pierce and Warriner (Chester, UK), Roche Diagnostics (Manheim, Germany) and Millipore (Bedford, MA, USA), respectively. For immunoblotting, primary antibodies to the CA3 isoform were purchased from Abcam (Cambridge, UK) and secondary peroxidase-conjugated antibodies were from Chemicon International, Inc. (Temecula, CA, USA). Ultrapure lysine for quenching the DIGE labelling reaction, DNase I and all general reagents were otained from Sigma Chemical Co. (Dorset, UK).

Human skeletal muscle biopsy samples

Since the neuromuscular system of the vastus lateralis muscle is intensively studied in human physiology and biochemistry, and frequently targeted during biopsy procedures for diagnostic purposes or routine biomedical investigations, age-related changes in this muscle were studied by MS-based proteomics. Two groupings of biopsy material representing healthy middle-aged vs. healthy aged vastus lateralis muscle were provided by the associated hospitals of the University of Bonn according to German ethics regulations (56). Middle-aged specimens derived from 47-, 55-, 59- and 62-year-old muscle tissue are marked in this study as samples MA1 to MA4. Aged specimens derived from 76-, 77-, 81- and 82-year-old muscle tissue are marked as samples OA1 to OA4. Following biopsy, fresh tissue specimens were immediately frozen in liquid nitrogen, transported on dry ice and then stored at −80°C prior to usage.

Preparation of urea-soluble fraction of skeletal muscle proteome

Freshly thawed skeletal muscle biopsy material (100 mg) was ground into a fine powder in the presence of liquid nitrogen using a pestle and mortar. Pulverised human muscle specimens of different ages were resuspended in 1 ml of ice-cold buffer containing 7 M urea, 2 M thiourea, 65 mM CHAPS, 10 mM Trizma base, 1% ampholytes pH 3–11, and 100 mM dithiothreitol. In order to eliminate excessive viscosity of the protein extract due to large amounts of muscle-derived DNA and to minimise protein degradation due to the presence of muscle-associated proteases, the solution was supplemented with 2 μl of DNase I (200 units; Sigma Chemical Co.) per 100 μl buffer and a protease inhibitor cocktail (Roche Diagnostics) (57). The muscle tissue homogenate was gently mixed by vortexing and then placed on a bench top shaker for 2.5 h at room temperature to precipitate the total urea-soluble protein complement. Following centrifugation of the suspension at 20,000 x g in an Eppendorf 5417R centrifuge (Eppendorf, Hamburg, Germany) for 20 min, the protein-containing pellet was washed in 5 ml of ice-cold 100% acetone and thoroughly broken up by vortexing and sonication. Using 80% acetone, washing and centrifugation was repeated twice and the final protein precipitate was collected by centrifugation and resuspended in 1 ml of the above described buffer. Individual protein samples were incubated for 1 h at room temperature with careful vortexing every 10 min for 5 sec. Determination of protein concentration and removal of potentially interfering contaminants was performed using the Bradford assay system (58) and 2D-Clean-Up kit from Amersham Biosciences/GE Healthcare (59), respectively. Finally, protein pellets representing middle-aged and aged skeletal muscle protein fractions were resuspended in DIGE lysis buffer (7 M urea, 2 M thiourea, 30 mM Tris, 4% CHAPS, pH 8.5) and adjusted to a protein concentration of 1 mg/ml.

Fluorescence DIGE analysis

The gel electrophoretic separation of fluorescently tagged muscle proteins was performed with a total amount of 100 μg protein per 2D-DIGE gel. To keep potential impurities to a minimum, electrophoretic separation steps were performed under designated fume hoods. For each protein preparation derived from differently aged vastus lateralis muscle, a 50 μg protein aliquote was fluorescently-labelled with 200 pmol DIGE fluor dyes for 30 min on ice and in the dark. Tissue extracts from eight different muscle samples representing 47–82 years of age were each labelled with Cy3 dye. In addition, pooled internal standards were prepared with Cy5 dye, as previously described in detail (60). Quenching of the fluorescent labelling reaction was carried out with 10 mM lysine for 10 min on ice. For separation in the first dimension, isoelectric focusing (IEF) was carried out with 24 cm strips in an Amersham IPGphor system using the following running conditions: 30 V for 2 h, 500 V for 2.5 h, 1,000 V for 1 h, 2,000 V for 1 h, 4000 V for 1 h, 6,000 V for 1 h, 800 V for 3 h, 500 V for 1.5 h and finally 8,000 V for 2.5 h. Gel strips were then equilibrated for 20 min in reducing buffer containing 100 mM dithiothreitol, followed by 10 min of alkylation in buffer containing 250 mM iodoacetamide. For separation in the second dimension, slab gel electrophoresis was carried out with an Amersham Ettan Dalt-twelve system, using 12.5% gels. Eight slab gels were run in parallel at 0.5 W/gel for 60 min and then 15 W/gel until the blue dye front had disappeared from the bottom of the gel. Cy3- and Cy5-labelled muscle-associated proteins were visualised with the assistance of an Amersham Typhoon Trio variable mode imager. Individual 2D-DIGE gels were warped to a single master gel prior to analysis. Potential changes in the protein expression profile of middle-aged vs. aged skeletal muscle samples were analysed using Progenesis SameSpots analysis software from Nonlinear Dynamics (Newcastle upon Tyne, UK), using the following parameters: n=4; t-test P<0.05; and a power value of >0.8. Muscle proteins with a significantly changed density were selected for tryptic digestion from Coomassie Brilliant Blue-stained preparative gels (61).

Mass spectrometric identification of skeletal muscle proteins

The mass spectrometric identification of proteins of particular interest was carried out in a dedicated and air-conditioned proteomics suite with semi-clean analytical status to avoid potential contamination of samples. Protein spots were digested by standardised in-gel trypsination to generate distinct peptide populations (62). MS analysis was performed on a Model 6340 Ion Trap LC/MS apparatus from Agilent Technologies (Santa Clara, CA, USA). Spot excision, washing, destaining and treatment with protease were performed by previously optimised methods (57,60,61). Trypsin-generated peptide mixtures were dried through vacuum centrifugation and then resuspended in MS-grade distilled water and 0.1% (v/v) formic acid, spun down through spin filter and added to LC-MS viles for identification by ion trap LC-MS analysis. A nanoflow Aligent 1200 series system was employed for peptide separation. Analytical samples were loaded into the enrichment at a capillary flow rate set to 2 μl/min with a mix of 0.1% (v/v) formic acid and 50% (v/v) acetonitrile and formic acid at a ratio of 19:1. The voltage was set to 1,700 V. Database searches were carried out with Mascot MS/MS Ion search (Matrix Science, London, UK). All searches used ‘Homo sapiens’ as a taxonomic category and the following parameters: i) two missed cleavages by trypsin, ii) mass tolerance of precursor ions ±2.5 Da and product ions ±0.7 Da, iii) carboxymethylated cysteins fixed modification, and iv) oxidation of methionine as variable modification. In addition, only hits with at least two matched distinct peptides and a Mascot score of at least 44 were considered significant proteomic findings.

Verification of age-related changes in the expression of carbonic anhydrase by immunoblotting

In order to verify age-related changes in the abundance of CA, immunoblot analysis was carried out to determine the expression levels of the CA3 isoform in middle-aged vs. aged vastus lateralis muscle preparations. The electrophoretic transfer of muscle proteins to Immobilon NC-pure nitrocellulose membranes was carried out for 1 h at 100 V and 4°C with a Mini-Protean transfer system from Bio-Rad Laboratories (Hemel-Hempstead, UK). Transfer efficiency was routinely evaluated by reversible Ponceau S-Red staining of membrane sheets. Nitrocellulose membranes were blocked for 1 h in 5% (w/v) fat-free milk powder dissolved in phosphate-buffered saline [PBS; 50 mM sodium phosphate, 0.9% (w/v) NaCl, pH 7.4]. Blocked and washed membranes were incubated for 3 h at room temperature with appropriately diluted primary antibody to the CA3 isoform, followed by a gentle washing in blocking buffer and then incubated with an appropriate dilution of peroxidase-conjugated secondary antibody for 1 h at room temperature (57). Immuno-decorated membranes were washed again in blocking solution and a final rinse in PBS. Antibody-labelled protein bands were visualised with the SuperSignal-type enhanced chemiluminescence kit from Pierce and Warriner. Densitometric scanning of immunoblots was performed on a 300 S computing densitometer (Molecular Dynamics, Sunnyvale, CA, USA) with ImageJ (NIH, USA) and GraphPad Prism (GraphPad Software, Inc., San Diego, CA, USA) software.

Results

In order to evaluate age-dependent alterations in the skeletal muscle proteome after the fifth decade of life, we applied the fluorescence 2D-DIGE technique and MS for studying vastus lateralis specimens from middle-aged vs. aged individuals. In our current study, the comparative gel electrophoresis-based proteomic study of crude muscle extracts focused on urea-soluble proteins, which is clearly reflected by the identification of mostly soluble and relatively abundant protein species. Shown is an overview of the analytical DIGE gels used in this proteomic study (Fig. 1), a DIGE master gel outlining protein spots with a significant age-related change in abundance (Fig. 2), a listing of changed proteins in senescent vastus lateralis muscle as determined by MS analysis (Table I) and immunoblotting of changed expression levels in a muscle-specific protein, the CA3 isoform (Fig. 3). In order to correlate MS-identified protein species listed in Table I with distinct protein spots of altered concentration in the DIGE master gel shown in Fig. 2, the numbering system of proteins in the table and corresponding gel is identical.

Table I.

List of DIGE-identified proteins with a changed abundance in middle aged vs. aged human vastus lateralis muscle.

Table I.

List of DIGE-identified proteins with a changed abundance in middle aged vs. aged human vastus lateralis muscle.

Spot no.Name of identified proteinAccession no.Molecular mass (kDa)Iso-electric point pIMascot scoreCoverage (%)Peptides matchedAnovaFold changeSequences
1Actin, α skeletal musclegi|33563240423725.2313528102.32E-04−3.8AGFAGDDAPR, AVFPSIVGRPR, HQGVMVGMGQK, DSYVGDEAQSK, DSYVGDEAQSKR, LDLAGR, DLTDYLMK, GYSFVTTAER, LCYVALDFENEMATAASSSSLEK, SYELPDGQVITIGNER
2Actin, α skeletal musclegi|33563240423725.2343536113.05E-04−2.6AGFAGDDAPR, DSYVGDEAQSK, DSYVGDEAQSKR, VAPEEHPTLLTEAPLNPK, DLTDYLMK, GYSFVTTAER, LCYVALDFENEMATAASSSSLEK, SYELPDGQVITIGNER, DLYANNVMSGGTTMYPGIADR, EITALAPSTMK, IIAPPER
3Annexin A5gi|6753060357884.8346520.002−2.5VLTEIIASR, MLVVLLQANR
4DJ-1 proteingi|55741460202406.32473667.00E-03−2.1ALVILAK, GAEEMETVIPVDVMR, VTVAGLAGK, GLIAAICAGPTALLAHEVGFGCK, DGLILTSR, APLVLKD
5Troponin T, slow skeletal musclegi|3449358299926.341981941.00E-03−2VDFDDIHR, DLLELQTLIDVHFEQR, VLSNMGAHFGGYLVK, YEINVLYNR
6Troponin T, slow skeletal musclegi|3449358299926.342232264.00E-03−2IPEGERVDFDDIHR, VDFDDIHR, DLLELQTLIDVHFEQR, KVLSNMGAHFGGYLVK, VLSNMGAHFGGYLVK, YEINVLYNR
7Glycogen phosphorylasegi|6755256976896.6560886.00E-03−1.9VIFLENYR, VIPAADLSEQISTAGTEASGTGNMK, GYNAQEYYDR, GYNAQEYYDRIPELR, IPELR, DIVNMLMHHDR, TIAQYAR, EIWGVEPSR
8Heat shock protein β-7gi|31542970186605.95441931.20E-02−1.8ALPAQDPPMEK, LAADGTVMNTFAHK, EDGSLTIR
9Actin, βgi|49868394515.7810123102.00E-03−1.7AVFPSIVGR, HQGVMVGMGQK, DSYVGDEAQSK, DSYVGDEAQSKR, LDLAGR, DLTDYLMK, SYELPDGQVITIGNER, EITALAPSTMK, IIAPPER, IIAPPERK
10Troponin C, slowgi|6678369185254.041212435.00E-03−1.7AAVEQLTEEQKNEFK, GKSEEELSDLFR, NADGYIDLDELK
11Myosin light chain MLC2gi|199985188704.711004066.00E-03−1.5DTFAALGR, EAPGPINFTVFLTMFGEK, GADPEETILNAFK, VFDPEGK, EMLTTQAER, NLVHIITHGEEKD
12Acyl-CoA dehydrogenase short chaingi|192659452088.961531241.30E-021.3GISAFLVPMPTPGLTLGK, IAMQTLDMGR, LADMALALESAR, ITEIYEGTSEIQR
13Heat shock-protein Hsp70gi|31560686698895.511751276.00E-031.4VEIIANDQGNR, TTPSYVAFTDTER, EIAEAYLGGK, DAGTITGLNVLR, IINEPTAAAIAYGLDK, FEELNADLFR, VCNPIISK
14Carbonic anhydrase 3gi|31982861296386.8947821.20E-021.5VVFDDTYDR, YAAELHLVHWNPK
15Creatine kinase M-typegi|6671762432506.58861241.00E-021.6GGDDLDPNYVLSSR, LSVEALNSLTGEFK, IEEIFK, GQSIDDMIPAQK
16Succinate dehydrogenase complex subunit Agi|3851614592666.1677421.30E-021.6LGANSLLDLVVFGR, SMQNHAAVFR
17Actin, α-cardiacgi|387090420485.233582193.00E-031.7AGFAGDDAPR, HQGVMVGMGQK, DSYVGDEAQSK, DSYVGDEAQSKR, DLTDYLMK, GYSFVTTAER, SYELPDGQVITIGNER, CDIDIRK, IIAPPER
18Calmodulingi|99032084166964.09594765.00E-031.7EAFSLFDKDGDGTITTK, MKDTDSEEEIR, DTDSEEEIREAFR, VFDKDGNGYISAAELR,DGNGYISAAELR, EADIDGDGQVNYEEFVQMMTAK
19 Phosphoglucomutasegi|33416468637056.0269954.35E-052VDLGVLGK, SMPTSGALDR, FFGNLMDASK, YDYEEVEAEGANK, LSGTGSAGATIR
Proteomic profiling of aging human vastus lateralis muscle

In this study, high-resolution fluorescence 2D gel electrophoresis, in combination with densitometric analysis using a Typhoon Trio variable imager and image analysis with progenesis 2D analysis software, resulted in the identification of 19 protein spots with a significant change in concentration levels in middle-aged vs. aged individuals. Shown are analytical DIGE gels of protein extracts from 47-, 55-, 59- and 62-year-old muscle tissue (Fig. 1A–D) and from 76-, 77-, 81-and 82-year-old muscle tissue (Fig. 1I–L), labelled with Cy3 dye. Pooled standards, labelled with Cy5 dye, are depicted in Fig. 1E–H and M–P.

MS identification of skeletal muscle proteins with an age-related change in concentration

A DIGE master gel with electrophoretically separated human vastus lateralis muscle proteins is shown for a molecular mass range of approximately 10–150 kDa and a pI range of pH 3 to pH 11 (Fig. 2). Muscle-associated proteins with a potential age-dependent alteration in density ranged in molecular mass from apparent 16.7 kDa (calmodulin) to 97.7 kDa (glycogen phosphorylase) and covered a pI range from approximately 4.09 (calmodulin) to 8.96 (Acyl-CoA dehydrogenase). MS analysis identified 19 proteins species, which are listed in Table I outlining protein name, protein accession number, molecular mass, pI-value, Mascot score, percentage sequence coverage, number of matched peptide sequences, Anova score, fold change of individual proteins affected by muscle aging, and peptide sequences used to identify proteins. The majority of muscle-associated proteins identified by DIGE screening in combination with MS analysis were shown to be associated with the actomyosin apparatus, the cytoskeleton, metabolism, signalling and the cellular stress response. A reduced density was determined for 11 proteins and 8 proteins showed an increase in their abundance.

The vastus lateralis muscle protein with the highest fold decrease was shown in the muscle isoform of α-actin (spots 1, 2). Phosphoglucomutase (spot 19) was identified as the significantly increased enzyme in aged muscle tissue. In addition to muscle α-actin, muscle β-actin (spot 9), Annexin (spot 3), DJ-1 protein (spot 4), troponin subunits (spots 5, 6, 10), glycogen phosphorylase (spot 7), heat shock protein (Hsp) β-7 (spot 8), and myosin light chain (spot 11) were decreased in senescent human muscle. Besides phosphoglucomutase, increased muscle proteins were identified as Acyl-CoA dehydrogenase (spot 12), Hsp70 (spot 13), CA3 isoform (spot 14), muscle creatine kinase (spot 15), succinate dehydrogenase (spot 16), cardiac α-actin (spot 17) and calmodulin (spot 18).

In addition to the proteins listed in Table I, the DIGE method in combination with MS analysis identified two other proteins with a changed abundance in aged muscle; slow myosin light chain MLC3 (gi|33563264|; 22523 kDa, pI 5.03) and the voltage-dependent anion-selective channel VDAC2 (gi|6755965|; 32351 kDa, pI 7.44). The contractile protein, MLC3, was identified by six peptides (ALGQNPTQAEVLR, MMDFETFLPMLQHISK, NKDTGTYEDFVEGLR, EGNGTVMGAELR, HVLATLGER and LTEDEVEK) with a 35% sequence coverage and a Mascot score of 86, and the mitochondrial ion channel, VDAC2, was identified by three peptides (LTFDTTFSPNTGK, VNNSSLIGVGYTQTLRPGVK and LTLSALVDGKSFNAGGHK) with a 17% sequence coverage and a Mascot score of 64. Although their identification was based on a sufficient number of peptide sequences, the DIGE-based determination of their respective 2D spots displayed only minor fragments. Therefore, these proteins were not included in the listing of the main findings of this proteomic survey of human skeletal muscle aging.

Immunoblot analysis of CA3 in aging human skeletal muscle

Although the DIGE technique represents one of the most powerful comparative methods in modern biochemistry and MS analysis is highly accurate in identifying distinct protein species, we were interested in independently verifying the differential expression of a muscle-specific protein identified in this proteomic study on old age. Since the CA3 isoform is a relatively muscle-specific and fibre type-specific enzyme, comparative immunoblotting was carried out to confirm a key finding of the proteomic data presented in this study. The increased immuno-decoration of CA in aged vs. middle-aged specimens from vastus lateralis muscle (Fig. 3A–D) is in agreement with the proteomic establishment of higher levels of this muscle enzyme in senescent contractile tissue (Fig. 2 and Table I). A comparison of 47- vs. 76-year-old, 55- vs. 77-year-old, 59- vs. 81-year-old and 62- vs. 82-year-old skeletal muscle specimens revealed a statistically significant increase in CA3 isoform expression in senescent skeletal muscle (Fig. 3E).

Discussion

The progressive decline in skeletal muscle mass and the weakening of contractile strength is a major pathophysiological feature of the aged neuromuscular system. Often the frailty syndrome and muscular dysfunction present personal care problems for elderly individuals, limiting their independence and requiring them to seek outside help, despite other medical ailments (63). This warrants large-scale genomic, proteomic and metabolomic surveys of aged skeletal muscle in order to establish the underlying mechanisms of sarcopenia in old age. Over the last few years, a considerable number of molecular investigations involved in the genetic basis of sarcopenia have identified a large collection of differentially expressed genes in aged muscle tissue (64). Based on these genetic findings, it is crucial to verify which of the identified age-dependent alterations in gene expression patterns translate into changed concentration levels and/or post-translational modifications in distinct protein products that markedly modify skeletal muscle functions. The MS-based proteomic survey of human aging presented in this study focused on the elucidation of potential alterations in the muscle protein complement after the fifth decade of life. Our fluorescent DIGE study of the vastus lateralis muscle compared middle-aged (47–62 years) vs. aged (76–82 years) individuals in order to supplement a set of previously determined proteomic data on the same skeletal muscle from young adults (20–25 years) and aged (70–76 years) individuals (52). Based on the proteome-wide changes revealed by these extensive gel electrophoresis-based surveys, the illustrative scheme in Fig. 4 summarises our knowledge of major molecular and cellular events during human skeletal muscle aging. Aged human vastus lateralis muscle tissue, which exhibits progressive denervation, faulty re-innervation and reduction in cross-sectional area (21,34), is characterised by excitation-contraction uncoupling, an altered cellular stress response, impaired ion homeostasis, fast-to-slow fibre transitions and glycolytic-to-oxidative metabolic shifts (5254).

The comparative proteomic study of middle-aged vs. aged human muscle identified age-related changes in the expression of key proteins involved in the excitation-contraction-relaxation cycle, ion handling, the cellular stress response and muscle fibre bioenergetics. The apparent switch between the muscle isoform of actin with its cardiac counter-part (65) in aged vastus lateralis muscle supports the idea of a fast-to-slow transformation process in the actomyosin apparatus during skeletal muscle aging (54). However, the decrease in slow subunits of the regulatory element, troponin, and myosin light chain 2 (MLC2) does not follow this general trend of isoform switching in a slower-twitching aged muscle population. On the other hand, increased levels of the mitochondrial enzyme, succinate dehydrogenase, clearly support the metabolic concept of glycolytic-to-oxidative transitions during fibre aging (53). This proteomic finding agrees with the findings from numerous published studies on skeletal muscle aging in both rodent models of sarcopenia and senescent human muscle (46,50,52). Most likely the observed contractile and metabolic transitions are not primary triggering events that render an aged skeletal muscle more susceptible to the loss of tissue mass and contractile strength, but a pathophysiological consequence of age-related abnormalities (54). It is important to stress that histological studies have established that certain aged human skeletal muscles exhibit a considerable reduction in fibre numbers, a drastic decline in fibre size and high levels of atrophying fast-twitching type II contractile fibres (21,27,29). Thus, the overall fast-to-slow transformation and glycolytic-to-oxidative metabolic shift during human skeletal muscle aging, as revealed by proteomics, possibly reflects the preferential loss of fast vs. slower contracting fibres in the senescent organism, and not an age-dependent adaptation due to enhanced muscle plasticity.

In agreement with the concept of age-related fibre type shifting, as exemplified by increased levels of slow contractile proteins and mitochondrial marker enzymes, is the observation of a higher concentration of the muscle-specific CA3 isoform (55) in aged human vastus lateralis muscle. This relatively abundant enzyme has not been previously identified by proteomic surveys of human skeletal muscle aging (38,52). CAs are widely distributed throughout the body and catalyse the reversible hydration of CO2 (66). The various isoforms play a crucial role in the acid-base balance, CO2-removal and provision of CO2 for metabolic processes, such as gluconeogenesis and the urea cycle, as well as the regulatory processes of ion homeostasis (6769). Mammalian skeletal muscles express several CA isoforms in a fiber type-specific manner, whereby the predominant CA3 isoform is mostly present in the cytosolic fraction of type I and IIa fibers (55). Changes in neuromuscular activity patterns, metabolic alterations, stretch-induced hypertrophy and disuse atrophy have profound effects on the expression of muscle CAs (7072). Of note, soluble isoforms of muscle CA are routinely used in clinical applications for assessing fibre damage (73). It has been suggested that the muscle-specific CA3 isoform presents a more sensitive biomarker of muscle damage compared to creatine kinase in neuromuscular disorders (74). Thus, CA is a well-established biomarker routinely used in muscle pathology (75), making it also an ideal candidate for evaluating potential fibre type shifting during muscle aging. The CA3 isoform levels were shown to be increased in senescent human skeletal muscle by both proteomics and independent immunoblot analysis. Although a higher concentration of CA3 may be due to an increased demand for efficient CO2-removal during fibre aging, the altered density of this abundant fibre type marker is more likely indicative of age-related fibre type shifting to slower-contracting muscle populations. In contrast to elevated levels of CA3 in aging human skeletal muscle, proteomic studies have recently established that the same muscle-specific CA isoform is decreased in dystrophic skeletal muscle (76) and non-obese, diabetic skeletal muscle (77). This suggests that the age-dependent increase in CA3 levels may be relatively specific with respect to sarcopenia in old age.

Other altered muscle-associated proteins of interest were Annexin A5, DJ-1 protein, glycogen phosphorylase, Hsp β-7 and Hsp70, creatine kinase, calmodulin and phosphoglucomutase. Increased levels of creatine kinase indicate a compensatory effect on the creatine phosphate shuttle system in aged muscle (78). An altered concentration of Annexin V and calmodulin may be due to altered ion handling during aging (79). Since calmodulin is a critical factor for Ca2+-sequestration and Ca2+-cycling during excitation-contraction coupling (80) and also plays a critical role in skeletal muscle plasticity (81), a changed density of this abundant Ca2+-binding protein presents an excellent new marker of age-related alterations in ion homeostasis. Since many metabolic enzymes involved in glycolysis, gluconeogenesis and glycogen metabolism are multi-functional (82), the alterations in phosphoglucomutase and glycogen phosphorylase during muscle aging are difficult to interpret. Both enzymes are of crucial importance for the bioenergetic utilization of muscle glucose. Phosphoglucomutase catalyses the inter-conversion of glucose-1-phosphate and glucose 6-phosphate, and glycogen phosphorylase facilitates the phosphorolytic cleavage of a glucosyl-residue from the glycogen polymer (83). The decreased levels of glycogen phosphorylase and increased density of phosphoglucomutase indicate altered flux rates of glucose and glycogen metabolism in senescent fibres. It has previously been shown that proteins involved in the cellular stress response are changed in various neuromuscular disorders (84), including sarcopenia in old age (40). The expression levels of various molecular chaperones have been shown to be altered after the fifth decade of life. However, distinct differences appear to exist between rodent models of aging and senescent human muscle (43). Of note, the mostly uncharacterised DJ-1 protein, which has been identified in this study by MS analysis, has previously been shown to play a role in neuro-degeneration (85). This makes this muscle-associated protein a potential new marker of sarcopenia.

In conclusion, aging of the human vastus lateralis muscle is associated with a plethora of proteome-wide changes, especially affecting fibre contraction, ion homeostasis, muscle metabolism and the cellular stress response. These proteomic findings are in agreement with findings from previous histological, physiological and biochemical investigations that have established cycles of denervation and impaired re-innervation causing the loss of entire motor units and muscular atrophy, excitation-contraction uncoupling at the triad junction, altered functioning of the actomyosin apparatus resulting in weaker contractility, impaired regulation of bioenergetic processes, disturbed ion handling and an altered cellular stress response by molecular chaperones. Importantly, the proteomic data presented in this study, are in agreement with altered levels of key metabolic enzymes and contractile elements that suggest a fast-to-slow transition of the contractile apparatus and a glycolytic-to-oxidative shift in metabolism during human muscle aging (54). The establishment of a novel biomarker signature of muscle aging after the fifth decade of life might be exploitable for the future design of a more reliable assay to diagnose sarcopenia of old age.

Acknowledgements

The present study was supported by grants from the Science Foundation Ireland, the Health Research Board and the Higher Education Authority. We thank Helen Kennelly for providing excellent technical support.

Abbreviations:

2D

2-dimensional;

CA

carbonic anhydrase;

DIGE

difference in-gel electrophoresis;

Hsp

heat shock protein;

IEF

isoelectric focusing;

MS

mass spectrometry;

SDS

sodium dodecyl sulfate;

PAGE

polyacrylamide gel electrophoresis;

PBS

phosphate-buffered saline

References

1. 

J VijgJY WeiUnderstanding the biology of aging: the key to prevention and therapyJ Am Geriatr Soc4342643419957706635

2. 

T FlattPS SchmidtIntegrating evolutionary and molecular genetics of agingBiochim Biophys Acta1790951962200910.1016/j.bbagen.2009.07.01019619612

3. 

TB KirkwoodSN AustadWhy do we age?Nature408233238200010.1038/3504168211089980

4. 

TB KirkwoodS MelovOn the programmed/non-programmed nature of ageing within the life historyCurr Biol21R701R707201110.1016/j.cub.2011.07.02021959160

5. 

GS LynchSarcopenia - Age-Related Muscle Wasting and Weakness: Mechanisms and TreatmentsSpringerNew York480201110.1007/978-90-481-9713-2

6. 

LV ThompsonAge-related muscle dysfunctionExp Gerontol44106111200910.1016/j.exger.2008.05.00318657920

7. 

WJ EvansSkeletal muscle loss: cachexia, sarcopenia, and inactivityAm J Clin Nutr911123S1127S201010.3945/ajcn.2010.28608A20164314

8. 

DR ThomasSarcopeniaClin Geriatr Med26331346201010.1016/j.cger.2010.02.012

9. 

MJ BergerTJ DohertySarcopenia: prevalence, mechanisms, and functional consequencesInterdiscip Top Gerontol3794114201010.1159/00031999720703058

10. 

GB ForbesJC ReinaAdult lean body mass declines with age: some longitudinal observationsMetabolism19653663197010.1016/0026-0495(70)90062-45459997

11. 

RN BaumgartnerPM StauberD McHughKM KoehlerPJ GarryCross-sectional age differences in body composition in persons 60+ years of ageJ Gerontol A Biol Sci Med Sci50M307M31619957583802

12. 

RS LindleEJ MetterNA LynchJL FlegJL FozardJ TobinTA RoyBF HurleyAge and gender comparisons of muscle strength in 654 women and men aged 20–93 yrJ Appl Physiol831581158719979375323

13. 

RN BaumgartnerKM KoehlerD GallagherL RomeroSB HeymsfieldRR RossPJ GarryRD LindemanEpidemiology of sarcopenia among the elderly in New MexicoAm J Epidemiol147755763199810.1093/oxfordjournals.aje.a0095209554417

14. 

LJ Melton IIIS KhoslaCS CrowsonMK O’ConnorWM O’FallonBL RiggsEpidemiology of sarcopeniaJ Am Geriatr Soc486256302000

15. 

I JanssenSB HeymsfieldR RossLow relative skeletal muscle mass (sarcopenia) in older persons is associated with functional impairment and physical disabilityJ Am Geriatr Soc50889896200210.1046/j.1532-5415.2002.50216.x12028177

16. 

BH GoodpasterSW ParkTB HarrisSB KritchevskyM NevittAV SchwartzEM SimonsickFA TylavskyM VisserAB NewmanThe loss of skeletal muscle strength, mass, and quality in older adults: the health, aging and body composition studyJ Gerontol A Biol Sci Med Sci6110591064200610.1093/gerona/61.10.105917077199

17. 

MJ DelmonicoTB HarrisM VisserSW ParkMB ConroyP Velasquez-MieyerR BoudreauTM ManiniM NevittAB NewmanBH GoodpasterHealth, aging, and body. Longitudinal study of muscle strength, quality, and adipose tissue infiltrationAm J Clin Nutr9015791585200910.3945/ajcn.2009.2804719864405

18. 

KK HedayatiM DittmarPrevalence of sarcopenia among older community-dwelling people with normal health and nutritional stateEcol Food Nutr49110128201010.1080/0367024090354115421883084

19. 

HP PatelHE SyddallHJ MartinCE StewartC CooperAA SayerHertfordshire sarcopenia study: design and methodsBMC Geriatr1043201010.1186/1471-2318-10-4320587018

20. 

DN ProctorPC O’BrienEJ AtkinsonKS NairComparison of techniques to estimate total body skeletal muscle mass in people of different age groupsAm J Physiol277E489E495199910484361

21. 

AA VandervoortAging of the human neuromuscular systemMuscle Nerve251725200210.1002/mus.121511754180

22. 

WR FronteraKF ReidEM PhillipsLS KrivickasVA HughesR RoubenoffFA FieldingMuscle fiber size and function in elderly humans: a longitudinal studyJ Appl Physiol105637642200810.1152/japplphysiol.90332.200818556434

23. 

JA FaulknerLM LarkinDR ClaflinSV BrooksAge-related changes in the structure and function of skeletal musclesClin Exp Pharmacol Physiol3410911096200710.1111/j.1440-1681.2007.04752.x17880359

24. 

D ScottL BlizzardJ FellG JonesThe epidemiology of sarcopenia in community living older adults: what role does lifestyle play?J Cachexia Sarcopenia Muscle2125134201110.1007/s13539-011-0036-421966639

25. 

RH CokerRR WolfeBedrest and sarcopeniaCurr Opin Clin Nutr Metab Care15711201210.1097/MCO.0b013e32834da629

26. 

P AagaardC SuettaP CaserottiSP MagnussonM KjaerRole of the nervous system in sarcopenia and muscle atrophy with aging: strength training as a countermeasureScand J Med Sci Sports204964201010.1111/j.1600-0838.2009.01084.x20487503

27. 

E EdstromM AltunE BergmanH JohnsonS KullbergV Ramirez-LeonB UlfhakeFactors contributing to neuromuscular impairment and sarcopenia during agingPhysiol Behav92129135200710.1016/j.physbeh.2007.05.04017585972

28. 

I BeyerT MetsI BautmansChronic low-grade inflammation and age-related sarcopeniaCurr Opin Clin Nutr Metab Care151222201210.1097/MCO.0b013e32834dd29722108098

29. 

L LarssonB SjodinJ KarlssonHistochemical and biochemical changes in human skeletal muscle with age in sedentary males, age 22–65 yearsActa Physiol Scand10331391978208350

30. 

M MuscaritoliSD AnkerJ ArgilésZ AversaJM BauerG BioloY BoirieI BosaeusT CederholmP CostelliConsensus definition of sarcopenia, cachexia and pre-cachexia: joint document elaborated by Special Interest Groups (SIG) ‘cachexia-anorexia in chronic wasting diseases’ and ‘nutrition in geriatrics’Clin Nutr29154159201020060626

31. 

AJ Cruz-JentoftJP BaeyensJM BauerY BoirieT CederholmF LandiFC MartinJP MichelY RollandSM SchneiderSarcopenia: European consensus on definition and diagnosis: Report of the European Working Group on Sarcopenia in Older PeopleAge Ageing39412423201010.1093/ageing/afq03420392703

32. 

AJ Cruz-JentoftF LandiE TopinkovaJP MichelUnderstanding sarcopenia as a geriatric syndromeCurr Opin Clin Nutr Metab Care1317201010.1097/MCO.0b013e328333c1c119915458

33. 

H PatelHE SyddallHJ MartinC CooperC StewartAA SayerThe feasibility and acceptability of muscle biopsy in epidemiological studies: findings from the Hertfordshire Sarcopenia Study (HSS)J Nutr Health Aging151015201110.1007/s12603-011-0006-821267515

34. 

J LexellCC TaylorM SjostromWhat is the cause of the ageing atrophy? Total number, size and proportion of different fiber types studied in whole vastus lateralis muscle from 15- to 83-year-old menJ Neurol Sci8427529419883379447

35. 

J LexellHuman aging, muscle mass, and fiber type compositionJ Gerontol A Biol Sci Med Sci5011161995

36. 

C LewisP DoranK OhlendieckProteomic analysis of dystrophic muscleMethods Mol Biol798357369201210.1007/978-1-61779-343-1_2022130847

37. 

K OhlendieckProteomics of skeletal muscle differentiation, neuromuscular disorders and fiber agingExpert Rev Proteomics7283296201010.1586/epr.10.220377394

38. 

C GelfiM VassoP CerretelliDiversity of human skeletal muscle in health and disease: contribution of proteomicsJ Proteomics74774795201110.1016/j.jprot.2011.02.02821414428

39. 

K OhlendieckSkeletal muscle proteomics: current approaches, technical challenges and emerging techniquesSkelet Muscle16201110.1186/2044-5040-1-621798084

40. 

P DoranP DonoghueK O’ConnellJ GannonK OhlendieckProteomics of skeletal muscle agingProteomics99891003200910.1002/pmic.20080036519180535

41. 

I PiecA ListratJ AlliotC ChambonRG TaylorD BechetDifferential proteome analysis of aging in rat skeletal muscleFASEB J1911431145200515831715

42. 

K O’ConnellJ GannonP DoranK OhlendieckProteomic profiling reveals a severely perturbed protein expression pattern in aged skeletal muscleInt J Mol Med20145153200717611631

43. 

D DoranJ GannonK O’ConnellK OhlendieckAging skeletal muscle shows a drastic increase in the small heat shock proteins αB-crystallin/HspB5 and cvHsp/HspB7Eur J Cell Biol86629640200717761354

44. 

J GannonL StauntonK O’ConnellP DoranK OhlendieckPhosphoproteomic analysis of aged skeletal muscleInt J Mol Med2233422008

45. 

K O’ConnellP DoranJ GannonK OhlendieckLectin-based proteomic profiling of aged skeletal muscle: decreased pyruvate kinase isozyme M1 exhibits drastically increased levels of N-glycosylationEur J Cell Biol877938052008

46. 

P DoranK O’ConnellJ GannonM KavanaghK OhlendieckOpposite pathobiochemical fate of pyruvate kinase and adenylate kinase in aged rat skeletal muscle as revealed by proteomic DIGE analysisProteomics8364377200810.1002/pmic.20070047518050275

47. 

D CapitanioM VassoC FaniaM MoriggiA ViganoP ProcacciV MagnaghiC GelfiComparative proteomic profile of rat sciatic nerve and gastrocnemius muscle tissues in ageing by 2-D DIGEProteomics920042020200910.1002/pmic.20070116219333999

48. 

J GannonP DoranA KirwanK OhlendieckDrastic increase of myosin light chain MLC-2 in senescent skeletal muscle indicates fast-to-slow fibre transition in sarcopenia of old ageEur J Cell Biol88685700200910.1016/j.ejcb.2009.06.00419616867

49. 

A LombardiE SilvestriF CioffiR SeneseA LanniF GogliaP de LangeM MorenoDefining the transcriptomic and proteomic profiles of rat ageing skeletal muscle by the use of a cDNA array, 2D- and Blue native-PAGE approachJ Proteomics72708721200910.1016/j.jprot.2009.02.00719268720

50. 

K O’ConnellK OhlendieckProteomic DIGE analysis of the mitochondria-enriched fraction from aged rat skeletal muscleProteomics955095524200919834913

51. 

P DonoghueL StauntonE MullenG ManningK OhlendieckDIGE analysis of rat skeletal muscle proteins using nonionic detergent phase extraction of young adult vs. aged gastrocnemius tissueJ Proteomics714411453201010.1016/j.jprot.2010.01.01420153846

52. 

C GelfiA ViganoM RipamontiA PontoglioS BegumMA PellegrinoB GrassiR BottinelliR WaitP CerretelliThe human muscle proteome in agingJ Proteome Res513441353200610.1021/pr050414x16739986

53. 

L StauntonK O’ConnellK OhlendieckProteomic profiling of mitochondrial enzymes during skeletal muscle agingJ Aging Res2011908035201110.4061/2011/90803521437005

54. 

K OhlendieckProteomic profiling of fast-to-slow muscle transitions during agingFront Physiol2105201122207852

55. 

P FremontPM CharestC CotePA RogersCarbonic anhydrase III in skeletal muscle fibers: an immunocytochemical and biochemical studyJ Histochem Cytochem36775782198810.1177/36.7.31334073133407

56. 

R SchäferU KnaufM ZweyerO HögemeierF de GuarriniX LiuHJ EichhornFW KochRR MundegarI ErzenA WernigAge dependence of the human skeletal muscle stem cell in forming muscle tissueArtif Organs30130140200616480387

57. 

L StauntonH JockuschC WiegandT AlbrechtK OhlendieckIdentification of secondary effects of hyperexcitability by proteomic profiling of myotonic mouse muscleMol Biosyst724802489201110.1039/c1mb05043e21629954

58. 

MM BradfordA rapid and sensitive method for the quantitation of microgram quantities of protein utilizing the principle of protein-dye bindingAnal Biochem72248254197610.1016/0003-2697(76)90527-3942051

59. 

T RabilloudM ChevalletS LucheC LelongTwo-dimensional gel electrophoresis in proteomics: Past, present and futureJ Proteomics7320642077201010.1016/j.jprot.2010.05.01620685252

60. 

L StauntonH JockuschK OhlendieckProteomic analysis of muscle affected by motor neuron degeneration: the wobbler mouse model of amyotrophic lateral sclerosisBiochem Biophys Res Commun406595600201110.1016/j.bbrc.2011.02.09921354103

61. 

C LewisK OhlendieckMass spectrometric identification of dystrophin isoform Dp427 by on-membrane digestion of sarcolemma from skeletal muscleAnal Biochem404197203201010.1016/j.ab.2010.05.01720507823

62. 

A ShevchenkoH TomasJ HavlisJV OlsenM MannIn-gel digestion for mass spectrometric characterization of proteins and proteomesNat Protoc128562860200610.1038/nprot.2006.46817406544

63. 

T LangT StreeperP CawthonK BaldwinDR TaaffeTB HarrisSarcopenia: etiology, clinical consequences, intervention, and assessmentOsteoporos Int21543559201010.1007/s00198-009-1059-y19779761

64. 

LJ TanSL LiuSF LeiCJ PapasianHW DengMolecular genetic studies of gene identification for sarcopeniaHum Genet131131201210.1007/s00439-011-1040-721706341

65. 

SY KhaitlinaFunctional specificity of actin isoformsInt Rev Cytol2023598200110.1016/S0074-7696(01)02003-411061563

66. 

C GeersG GrosCarbon dioxide transport and carbonic anhydrase in blood and musclePhysiol Rev80681715200010747205

67. 

P WetzelG GrosInhibition and kinetic properties of membrane-bound carbonic anhydrases in rabbit skeletal musclesArch Biochem Biophys356151158199810.1006/abbi.1998.07629705205

68. 

P WetzelT KleinkeS PapadopoulosG GrosInhibition of muscle carbonic anhydrase slows the Ca2+ transient in rat skeletal muscle fibersAm J Physiol283C1242C1253200210.1152/ajpcell.00106.200212225987

69. 

P FremontH RiverinJ FrenettePA RogersC CoteFatigue and recovery of rat soleus muscle are influenced by inhibition of an intracellular carbonic anhydrase isoformAm J Physiol260R615R62119912001010

70. 

C BrownsonH IsenbergW BrownS SalmonsY EdwardsChanges in skeletal muscle gene transcription induced by chronic stimulationMuscle Nerve1111831189198810.1002/mus.8801111133147374

71. 

C BrownsonP LoughnaAlterations in the mRNA levels of two metabolic enzymes in rat skeletal muscle during stretch-induced hypertrophy and disuse atrophyPfluegers Arch431990992199610.1007/s0042400500978927521

72. 

CH CoteF AmbrosioG PerreaultMetabolic and contractile influence of carbonic anhydrase III in skeletal muscle is age dependentAm J Physiol276R559R56519999950937

73. 

AH WuMB PerrymanClinical applications of muscle enzymes and proteinsCurr Opin Rheumatol481582019921457275

74. 

HK VäänänenTE TakalaU TolonenJ VuoriVV MyllyläMuscle-specific carbonic anhydrase III is a more sensitive marker of muscle damage than creatine kinase in neuromuscular disordersArch Neurol451254125619883142447

75. 

P BrancaccioG LippiN MaffulliBiochemical markers of muscular damageClin Chem Lab Med48757767201010.1515/CCLM.2010.179

76. 

P DoranG MartinP DowlingH JockuschK OhlendieckProteome analysis of the dystrophin-deficient MDX diaphragm reveals a drastic increase in the heat shock protein cvHSPProteomics646104621200610.1002/pmic.20060008216835851

77. 

E MullenK OhlendieckProteomic profiling of non-obese type 2 diabetic skeletal muscleInt J Mol Med25445458201020127051

78. 

T WallimannM Tokarska-SchlattnerU SchlattnerThe creatine kinase system and pleiotropic effects of creatineAmino Acids4012711296201110.1007/s00726-011-0877-321448658

79. 

M YanezJ Gil-LongoM Campos-ToimilCalcium binding proteinsAdv Exp Med Biol740461482201210.1007/978-94-007-2888-2_19

80. 

W TangS SencerSL HamiltonCalmodulin modulation of proteins involved in excitation-contraction couplingFront Biosci7d1583d1589200210.2741/tang12045019

81. 

P TaviH WesterbladThe role of in vivo Ca²+ signals acting on Ca²+-calmodulin-dependent proteins for skeletal muscle plasticityJ Physiol589502150312011

82. 

K OhlendieckProteomics of skeletal muscle glycolysisBiochim Biophys Acta180420892101201010.1016/j.bbapap.2010.08.00120709194

83. 

TE JensenEA RichterRegulation of glucose and glycogen metabolism during and after exerciseJ Physiol59010691076201210.1113/jphysiol.2011.22497222199166

84. 

RN NishimuraFR SharpHeat shock proteins and neuromuscular diseaseMuscle Nerve32693709200510.1002/mus.2037315962334

85. 

V BonifatiBA OostraP HeutinkLinking DJ-1 to neuro-degeneration offers novel insights for understanding the pathogenesis of Parkinson’s diseaseJ Mol Med82163174200414712351

Related Articles

Journal Cover

October 2012
Volume 30 Issue 4

Print ISSN: 1107-3756
Online ISSN:1791-244X

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Staunton L, Zweyer M, Swandulla D and Ohlendieck K: Mass spectrometry-based proteomic analysis of middle-aged vs. aged vastus lateralis reveals increased levels of carbonic anhydrase isoform 3 in senescent human skeletal muscle. Int J Mol Med 30: 723-733, 2012
APA
Staunton, L., Zweyer, M., Swandulla, D., & Ohlendieck, K. (2012). Mass spectrometry-based proteomic analysis of middle-aged vs. aged vastus lateralis reveals increased levels of carbonic anhydrase isoform 3 in senescent human skeletal muscle. International Journal of Molecular Medicine, 30, 723-733. https://doi.org/10.3892/ijmm.2012.1056
MLA
Staunton, L., Zweyer, M., Swandulla, D., Ohlendieck, K."Mass spectrometry-based proteomic analysis of middle-aged vs. aged vastus lateralis reveals increased levels of carbonic anhydrase isoform 3 in senescent human skeletal muscle". International Journal of Molecular Medicine 30.4 (2012): 723-733.
Chicago
Staunton, L., Zweyer, M., Swandulla, D., Ohlendieck, K."Mass spectrometry-based proteomic analysis of middle-aged vs. aged vastus lateralis reveals increased levels of carbonic anhydrase isoform 3 in senescent human skeletal muscle". International Journal of Molecular Medicine 30, no. 4 (2012): 723-733. https://doi.org/10.3892/ijmm.2012.1056