Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
J Colloid Interface Sci ; 594: 362-371, 2021 Jul 15.
Article in English | MEDLINE | ID: mdl-33774393

ABSTRACT

Microencapsulation helps to improve bioavailability of a functional whey protein, lactoferrin (Lf), in adults. Herein, we report the Lf loading capacity (LC) and retention efficiency (RE) in the microparticles of surface-reacted calcium carbonate (SRCC) of different types and compare them to those of widely used vaterite microparticles. The LCs and REs are analyzed in connection to the total surface area and the volume of intraparticle pores. The best performing SRCC3 demonstrates Lf LC of 11.00 wt% achieved in a single absorption step and 74% RE after two cycles of washing with deionized water. A much larger surface area of SRCC templates and a lower pH required to release Lf do not affect its antitumor activity in MCF-7 assay. Layer-by-Layer assembly of pepsin-tannic acid multilayer shell around Lf-loaded microparticles followed by acidic decomposition of the inorganic core produces microencapsulated Lf with a yield ~36 times higher than from vaterite templates reported earlier, while the scale of encapsulated Lf production is ~12,000 times larger. In vitro digestion tests demonstrate the protection of ~65% of encapsulated Lf from gastric digestion. The developed capsules are prospective candidates for functional foods fortified with Lf.


Subject(s)
Calcium Carbonate , Lactoferrin , Capsules , Lactoferrin/metabolism , Prospective Studies , Tannins
2.
J Endocrinol ; 234(2): 187-200, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28533420

ABSTRACT

Insulin-like growth factors (IGFs) and myostatin have opposing roles in regulating the growth and size of skeletal muscle, with IGF1 stimulating, and myostatin inhibiting, growth. However, it remains unclear whether these proteins have mutually dependent, or independent, roles. To clarify this issue, we crossed myostatin null (Mstn-/-) mice with mice overexpressing Igf1 in skeletal muscle (Igf1+) to generate six genotypes of male mice; wild type (Mstn+/+ ), Mstn+/-, Mstn-/-, Mstn+/+:Igf1+, Mstn+/-:Igf1+ and Mstn-/-:Igf1+ Overexpression of Igf1 increased the mass of mixed fibre type muscles (e.g. Quadriceps femoris) by 19% over Mstn+/+ , 33% over Mstn+/- and 49% over Mstn-/- (P < 0.001). By contrast, the mass of the gonadal fat pad was correspondingly reduced with the removal of Mstn and addition of Igf1 Myostatin regulated the number, while IGF1 regulated the size of myofibres, and the deletion of Mstn and Igf1+ independently increased the proportion of fast type IIB myosin heavy chain isoforms in T. anterior (up to 10% each, P < 0.001). The abundance of AKT and rpS6 was increased in muscles of Mstn-/-mice, while phosphorylation of AKTS473 was increased in Igf1+mice (Mstn+/+:Igf1+, Mstn+/-:Igf1+ and Mstn-/-:Igf1+). Our results demonstrate that a greater than additive effect is observed on the growth of skeletal muscle and in the reduction of body fat when myostatin is absent and IGF1 is in excess. Finally, we show that myostatin and IGF1 regulate skeletal muscle size, myofibre type and gonadal fat through distinct mechanisms that involve increasing the total abundance and phosphorylation status of AKT and rpS6.


Subject(s)
Gene Expression Regulation/physiology , Insulin-Like Growth Factor I/metabolism , Muscle, Skeletal/physiology , Myostatin/metabolism , Adipose Tissue/physiology , Animals , Genotype , Insulin-Like Growth Factor I/genetics , Male , Mice , Mice, Knockout , Mice, Transgenic , Myostatin/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism
3.
PLoS One ; 8(12): e81713, 2013.
Article in English | MEDLINE | ID: mdl-24312578

ABSTRACT

Myostatin plays a fundamental role in regulating the size of skeletal muscles. To date, only a single myostatin gene and no splice variants have been identified in mammals. Here we describe the splicing of a cryptic intron that removes the coding sequence for the receptor binding moiety of sheep myostatin. The deduced polypeptide sequence of the myostatin splice variant (MSV) contains a 256 amino acid N-terminal domain, which is common to myostatin, and a unique C-terminus of 65 amino acids. Western immunoblotting demonstrated that MSV mRNA is translated into protein, which is present in skeletal muscles. To determine the biological role of MSV, we developed an MSV over-expressing C2C12 myoblast line and showed that it proliferated faster than that of the control line in association with an increased abundance of the CDK2/Cyclin E complex in the nucleus. Recombinant protein made for the novel C-terminus of MSV also stimulated myoblast proliferation and bound to myostatin with high affinity as determined by surface plasmon resonance assay. Therefore, we postulated that MSV functions as a binding protein and antagonist of myostatin. Consistent with our postulate, myostatin protein was co-immunoprecipitated from skeletal muscle extracts with an MSV-specific antibody. MSV over-expression in C2C12 myoblasts blocked myostatin-induced Smad2/3-dependent signaling, thereby confirming that MSV antagonizes the canonical myostatin pathway. Furthermore, MSV over-expression increased the abundance of MyoD, Myogenin and MRF4 proteins (P<0.05), which indicates that MSV stimulates myogenesis through the induction of myogenic regulatory factors. To help elucidate a possible role in vivo, we observed that MSV protein was more abundant during early post-natal muscle development, while myostatin remained unchanged, which suggests that MSV may promote the growth of skeletal muscles. We conclude that MSV represents a unique example of intra-genic regulation in which a splice variant directly antagonizes the biological activity of the canonical gene product.


Subject(s)
Alternative Splicing , Muscle Development , Myostatin/genetics , Myostatin/metabolism , Amino Acid Sequence , Animals , Cattle , Cell Line , Gene Expression Regulation, Developmental , Male , Molecular Sequence Data , Muscle Development/genetics , Muscle, Skeletal/cytology , Muscle, Skeletal/growth & development , Muscle, Skeletal/metabolism , Myogenic Regulatory Factors/metabolism , Myostatin/chemistry , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Isoforms/metabolism , Sheep , Signal Transduction , Smad2 Protein/metabolism , Smad3 Protein/metabolism
4.
Am J Physiol Cell Physiol ; 296(3): C525-34, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19129464

ABSTRACT

Myostatin (Mstn) is a secreted growth factor belonging to the tranforming growth factor (TGF)-beta superfamily. Inactivation of murine Mstn by gene targeting, or natural mutation of bovine or human Mstn, induces the double muscling (DM) phenotype. In DM cattle, Mstn deficiency increases fast glycolytic (type IIB) fiber formation in the biceps femoris (BF) muscle. Using Mstn null ((-/-)) mice, we suggest a possible mechanism behind Mstn-mediated fiber-type diversity. Histological analysis revealed increased type IIB fibers with a concomitant decrease in type IIA and type I fibers in the Mstn(-/-) tibialis anterior and BF muscle. Functional electrical stimulation of Mstn(-/-) BF revealed increased fatigue susceptibility, supporting increased type IIB fiber content. Given the role of myocyte enhancer factor 2 (MEF2) in oxidative type I fiber formation, MEF2 levels in Mstn(-/-) tissue were quantified. Results revealed reduced MEF2C protein in Mstn(-/-) muscle and myoblast nuclear extracts. Reduced MEF2-DNA complex was also observed in electrophoretic mobility-shift assay using Mstn(-/-) nuclear extracts. Furthermore, reduced expression of MEF2 downstream target genes MLC1F and calcineurin were found in Mstn(-/-) muscle. Conversely, Mstn addition was sufficient to directly upregulate MLC promoter-enhancer activity in cultured myoblasts. Since high MyoD levels are seen in fast fibers, we analyzed MyoD levels in the muscle. In contrast to MEF2C, MyoD levels were increased in Mstn(-/-) muscle. Together, these results suggest that while Mstn positively regulates MEF2C levels, it negatively regulates MyoD expression in muscle. We propose that Mstn could regulate fiber-type composition by regulating the expression of MEF2C and MyoD during myogenesis.


Subject(s)
Gene Expression Regulation , Muscle Fibers, Fast-Twitch/metabolism , Muscle Fibers, Slow-Twitch/metabolism , MyoD Protein/genetics , Myogenic Regulatory Factors/genetics , Myostatin/metabolism , Animals , Calcineurin/genetics , Cattle , Cell Line , Electric Stimulation , MEF2 Transcription Factors , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle Contraction , Muscle Fatigue , MyoD Protein/metabolism , Myogenic Regulatory Factors/metabolism , Myosin Heavy Chains/genetics , Myosin Light Chains/genetics , Myostatin/deficiency , Myostatin/genetics , Promoter Regions, Genetic , RNA, Messenger/metabolism , Time Factors , Transcription, Genetic
5.
Exp Cell Res ; 314(5): 1013-29, 2008 Mar 10.
Article in English | MEDLINE | ID: mdl-18255059

ABSTRACT

Genetic analysis has revealed an important function in myogenesis for Myostatin, a member of the TGF-beta superfamily. However, the cascade of genes that responds to Myostatin signalling to regulate myogenesis is not well understood. Thus, a suppressive subtraction hybridization to identify such genes was undertaken and here we report the cloning and characterization of a novel gene, Mighty. Mighty is expressed in a variety of different tissues but appears to be specifically regulated by Myostatin in skeletal muscle. Overexpression of Mighty in C2C12 cells results in early withdrawal of myoblasts from the cell cycle, enhanced and accelerated differentiation and hypertrophy of myotubes. Most importantly, Mighty overexpression leads to increased and earlier expression of MyoD and increased secretion of another known differentiation inducing factor, IGF-II. Furthermore, viral expression of Mighty in mdx mice resulted in an increase in the number of larger healthy muscle fibers. Given its role in myogenesis, we propose that Mighty is a critical promyogenic factor which plays a key role in the signalling pathway downstream of Myostatin.


Subject(s)
Insulin-Like Growth Factor II/metabolism , Muscle Development , Muscle Proteins/physiology , Muscle, Skeletal/growth & development , MyoD Protein/genetics , Myogenic Regulatory Factors/physiology , Animals , Cell Differentiation , Cloning, Molecular , Gene Expression Regulation , Mice , Muscle Fibers, Skeletal , Muscle Proteins/genetics , Myoblasts, Skeletal/cytology , Myogenic Regulatory Factors/genetics , Myostatin , Transforming Growth Factor beta/physiology
6.
Mol Ther ; 15(8): 1463-70, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17551508

ABSTRACT

A reduction in muscle mass and strength is often observed with aging, and this phenomenon is known as sarcopenia. This age-related atrophy frequently correlates with insufficient levels of muscle regeneration resulting from impairment of satellite cell involvement and myogenesis brought about by the aged environment. Using myostatin-null mice, we recently showed that negative regulators of muscle mass such as myostatin play an active role in the regulation of myogenesis during aging. The present study specifically tests the therapeutic value of a myostatin antagonist in sarcopenia. We report here that a short-term blockade of myostatin, through stage-specific administration of a myostatin antagonist, significantly enhanced muscle regeneration in aged mice after injury and during sarcopenia. Antagonism of myostatin led to satellite cell activation, increased Pax7 and MyoD protein levels, and greater myoblast and macrophage cell migration, resulting in enhanced muscle regeneration after notexin injury in aged mice. In addition, the antagonist demonstrated a high degree of efficacy, as only minimal doses during the critical period of regeneration after injury were sufficient to restore the myogenic and inflammatory responses in the aged environment. Thus, we propose that the antagonism of myostatin has significant therapeutic potential in the alleviation of sarcopenia.


Subject(s)
Aging/physiology , Muscles/physiology , Regeneration/physiology , Transforming Growth Factor beta/antagonists & inhibitors , Animals , Cell Movement , Cell Proliferation , Macrophages/cytology , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Muscles/injuries , MyoD Protein/genetics , MyoD Protein/metabolism , Myoblasts/cytology , Myoblasts/metabolism , Myostatin , PAX7 Transcription Factor/genetics , PAX7 Transcription Factor/metabolism , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism
7.
J Cell Physiol ; 206(1): 255-63, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16110473

ABSTRACT

Myostatin is a transforming growth factor-beta (TGF-beta) superfamily member and a key negative regulator of embryonic and postnatal muscle growth. In order to identify downstream target genes regulated by Myostatin, we performed suppressive subtraction hybridization (SSH) on cDNA generated from the biceps femoris muscle of wild-type and myostatin-null mice. Sequence analysis identified several known and unknown genes as Myostatin downstream target genes. Here, we have investigated the regulation of gene expression of an androgen receptor (AR) binding co-factor, androgen receptor associated protein-70 (ARA70), by Myostatin. We show that in mouse there are two isoforms of ARA70 with high homology (79%) to human ARA70; an alpha-isoform which is a canonical ARA70 and a beta-isoform which has a 9 consecutive amino acid deletion and 6 amino acid substitutions in the carboxyl-terminal portion. Reverse Northern analysis on the differentially expressed cDNA library indicated that there is increased expression of ARA70 in the muscles of myostatin-null mice. In addition, Northern blot, together with semi-quantitative PCR analysis, confirmed that there is increased expression of ARA70 in myostatin-null biceps femoris muscle when compared to wild-type muscle. In corroboration of these results, addition of exogenous Myostatin results in down-regulation of ARA70 expression confirming that Myostatin is a negative regulator of ARA70 gene expression. Expression analysis further confirmed that ARA70 is up-regulated during myogenesis and that peak expression of ARA70 is observed following the peak expression of MyoD in differentiating myoblasts. Given that lack of Myostatin and increased expression of AR leads to hypertrophy, we propose that absence of Myostatin, at least in part, induces the hypertrophy phenotype by increasing the activity of AR by up-regulating the expression of ARA70, a known stimulating co-factor of AR.


Subject(s)
Gene Expression Regulation , Oncogene Proteins/metabolism , Transcription Factors/metabolism , Transforming Growth Factor beta/metabolism , Amino Acid Sequence , Animals , Cells, Cultured , Humans , In Situ Hybridization , Mice , Mice, Knockout , Molecular Sequence Data , Muscle, Skeletal/cytology , Muscle, Skeletal/metabolism , Myoblasts/cytology , Myoblasts/metabolism , Myostatin , Nuclear Receptor Coactivators , Nucleic Acid Hybridization , Oncogene Proteins/chemistry , Oncogene Proteins/genetics , Protein Isoforms/genetics , Protein Isoforms/metabolism , Sequence Alignment , Sequence Homology, Amino Acid , Tissue Distribution , Transcription Factors/chemistry , Transcription Factors/genetics , Transforming Growth Factor beta/genetics , Two-Hybrid System Techniques
8.
Biochem Biophys Res Commun ; 340(2): 675-80, 2006 Feb 10.
Article in English | MEDLINE | ID: mdl-16380093

ABSTRACT

Myostatin, a member of TGF-beta superfamily of growth factors, acts as a negative regulator of skeletal muscle mass. The mechanism whereby myostatin controls the proliferation and differentiation of myogenic cells is mostly clarified. However, the regulation of myostatin activity to myogenic cells after its secretion in the extracellular matrix (ECM) is still unknown. Decorin, a small leucine-rich proteoglycan, binds TGF-beta and regulates its activity in the ECM. Thus, we hypothesized that decorin could also bind to myostatin and participate in modulation of its activity to myogenic cells. In order to test the hypothesis, we investigated the interaction between myostatin and decorin by surface plasmon assay. Decorin interacted with mature myostatin in the presence of concentrations of Zn(2+) greater than 10microM, but not in the absence of Zn(2+). Kinetic analysis with a 1:1 binding model resulted in dissociation constants (K(D)) of 2.02x10(-8)M and 9.36x10(-9)M for decorin and the core protein of decorin, respectively. Removal of the glycosaminoglycan chain by chondroitinase ABC digestion did not affect binding, suggesting that decorin could bind to myostatin with its core protein. Furthermore, we demonstrated that immobilized decorin could rescue the inhibitory effect of myostatin on myoblast proliferation in vitro. These results suggest that decorin could trap myostatin and modulate its activity to myogenic cells in the ECM.


Subject(s)
Extracellular Matrix Proteins/metabolism , Myoblasts/metabolism , Proteoglycans/metabolism , Transforming Growth Factor beta/metabolism , Animals , Cattle , Cell Line , Cell Proliferation , Decorin , Extracellular Matrix Proteins/physiology , Mice , Myoblasts/cytology , Myostatin , Protein Binding , Proteoglycans/physiology , Surface Plasmon Resonance , Zinc
9.
Am J Physiol Cell Physiol ; 283(1): C135-41, 2002 Jul.
Article in English | MEDLINE | ID: mdl-12055081

ABSTRACT

Myostatin, a member of the transforming growth factor-beta superfamily, is a secreted growth factor that is proteolytically processed to give COOH-terminal mature myostatin and NH2-terminal latency-associated peptide in myoblasts. Piedmontese cattle are a heavy-muscled breed that express a mutated form of myostatin in which cysteine (313) is substituted with tyrosine. Here we have characterized the biology of this mutated Piedmontese myostatin. Northern and Western analyses indicate that there is increased expression of myostatin mRNA and precursor myostatin protein in the skeletal muscle of Piedmontese cattle. In contrast, a decrease in mature myostatin was observed in Piedmontese skeletal muscle. However, there is no detectable change in the circulatory levels of mature myostatin in Piedmontese cattle. Myoblast proliferation assay performed with normal and Piedmontese myostatin indicated that mature wild-type myostatin protein inhibited the proliferation of C2C12 myoblasts. Piedmontese myostatin, by contrast, failed to inhibit myoblast proliferation. In addition, when added in molar excess, Piedmontese myostatin acted as a potent "competitive inhibitor" molecule. These results indicate that, in Piedmontese myostatin, substitution of cysteine with tyrosine results in the distortion of the "cystine knot" structure and a loss of biological activity of the myostatin. This mutation also appears to affect either processing or stability of mature myostatin without altering the secretion of myostatin.


Subject(s)
Growth Inhibitors/physiology , Mutation/physiology , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/physiology , Amino Acid Substitution , Animals , Cattle , Cell Division/drug effects , Cell Line , Cysteine , Muscles/cytology , Myostatin , RNA Stability , RNA, Messenger/metabolism , Reference Values , Transforming Growth Factor beta/isolation & purification , Transforming Growth Factor beta/pharmacology , Tyrosine
SELECTION OF CITATIONS
SEARCH DETAIL
...