Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
Medicina (Kaunas) ; 60(9)2024 Aug 26.
Article in English | MEDLINE | ID: mdl-39336440

ABSTRACT

Background and Objectives: Urinary incontinence (UI) is the involuntary loss of urine caused by a weakness in the pelvic floor muscles (PFMs) that affects urethral closure. Myostatin, which prevents the growth of muscles, is a protein expressed by human skeletal muscle cells. Indeed, it has been observed that myostatin concentration rises during skeletal muscle inactivity and that suppressing serum myostatin promotes muscle growth and strength. Furthermore, therapeutic interventions that reduce myostatin signalling may lessen the effects of aging on skeletal muscle mass and function. For this reason, the aim of the study was to assess if flat magnetic stimulation technology affects serum myostatin levels, as myostatin can block cell proliferation at the urethral sphincter level. Materials and Methods: A total of 19 women, 75% presenting stress urinary incontinence (SUI) and 25% urgency urinary incontinence (UUI), were enrolled. A non-invasive electromagnetic therapeutic system designed for deep pelvic floor area stimulation was used for eight sessions. Results: The ELISA (enzyme linked immunosorbent assay) test indicated that the myostatin levels in blood sera had significantly decreased. Patients' ultrasound measurements showed a significant genital hiatus length reduction at rest and in a stress condition. The Pelvic Floor Bother Questionnaire consistently revealed a decrease in mean scores when comparing the pre- and post-treatment data. Conclusions: Effective flat magnetic stimulation reduces myostatin concentration and genital hiatus length, minimizing the severity of urinary incontinence. The results of the study show that without causing any discomfort or unfavourable side effects, the treatment plan significantly improved the PFM tone and strength in patients with UI.


Subject(s)
Myostatin , Pelvic Floor , Humans , Female , Myostatin/blood , Myostatin/analysis , Middle Aged , Magnetic Field Therapy/methods , Aged , Urinary Incontinence/therapy , Urinary Incontinence, Stress/therapy , Adult , Surveys and Questionnaires , Enzyme-Linked Immunosorbent Assay/methods
2.
Rheumatology (Oxford) ; 59(9): 2491-2501, 2020 09 01.
Article in English | MEDLINE | ID: mdl-31990347

ABSTRACT

OBJECTIVES: The aim of this study was to investigate the systemic and skeletal muscle levels of atrophy-associated myokines in patients with idiopathic inflammatory myopathies (IIM) and their association with clinical characteristics of myositis. METHODS: A total of 94 IIM patients and 162 healthy controls were recruited. Of those, 20 IIM patients and 28 healthy controls underwent a muscle biopsy. Circulating concentrations of myostatin, follistatin, activin A and TGF-ß1 were assessed by ELISA. The expression of myokines and associated genes involved in the myostatin signalling pathway in muscle tissue was determined by real-time PCR. RESULTS: We report decreased levels of circulating myostatin (median 1817 vs 2659 pg/ml; P = 0.003) and increased follistatin (1319 vs 1055 pg/ml; P = 0.028) in IIM compared with healthy controls. Activin A levels were also higher in IIM (414 vs 309 pg/ml; P = 0.0005) compared with controls. Myostatin was negatively correlated to muscle disease activity assessed by physician on visual analogue scale (MDA) (r = -0.289, P = 0.015) and positively to manual muscle testing of eight muscles (r = 0.366, P = 0.002). On the other hand, follistatin correlated positively with MDA (r = 0.235, P = 0.047). Gene expression analysis showed higher follistatin (P = 0.003) and myostatin inhibitor follistatin-like 3 protein (FSTL3) (P = 0.008) and lower expression of activin receptor type 1B (ALK4) (P = 0.034), signal transducer SMAD3 (P = 0.023) and atrophy marker atrogin-1 (P = 0.0009) in IIM muscle tissue compared with controls. CONCLUSION: This study shows lower myostatin and higher follistatin levels in circulation and attenuated expression of myostatin pathway signalling components in skeletal muscle of patients with myositis, a newly emerging pattern of the activin A-myostatin-follistatin system in muscle wasting diseases.


Subject(s)
Follistatin/analysis , Muscle, Skeletal , Muscular Atrophy , Myositis , Myostatin/analysis , Activin Receptors, Type I/genetics , Correlation of Data , Female , Follistatin-Related Proteins/genetics , Gene Expression Profiling , Humans , Male , Middle Aged , Muscle Proteins/genetics , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscular Atrophy/metabolism , Muscular Atrophy/pathology , Myositis/blood , Myositis/diagnosis , Myositis/etiology , Myositis/physiopathology , Patient Acuity , Physical Examination/methods , SKP Cullin F-Box Protein Ligases/genetics , Signal Transduction , Smad3 Protein/genetics
3.
Biochemistry ; 58(46): 4632-4640, 2019 11 19.
Article in English | MEDLINE | ID: mdl-31638376

ABSTRACT

The mature forms of the TGF-ß family members GDF-11 and GDF-8 are highly similar 25 kDa homodimers with 90% amino acid sequence identity and 99% similarity. Cross-reactivity of GDF-11 and GDF-8 binding reagents is common, making it difficult to attribute distinct roles of these two proteins in biology. We report the selection of GDF-11 and GDF-8 specific SOMAmer (Slow Off-rate Modified Aptamer) reagents aided by a combination of positive selection for one protein coupled with counter-selection against the other. We identified GDF-11 specific SOMAmer reagents from four modified DNA libraries that showed a high affinity (Kd range 0.05-1.2 nM) for GDF-11 but did not bind to GDF-8 (Kd > 1 µM). Conversely, we identified one SOMAmer reagent for GDF-8 from one of the modified libraries that demonstrated excellent affinity (Kd = 0.23 nM) and specificity. In contrast, standard protocols that utilized only positive selection produced binding reagents with similar affinity for both proteins. High affinity and specificity were efficiently encoded in minimal sequences of 21 nucleotides for GDF-11 and 24 nucleotides for GDF-8. Further characterization in pull-down, competition, sandwich-binding, and kinetic studies revealed robust binding under a wide range of buffer and assay conditions. For highly similar proteins like GDF-11 and GDF-8, our method of selection coupled with counter-selection was essential for identification of high-affinity, specific reagents that have the potential to elucidate the fundamental distinction of these growth factors in biology.


Subject(s)
Aptamers, Nucleotide/chemistry , Bone Morphogenetic Proteins/analysis , Growth Differentiation Factors/analysis , Myostatin/analysis , Amino Acid Sequence , Base Sequence , Binding Sites , Epitopes/analysis , Humans , Indicators and Reagents , Recombinant Proteins/analysis , SELEX Aptamer Technique
4.
Dev Dyn ; 247(12): 1241-1252, 2018 12.
Article in English | MEDLINE | ID: mdl-30325085

ABSTRACT

BACKGROUND: Myostatin (MSTN), a member of the transforming growth factor-ß (TGF-ß) superfamily, has been implicated in the negative regulation of skeletal myogenesis. However, the molecular mechanism through which MSTN regulates early embryonic myogenesis is not well understood. RESULTS: We demonstrate that MSTN regulates early embryonic myogenesis by promoting the epithelial-to-mesenchymal transition (EMT) of the dermomyotome during somitogenesis in chicks. We show that the MSTN gene is first expressed at the center of the dermomyotome. As somitogenesis progresses, its expression extends dorsally and ventrally along the plane of the dermomyotome. By combining in situ hybridization and immunofluorescence assays, we demonstrate that the expression pattern of MSTN is spatiotemporally well correlated with EMT of the dermomyotome. Our gain- and loss-of-function experiments further reveal that MSTN can induce EMT of the chick dermomyotome. We also show that MSTN induces EMT of a nonsmall cell lung carcinoma cell line (A549) and Madin-Darby canine kidney cells in vitro. CONCLUSIONS: Our experimental data suggest that MSTN regulates myogenesis by promoting EMT during somitogenesis. These findings provide novel insights into the functions of MSTN during early embryonic myogenesis. Developmental Dynamics 247:1241-1252, 2018. © 2018 Wiley Periodicals, Inc.


Subject(s)
Epithelial-Mesenchymal Transition/drug effects , Muscle Development/drug effects , Myostatin/analysis , Myostatin/pharmacology , Somites/growth & development , A549 Cells , Animals , Carcinoma, Non-Small-Cell Lung/pathology , Chick Embryo , Dogs , Embryonic Development , Epithelium/growth & development , Humans , Madin Darby Canine Kidney Cells , Myostatin/genetics , Somites/embryology
5.
J Clin Lab Anal ; 31(5)2017 Sep.
Article in English | MEDLINE | ID: mdl-27878995

ABSTRACT

OBJECTIVE: Myostatin, a member of the transforming growth factor-ß family, contributes to joint deterioration in mice. Thus, we aimed to assess the correlation of myostatin concentrations with the presence and severity of knee osteoarthritis (OA). MATERIAL AND METHODS: We determined serum and synovial fluid (SF) myostatin concentrations in a population of 184 patients with knee OA and 109 healthy controls. RESULTS: The knee OA group presented with higher serum myostatin concentrations than the controls. Knee OA patients with KL grade 4 showed higher serum and SF myostatin concentrations compared with those with KL grade 2 and 3. Knee OA patients with KL grade 3 had higher serum and SF myostatin concentrations compared with those with KL grade 2. Serum and SF myostatin concentrations were significantly correlated with KL grading. CONCLUSION: Serum and SF myostatin concentrations were correlated with the presence and severity of knee OA.


Subject(s)
Myostatin/analysis , Osteoarthritis, Knee , Aged , Biomarkers/analysis , Biomarkers/blood , Biomarkers/chemistry , Case-Control Studies , Female , Humans , Male , Middle Aged , Myostatin/blood , Myostatin/chemistry , Osteoarthritis, Knee/blood , Osteoarthritis, Knee/diagnosis , Osteoarthritis, Knee/epidemiology , Osteoarthritis, Knee/metabolism , Severity of Illness Index , Synovial Fluid/chemistry
6.
Anim Biotechnol ; 25(4): 223-33, 2014.
Article in English | MEDLINE | ID: mdl-24813217

ABSTRACT

The purpose of the present study was to compare mRNA levels of myostatin (MSTN), myogenin (MyoG), and fiber type compositions in terms of myosin heavy chain (MyHC) in skeletal muscles of two rabbit breeds with different body sizes and growth rates. Longissimus dorsi and biceps femoris muscles of 16 Californian rabbits (CW) and 16 Germany great line of ZIKA rabbits (GZ) were collected at the ages of 35d and 84d (slaughter age). The results showed that the live weights of GZ rabbits of 35d and 84d old were approximately 36% and 26% greater than those of CW rabbits, respectively. Quantitative real-time PCR analysis revealed that at the age of 84d GZ rabbits contained significantly lower MSTN mRNA level and higher MyoG mRNA level in both longissimus dorsi and biceps femoris muscles than CW rabbits, and mRNA levels of MSTN and MyoG exhibited opposite changes from the age of 35d to 84d, suggesting that GZ rabbits were subjected to less growth inhibition from MSTN at slaughter age, which occurred most possibly in skeletal muscles. Four types of fiber were identified by real-time PCR in rabbit muscles, with MyHC-1 and MyHC-2D, MyHC-2B were the major types in biceps femoris and longissimus dorsi muscles, respectively. At the age of 84d, GZ rabbits contained greater proportion of MyHC-1 and decreased proportion of MyHC-2D and decreased lactate dehydrogenase activity in biceps femoris than CW rabbits, and the results were exactly opposite in longissimus dorsi, suggesting that GZ rabbits show higher oxidative capacity in biceps femoris muscle than CW rabbits. In conclusion, the trends of mRNA levels of MSTN and fiber types in GZ rabbits' skeletal muscles might be consistent with the putative fast growth characteristic of GZ rabbits compared to CW rabbits.


Subject(s)
Muscle, Skeletal/growth & development , Muscle, Skeletal/metabolism , Myogenin/analysis , Myosin Heavy Chains/analysis , Myostatin/analysis , Rabbits/growth & development , Animals , Female , Gene Expression , Livestock , Male , Muscle, Skeletal/chemistry , Myogenin/genetics , Myogenin/metabolism , Myosin Heavy Chains/genetics , Myosin Heavy Chains/metabolism , Myostatin/genetics , Myostatin/metabolism , Rabbits/genetics , Rabbits/metabolism , Species Specificity
7.
J Oral Maxillofac Surg ; 70(2): 440-8, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21821327

ABSTRACT

PURPOSE: We identified masseter muscle fiber type property differences in subjects with dentofacial deformities. PATIENTS AND METHODS: Samples of masseter muscle were collected from 139 young adults during mandibular osteotomy procedures to assess mean fiber areas and percent tissue occupancies for the 4 fiber types that comprise the muscle. Subjects were classified into 1 of 6 malocclusion groups based on the presence of a skeletal Class II or III sagittal dimension malocclusion and either a skeletal open, deep, or normal bite vertical dimension malocclusion. In a subpopulation, relative quantities of the muscle growth factors IGF-I and GDF-8 gene expression were quantified by real-time polymerase chain reaction. RESULTS: Fiber properties were not different in the sagittal malocclusion groups, but were very different in the vertical malocclusion groups (P ≤ .0004). There were significant mean fiber area differences for type II (P ≤ .0004) and type neonatal-atrial (P = .001) fiber types and for fiber percent occupancy differences for both type I-II hybrid fibers and type II fibers (P ≤ .0004). Growth factor expression differed by gender for IGF-I (P = .02) and GDF-8 (P < .01). The ratio of IGF-I:GDF-8 expression associates with type I and II mean fiber areas. CONCLUSION: Fiber type properties are very closely associated with variations in vertical growth of the face, with statistical significance for overall comparisons at P ≤ .0004. An increase in masseter muscle type II fiber mean fiber areas and percent tissue occupancies is inversely related to increases in vertical facial dimension.


Subject(s)
Insulin-Like Growth Factor I/analysis , Malocclusion, Angle Class III/pathology , Malocclusion, Angle Class II/pathology , Masseter Muscle/ultrastructure , Muscle Fibers, Skeletal/ultrastructure , Myostatin/analysis , Adolescent , Adult , Cardiac Myosins/analysis , Female , Humans , Insulin-Like Growth Factor I/genetics , Male , Maxillofacial Development/physiology , Muscle Fibers, Fast-Twitch/ultrastructure , Muscle Fibers, Slow-Twitch/ultrastructure , Myosin Type I/analysis , Myosin Type II/analysis , Myostatin/genetics , Open Bite/pathology , Overbite/pathology , Polymerase Chain Reaction , RNA/analysis , Sex Factors , Vertical Dimension , Young Adult
8.
J Exp Biol ; 214(Pt 15): 2522-7, 2011 Aug 01.
Article in English | MEDLINE | ID: mdl-21753045

ABSTRACT

Myostatin, a negative regulator of muscle mass, is elevated during disuse and starvation. Mammalian hibernation presents a unique scenario, where animals are hypocaloric and in torpor, but the extent of muscle protein loss is minimized. We hypothesized that myostatin expression, which is usually increased early in disuse and under hypocaloric conditions, could be suppressed in this unique model. Skeletal muscle was collected from thirteen-lined ground squirrels, Spermophilus tridecemlineatus, at six time points during hibernation: control euthermic (CON); entrance into hibernation (ENT), body temperature (T(b)) falling; early hibernation (EHib), stable T(b) in torpor for 24 h; late hibernation (LHib), stable T(b) in torpor for 3 days; early arousal (EAr), T(b) rising; and arousal (AR), T(b) restored to 34-37°C for about 18 h. There was no significant increase of myostatin during ENT, EHib or LHib. Unexpectedly, there were approximately sixfold increases in myostatin protein levels as squirrels arose from torpor. The elevation during EAr remained high in AR, which represented an interbout time period. Mechanisms that could release the suppression or promote increased levels of myostatin were assessed. SMAD2 and phosphorylated SMAD2 were increased during EHib, but only the phosphorylated SMAD2 during AR mirrored increases in myostatin. Follistatin, a negative regulator of myostatin, did not follow the same time course as myostatin or its signaling pathway, indicating more control of myostatin at the signaling level. However, SMAD7, an inhibitory SMAD, did not appear to play a significant role during deep hibernation. Hibernation is an excellent natural model to study factors involved in the endogenous intracellular mechanisms controlling myostatin.


Subject(s)
Hibernation , Muscle, Skeletal/metabolism , Myostatin/metabolism , Sciuridae/physiology , Smad Proteins/metabolism , Animals , Follistatin/analysis , Follistatin/metabolism , Male , Models, Animal , Myostatin/analysis , Phosphorylation , Signal Transduction , Smad Proteins/analysis
9.
Nutrients ; 12(8)2020 Aug 11.
Article in English | MEDLINE | ID: mdl-32796600

ABSTRACT

Sarcopenia is a geriatric syndrome with a significant impact on older patients' quality of life, morbidity and mortality. Despite the new available criteria, its early diagnosis remains difficult, highlighting the necessity of looking for a valid muscle wasting biomarker. Myostatin, a muscle mass negative regulator, is one of the potential candidates. The aim of this work is to point out various factors affecting the potential of myostatin as a biomarker of muscle wasting. Based on the literature review, we can say that recent studies produced conflicting results and revealed a number of potential confounding factors influencing their use in sarcopenia diagnosing. These factors include physiological variables (such as age, sex and physical activity) as well as a variety of disorders (including heart failure, metabolic syndrome, kidney failure and inflammatory diseases) and differences in laboratory measurement methodology. Our conclusion is that although myostatin alone might not prove to be a feasible biomarker, it could become an important part of a recently proposed panel of muscle wasting biomarkers. However, a thorough understanding of the interrelationship of these markers, as well as establishing a valid measurement methodology for myostatin and revising current research data in the light of new criteria of sarcopenia, is needed.


Subject(s)
Geriatric Assessment , Myostatin/analysis , Nutrition Assessment , Sarcopenia/diagnosis , Wasting Syndrome/diagnosis , Aged , Biomarkers/analysis , Female , Humans , Male , Muscle, Skeletal/metabolism
10.
Adv Protein Chem Struct Biol ; 108: 227-256, 2017.
Article in English | MEDLINE | ID: mdl-28427562

ABSTRACT

Almost 30 years ago, the protein, atrial natriuretic peptide, was identified as a heart-secreted hormone that provides a peripheral signal from the myocardium that communicates to the rest of the organism to modify blood pressure and volume under conditions of heart failure. Since then, additional peripheral factors secreted by the heart, termed cardiokines, have been identified and shown to coordinate this interorgan cross talk. In addition to this interorgan communication, cardiokines also act in an autocrine/paracrine manner to play a role in intercellular communication within the myocardium. This review focuses on the roles of newly emerging cardiokines that are mainly increased in stress-induced cardiac diseases. The potential of these cardiokines as clinical biomarkers for diagnosis and prognosis of cardiac disorders is also discussed.


Subject(s)
Heart Diseases/immunology , Inflammation/immunology , Myocardium/immunology , Activins/analysis , Activins/immunology , Animals , Biomarkers/analysis , Fibroblast Growth Factors/analysis , Fibroblast Growth Factors/immunology , Follistatin/analysis , Follistatin/immunology , Follistatin-Related Proteins/analysis , Follistatin-Related Proteins/immunology , Growth Differentiation Factor 15/analysis , Growth Differentiation Factor 15/immunology , Heart Diseases/complications , Heart Diseases/pathology , Humans , Inflammation/complications , Inflammation/pathology , Interleukin-33/analysis , Interleukin-33/immunology , Myocardium/pathology , Myostatin/analysis , Myostatin/immunology , Paracrine Communication , Stress, Physiological , Transforming Growth Factor beta/analysis , Transforming Growth Factor beta/immunology
11.
Bioanalysis ; 8(3): 193-204, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26811930

ABSTRACT

BACKGROUND: Therapeutic protein discovery study highlights the need for the development of quantitative bioanalytical methods for determining the levels of both the therapeutic protein and the target protein, as well. RESULTS: For the quantitation of BMS-986089, both accuracy (99-103%) and precision (2.4-12%) were obtained for the analysis of the surrogate peptide (ITYGGNSPVQEFTVPGR), in addition to the accuracy (100-108%) and precision (0.7-18%) that were obtained for the analysis of the surrogate peptide (VVSVLTVLHQDWLNGK). For Myostatin, accuracy (94-103%) and precision (2.4-14.9%) were obtained for the analysis of the surrogate peptide (IPAMVVDR). CONCLUSION: The developed method was applied to the analysis of samples following dosing of BMS-986089 to mice. This method highlights the potential of LC-MS/MS-based methods to eventually assess in vivo drug-target engagement.


Subject(s)
Chromatography, Liquid/methods , Myostatin/analysis , Recombinant Fusion Proteins/analysis , Tandem Mass Spectrometry/methods , Amino Acid Sequence , Animals , Chromatography, Liquid/economics , Cost-Benefit Analysis , Humans , Immunoglobulin G/analysis , Immunoglobulin G/metabolism , Immunoglobulin G/pharmacology , Immunoglobulin G/therapeutic use , Male , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Myostatin/metabolism , Peptide Fragments/analysis , Peptide Fragments/chemistry , Rats , Recombinant Fusion Proteins/pharmacokinetics , Recombinant Fusion Proteins/pharmacology , Recombinant Fusion Proteins/therapeutic use , Tandem Mass Spectrometry/economics , Trypsin/metabolism
12.
Biotechnol Prog ; 31(2): 452-9, 2015.
Article in English | MEDLINE | ID: mdl-25395261

ABSTRACT

Myostatin (MSTN) is a secreted growth factor that negatively regulates skeletal muscle mass, and therefore, strategies to block myostatin-signaling pathway have been extensively pursued to increase the muscle mass in livestock. Here, we report a lentiviral vector-based delivery of shRNA to disrupt myostatin expression into goat fetal fibroblasts (GFFs) that were commonly used as karyoplast donors in somatic-cell nuclear transfer (SCNT) studies. Sh-RNA positive cells were screened by puromycin selection. Using real-time polymerase chain reaction (PCR), we demonstrated efficient knockdown of endogenous myostatin mRNA with 64% down-regulation in sh2 shRNA-treated GFF cells compared to GFF cells treated by control lentivirus without shRNA. Moreover, we have also demonstrated both the induction of interferon response and the expression of genes regulating myogenesis in GFF cells. The results indicate that myostatin-targeting siRNA produced endogenously could efficiently down-regulate myostatin expression. Therefore, targeted knockdown of the MSTN gene using lentivirus-mediated shRNA transgenics would facilitate customized cell engineering, allowing potential use in the establishment of stable cell lines to produce genetically engineered animals.


Subject(s)
Gene Knockdown Techniques/methods , Lentivirus/genetics , Myostatin/genetics , RNA Interference/physiology , Animals , Cells, Cultured , Fibroblasts , Genetic Vectors/genetics , Goats/genetics , HEK293 Cells , Humans , Myostatin/analysis , Myostatin/metabolism , RNA, Small Interfering/genetics
13.
Domest Anim Endocrinol ; 52: 17-24, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25700268

ABSTRACT

Undernutrition suppresses the growth of skeletal muscles and alters the expression of insulin-like growth factor 1 (IGF1), a key mitogen, and myostatin, a potent inhibitor of myogenesis. These changes can explain, at least in part, the reduced growth of skeletal muscles in underfed lambs. We have recently identified a myostatin splice variant (MSV) that binds to and antagonizes the canonical signaling of myostatin. In the present study, we hypothesized that the expression of MSV would be reduced in conjunction with myostatin and IGF1 in response to underfeeding in skeletal muscles of sheep. Young growing ewes were fed either ad libitum or an energy-restricted diet (30% of maintenance requirements) for 28 d. This regime of underfeeding resulted in a 24% reduction in body mass (P < 0.001) and a 36% reduction in the mass of the semitendinosus muscles relative to controls (P < 0.001) by day 28. The concentrations of MSV and IGF1 messenger RNA (mRNA) were reduced (both P < 0.001), but myostatin mRNA was not altered in semitendinosus muscles. Unlike the reduced expression of mRNA, the abundance of MSV protein was increased (P < 0.05) and there was no change in the abundance of myostatin protein. Our results suggest that undernutrition for 28 d decreases the signaling of myostatin by increasing the abundance of MSV protein. Although this action may reduce the growth inhibitory activity of myostatin, it cannot prevent the loss of growth of skeletal muscles during undernutrition.


Subject(s)
Insulin-Like Growth Factor I/genetics , Malnutrition/veterinary , Muscle, Skeletal/metabolism , Myostatin/genetics , Protein Isoforms/genetics , Sheep Diseases/metabolism , Animals , Female , Food Deprivation , Gene Expression Regulation/physiology , Insulin-Like Growth Factor I/analysis , Muscle, Skeletal/chemistry , Myostatin/analysis , Protein Isoforms/analysis , RNA, Messenger/analysis , Sheep/metabolism , Signal Transduction
14.
Fertil Steril ; 104(3): 744-52.e1, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26086422

ABSTRACT

OBJECTIVE: To evaluate the expression pattern of activins and related growth factor messenger RNA (mRNA) levels in adenomyotic nodules and in their endometrium. DESIGN: Prospective study. SETTING: University hospital. PATIENT(S): Symptomatic premenopausal women scheduled to undergo hysterectomy for adenomyosis. INTERVENTION(S): Samples from adenomyotic nodules and homologous endometria were collected. Endometrial tissue was also obtained from a control group. MAIN OUTCOME MEASURE(S): Quantitative real-time polymerase chain reaction (PCR) analysis and immunohistochemical localization of activin-related growth factors (activin A, activin B, and myostatin), binding protein (follistatin), antagonists (inhibin-α, cripto), and receptors (ActRIIa, ActRIIb) were performed. RESULT(S): Myostatin mRNA levels in adenomyotic nodule were higher than in eutopic endometrium and myostatin, activin A, and follistatin concentrations were higher than in control endometrium. No difference was observed for inhibin-α, activin B, and cripto mRNA levels. Increased mRNA levels of ActRIIa and ActRIIb were observed in adenomyotic nodules compared with eutopic endometrium and control endometrium. Immunofluorescent staining for myostatin and follistatin confirmed higher protein expression in both glands and stroma of patients with adenomyosis than in controls. CONCLUSION(S): The present study showed for the first time that adenomyotic tissues express high levels of myostatin, follistatin, and activin A (growth factors involved in proliferation, apoptosis, and angiogenesis). Increased expression of their receptors supports the hypothesis of a possible local effect of these growth factors in adenomyosis. The augmented expression of ActRIIa, ActRIIb, and follistatin in the endometrium of these patients may play a role in adenomyosis-related infertility.


Subject(s)
Activin Receptors, Type II/analysis , Adenomyosis/metabolism , Endometrium/chemistry , Follistatin/analysis , Myostatin/analysis , Activin Receptors, Type II/genetics , Activins/analysis , Adenomyosis/genetics , Adenomyosis/surgery , Adult , Case-Control Studies , Endometrium/surgery , Female , Follistatin/genetics , Humans , Immunohistochemistry , Myostatin/genetics , Prospective Studies , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction
15.
Reprod Sci ; 22(12): 1597-602, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26138721

ABSTRACT

Uterine leiomyoma is the most common benign neoplasm of female reproductive system, found in about 50% of women in reproductive age. The mechanisms of leiomyoma growth include cell proliferation, which is modulated by growth factors, and deposition of extracellular matrix (ECM). Activin A and myostatin are growth factors that play a role in proliferation of leiomyoma cells. Matrix metalloproteinases (MMPs) are known for their ability to remodel the ECM in different biological systems. The aim of this study was to evaluate the expression levels of activin ßA-subunit, myostatin, and MMP14 messenger RNAs (mRNAs) in uterine leiomyomas and the possible correlation of these factors with clinical features of the disease. Matrix metalloproteinase 14 was highly expressed in uterine leiomyoma and correlated with myostatin and activin A mRNA expression. Moreover, MMP14 and myostatin mRNA expression correlated significantly and directly with the intensity of dysmenorrhea. Overall, the present findings showed that MMP14 mRNA is highly expressed in uterine leiomyoma, where it correlates with the molecular expression of growth factors and is further increased in cases of intense dysmenorrhea.


Subject(s)
Biomarkers, Tumor/genetics , Dysmenorrhea/genetics , Leiomyoma/genetics , Matrix Metalloproteinase 14/genetics , Myostatin/genetics , RNA, Messenger/genetics , Uterine Neoplasms/genetics , Adult , Dysmenorrhea/diagnosis , Dysmenorrhea/enzymology , Female , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Humans , Immunohistochemistry , Inhibin-beta Subunits/genetics , Leiomyoma/complications , Leiomyoma/enzymology , Matrix Metalloproteinase 14/analysis , Middle Aged , Myostatin/analysis , Prospective Studies , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Severity of Illness Index , Uterine Neoplasms/complications , Uterine Neoplasms/enzymology , Uterine Neoplasms/pathology , Young Adult
16.
Exp Biol Med (Maywood) ; 238(12): 1421-30, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24186267

ABSTRACT

The mechanisms of muscle wasting and decreased mobility have a major functional effect in rheumatoid arthritis, but they have been poorly studied. The objective of our study is to describe muscular involvement and the pathways in an experimental model of arthritis compared to the pathways in disuse atrophy. Female Wistar rats were separated into three groups: control (CO), collagen-induced arthritis (CIA), and immobilized (IM). Spontaneous locomotion and weight were evaluated weekly. The gastrocnemius muscle was evaluated by histology and immunoblotting to measure the expression of myostatin (a negative regulator), LC3 (autophagy), MuRF-1 (proteasome-mediated proteolysis), MyoD, and myogenin (satellite-cell activation). The significance level was set at P < 0.05, and histological analysis of joints confirmed the severity of the arthropathy. There was a significant difference in spontaneous locomotion in the CIA group. Animal body weight, gastrocnemius muscle weight, and relative muscle weight decreased 20%, 30%, and 20%, respectively, in the CIA rats. Inflammatory infiltration and swelling were present in the gastrocnemius muscles of the CIA rats. The mean cross-sectional area was reduced by 30% in the CIA group and by 60% in the IM group. The expressions of myostatin and LC3 between the groups were similar. There was increased expression of MuRF-1 in the IM (1.9-fold) and CIA (3.1-fold) groups and of myogenin in the muscles of the CIA animals (1.7-fold), while MyoD expression was decreased in the IM (20%) rats. This study demonstrated that the development of experimental arthritis is associated with decreased mobility, body weight, and muscle loss. Both IM and CIA animal models presented muscle atrophy, but while proteolysis and the regeneration pathways were activated in the CIA model, there was no activation of regeneration in the IM model. We can assume that muscle atrophy in experimental arthritis is associated with the disease itself and not simply with decreased mobility.


Subject(s)
Arthritis/complications , Muscles/pathology , Muscular Atrophy/etiology , Animals , Arthritis/chemically induced , Arthritis/physiopathology , Collagen/pharmacology , Disease Models, Animal , Female , Microtubule-Associated Proteins/analysis , Muscle Proteins/analysis , Muscles/chemistry , Muscles/physiopathology , Muscular Atrophy/pathology , Muscular Atrophy/physiopathology , MyoD Protein/analysis , Myogenin/analysis , Myostatin/analysis , Rats , Rats, Wistar , Restraint, Physical/adverse effects , Tripartite Motif Proteins , Ubiquitin-Protein Ligases/analysis
17.
Meat Sci ; 89(4): 451-6, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21641731

ABSTRACT

This study investigated the growth performance and gene expression for muscle development between grass hay-fed (GH) and concentrate-fed (CT) steers. Daily gain and energy intake during the fattening period of the GH group were lower than those of the CT group. Analysis of C/EBPα, PPARγ2, myosin heavy chain (MHC), and myostatin gene expressions was performed by real-time PCR. Expressions of C/EBPα and myostatin in semitendinosus and longissimus lumborum (LL) muscles were higher in the CT group than in the GH group at the end of fattening. In LL muscle, MHC expression at the end of fattening was greater in the GH group than in the CT group. These results suggest that regulation of adipogenesis and myogenesis by the expression of genes involved in muscle development might have occurred in the skeletal muscle of the GH group by the feeding of grass hay and/or because of the low energy intakes.


Subject(s)
Animal Feed , Muscle Development/genetics , Muscle, Skeletal/growth & development , Adipogenesis/genetics , Animals , CCAAT-Enhancer-Binding Protein-alpha/analysis , CCAAT-Enhancer-Binding Protein-alpha/genetics , Cattle , Energy Intake , Gene Expression Regulation , Male , Muscle, Skeletal/drug effects , Myosin Heavy Chains/analysis , Myosin Heavy Chains/genetics , Myostatin/analysis , Myostatin/genetics , PPAR gamma/analysis , PPAR gamma/genetics , RNA/isolation & purification
18.
Arch Bronconeumol ; 47(12): 590-8, 2011 Dec.
Article in Spanish | MEDLINE | ID: mdl-22056524

ABSTRACT

INTRODUCTION: Among the extrapulmonary manifestations of COPD, dysfunction and loss of muscle mass/weight are those that have the greatest impact on the quality of life of patients. Our objective was to evaluate the molecular mechanisms that are potentially implicated in the limited development of muscle mass in the diaphragm and gastrocnemius of mice with experimentally-induced emphysema. METHODS: An experimental model in mice, in which emphysema was induced by means of the local instillation of elastase (n=6), while saline was administered to the controls (n=7). We determined the levels of oxidative stress, proteolytic systems, signaling pathways, growth factors and cell differentiation (western-blot) in the diaphragm and gastrocnemius of all the mice after 34 weeks. RESULTS: Upon comparing the mice with emphysema with the controls, the following findings were observed: (1) lower total body weight and lower weight of the diaphragm and gastrocnemius; (2) in the diaphragm, the levels of protein oxidation were increased, the mitochondrial antioxidant systems reduced, the levels of myostatin and of the ERK1/2 and FoxO1 signaling pathways were higher, and the myosin content was lower (67%); and (3) in the gastrocnemius of the emphysematous mice, the cytosolic antioxidants were decreased and the levels of myostatin and of the JNK and NF-kB signaling pathways were increased. CONCLUSIONS: The reduction of the myosin content observed in the diaphragm of mice with emphysema could explain their smaller size. Oxidative stress, myostatin and FoxO could be implicated in the loss of this structural protein.


Subject(s)
Muscle, Skeletal/pathology , Muscular Atrophy/etiology , Myosins/deficiency , Myostatin/physiology , Pulmonary Emphysema/complications , Respiratory Muscles/pathology , Animals , Antioxidants/analysis , Cytosol/chemistry , Diaphragm/chemistry , Diaphragm/pathology , Forkhead Box Protein O1 , Forkhead Transcription Factors/analysis , Forkhead Transcription Factors/physiology , MAP Kinase Signaling System , Male , Mice , Mice, Inbred A , Muscle Development , Muscle, Skeletal/chemistry , Muscle, Skeletal/metabolism , Muscular Atrophy/metabolism , Myostatin/analysis , NF-kappa B/metabolism , Organ Size , Oxidative Stress , Pancreatic Elastase/toxicity , Proteolysis , Pulmonary Emphysema/metabolism , Pulmonary Emphysema/pathology , Respiratory Muscles/metabolism , Weight Loss
19.
Arq. bras. med. vet. zootec ; Arq. bras. med. vet. zootec. (Online);65(1): 75-81, fev. 2013. graf, tab
Article in Portuguese | LILACS | ID: lil-667539

ABSTRACT

Neste trabalho foi estudada a correlação entre o perfil proteico do plasma seminal e a motilidade e viabilidade espermática em coelhos submetidos ao tratamento com vetores de expressão contendo o gene da eritropoetina (EPO) e com EPO recombinante humana. Foram identificadas, em coelhos submetidos ao tratamento com vetor de DNA contendo o gene da EPO, duas bandas proteicas associadas a alterações na motilidade espermática - 48kDa à baixa motilidade (P<0,05) e 18kDa à alta motilidade (P<0,05) - e esse fator foi associado a maior viabilidade espermática (P<0,05). Em coelhos submetidos ao tratamento com EPO recombinante, um fator proteico, 63kDa, associou-se à alta motilidade espermática (P<0,05), enquanto dois, 26 e 40kDa, foram associados à alta viabilidade espermática (P<0,05). Esses resultados sugerem que o doping genético pode ocasionar mudanças no perfil proteico do plasma seminal, provocando alterações na motilidade e viabilidade espermática.


In this study the correlation between seminal plasma protein profile and the sperm motility and sperm viability in rabbits submitted to treatment with an expression vector containing EPO gene and with human recombinant EPO was evaluated. In rabbits submitted to treatment with EPO expression vector, two protein bands were associated to sperm motility - 48kDa associated to low motility (P<0.05) and 18kDa to high motility (P<0.05) - and this protein band was also associated to high sperm viability (P<0.05). In rabbits submitted to treatment with human recombinant EPO, a protein factor, 63kDa, was associated to high sperm motility (P<0.05) while two protein factors, 26 and 40kDa, were associated to high sperm viability (P<0.05). These results suggest that gene doping leads to changes in rabbit seminal plasma protein, altering sperm motility and sperm viability.


Subject(s)
Animals , Rabbits , Semen Analysis/veterinary , Erythropoietin/analysis , Erythropoietin/physiology , Myostatin/analysis , Rabbits/genetics , Reproduction , Semen/immunology , Semen/parasitology , Veterinary Medicine
20.
J Endocrinol ; 197(1): 85-93, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18372235

ABSTRACT

Angiotensin II (AngII) plays a critical role in cardiac remodeling and promotes cardiac myocyte hypertrophy. Myostatin, a negative regulator of muscle growth, is increased in hypertrophied and infarcted heart. The direct effect of AngII on cardiac myocyte myostatin expression has not been previously investigated. We hypothesized that myostatin may act as a cardiac endocrine inhibitor for AngII. AngII-induced myostatin protein expression in cultured rat neonatal cardiomyocytes was dose-dependent. AngII significantly increased myostatin protein and mRNA expression in a time-dependent manner. Addition of losartan, SB203580, or p38 siRNA 30 min before AngII stimulation significantly blocked the increase of myostatin protein by AngII. AngII significantly increased phosphorylation of p38 while SB205380 and losartan attenuated the phosphorylation of p38 induced by AngII. AngII increased, while myostatin-Mut plasmid, SB203580, losartan, and myocyte enhance factor 2 (MEF-2) antibody abolished the myostatin promoter activity. Co-stimulation with myostatin and AngII significantly inhibited the protein synthesis induced by AngII. In conclusion, AngII enhances myostatin expression in cultured rat neonatal cardiomyocytes. The AngII-induced myostatin is mediated through p38 MAP kinase and MEF-2 pathway.


Subject(s)
Angiotensin II/pharmacology , MADS Domain Proteins/physiology , Myocytes, Cardiac/drug effects , Myogenic Regulatory Factors/physiology , Myostatin/analysis , p38 Mitogen-Activated Protein Kinases/physiology , Animals , Animals, Newborn , Cardiomegaly/etiology , MAP Kinase Signaling System , MEF2 Transcription Factors , Myocytes, Cardiac/chemistry , Myostatin/genetics , Promoter Regions, Genetic , Protein Biosynthesis , Rats , Rats, Wistar , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL