Myostatin Human, His

Myostatin Human Recombinant, His Tag
Shipped with Ice Packs
In Stock

Description

Production and Expression Systems

Recombinant Myostatin Human, His is produced in two primary systems:

Escherichia coli

  • CYT-445 Construct: 152 amino acids, N-terminal His-tag, molecular weight 16.7 kDa .

  • Purity: >90% by SDS-PAGE, endotoxin levels <1.0 EU/µg .

HEK293 Cells

  • HEK293 Construct: 360 amino acids, includes His-tag and linker sequences (MW 41.1 kDa) .

  • Post-Translational Modifications: Glycosylation observed, enhancing stability .

Inhibition of Muscle Hypertrophy

  • Myostatin binds activin type II receptors (ActRIIB), activating SMAD2/3 signaling to suppress muscle differentiation .

  • Mechanism:

    • Blocks Akt/mTOR pathway, reducing protein synthesis .

    • Upregulates ubiquitin ligases (e.g., MuRF1), promoting proteasomal degradation .

Mitochondrial and Metabolic Effects

  • Mstn−/− mice exhibit 26% fewer mitochondria and reduced oxidative capacity, leading to weaker specific muscle force .

  • Mitochondrial DNA/nuclear DNA ratio decreases by 30% in myostatin-deficient muscle .

Clinical Correlations

  • HIV Wasting Syndrome: Elevated serum myostatin inversely correlates with fat-free mass (r = -0.62, P < 0.01) .

  • Therapeutic Potential: Antibody blockade increases muscle mass in preclinical models but risks mitochondrial depletion .

Assays and Diagnostics

  • ELISA/Western Blot: Used to quantify myostatin levels in serum and muscle biopsies .

  • Muscle Atrophy Studies: Recombinant Myostatin Human, His induces atrophy in cultured myotubes (EC₅₀ ~10 ng/mL) .

Drug Development

  • Ligand Traps: Soluble ActRIIB-Fc fusion proteins neutralize myostatin, increasing lean mass by 15–25% in murine models .

Comparative Analysis of Recombinant Constructs

ParameterE. coli-Expressed HEK293-Expressed
GlycosylationNoYes
Endotoxin Levels<1.0 EU/µg<1.0 EU/µg
Yield0.25–0.8 mg/mL0.25–0.8 mg/mL
ApplicationsStructural studies, in vitroCell signaling, in vivo

Evolutionary and Genetic Considerations

  • Positive Selection in Humans: Sub-Saharan populations show high frequencies (31%) of GDF8 variants linked to muscle hypertrophy, suggesting adaptive evolution .

  • Loss-of-Function Mutations: Rare human cases (e.g., German and American boys) exhibit 50–100% greater muscle mass .

Challenges and Future Directions

  • Therapeutic Risks: Chronic myostatin inhibition may deplete satellite cells, exacerbating muscular dystrophy .

  • Mitochondrial Rescue: Co-administration with PPAR-δ agonists (e.g., GW501516) restores oxidative capacity in Mstn−/− mice .

Product Specs

Introduction
Myostatin (GDF-8), a member of the transforming growth factor β superfamily, is a 12 kDa mature glycoprotein composed of 113 amino acid residues. It is exclusively expressed in human skeletal muscle and secreted into the plasma. Myostatin plays a crucial role in regulating skeletal muscle mass. Research indicates that myostatin may also be involved in cardiac development and function.
Description
This protein consists of a total of 152 amino acids, with a molecular weight of 16.7 kDa (calculated). It features an N-terminal His-tag and a spacer (43AA, highlighted). The amino acid sequence of the human myostatin portion within the fusion protein aligns with the UniProtKB/Swiss-Prot entry O14793.
Physical Appearance
White lyophilized powder (freeze-dried) after filtration.
Formulation
The protein solution (0.5 mg/ml in 0.05M acetate buffer, pH 4.5) is filtered through a 0.4 μm filter and then lyophilized.
Solubility
To prepare a working stock solution of around 0.5 mg/mL, add 0.1M Acetate buffer (pH 4) to the lyophilized pellet and allow it to dissolve completely. For use at higher pH values, dilute the solution with the appropriate buffer to a concentration of 10 μg/ml. Note that the solubility of this antigen is limited at higher concentrations.
Stability
Store the lyophilized protein at -20°C. After reconstitution, aliquot the product to prevent repeated freeze-thaw cycles. The reconstituted protein remains stable at 4°C for a limited time and shows no significant change after two weeks at this temperature.
Purity
Purity determined by SDS-PAGE is greater than 95%.
Synonyms
GDF-8, MSTN, Growth/Differentiation Factor 8,MSTN Muscle Hypertrophy.
Source
Escherichia Coli.
Amino Acid Sequence
MRGSHHHHHH GMASMTGGQQ MGRDLYDDDD KDPSSRSAVR SRRDFGLDCD EHSTESRCCR YPLTVDFEAFGWDWIIAPKR YKANYCSGEC EFVFLQKYPH THLVHQANPR GSAGPCCTPT KMSPINMLYF NGKEQIIYGKIPAMVVDRCG CS.

Q&A

What is myostatin and what is its primary function in human physiology?

Myostatin (also known as growth differentiation factor 8 or GDF8) is a protein encoded by the MSTN gene in humans that functions as a negative regulator of skeletal muscle mass. It is a myokine that is produced and released by myocytes and acts on muscle cells to inhibit muscle growth and differentiation . Myostatin belongs to the transforming growth factor-β (TGF-β) superfamily of proteins that regulate cellular growth and differentiation . In physiological contexts, myostatin is predominantly expressed in skeletal muscle tissue where it helps maintain homeostasis by balancing muscle growth and preventing excessive muscle hypertrophy . The protein is assembled in skeletal muscle before being released into the bloodstream, where it can exert systemic effects . Animals with myostatin deficiency or those treated with myostatin inhibitors exhibit significantly increased muscle mass, demonstrating its crucial role in regulating muscle development .

What is the molecular structure of mature human myostatin protein?

Human myostatin consists of two identical subunits forming a homodimer, with each subunit consisting of 109 amino acid residues in its mature form . The full-length gene encodes a 375-amino acid prepro-protein that undergoes proteolytic processing to generate the mature, active form . The mature myostatin protein has a molecular weight of approximately 25.0 kDa and features a cystine knot structure that is characteristic of TGF-β family members . Each mature myostatin monomer consists of four antiparallel β-strands (referred to as "fingers") and the cystine-knot motif . The two protomers associate through their concave "palms" and are linked covalently through a disulfide bond between equivalent Cys339 residues in the "wrist" region . This specific structural arrangement is critical for myostatin's biological activity and receptor binding properties .

How are His-tagged myostatin recombinant proteins typically produced for research use?

Recombinant human myostatin proteins with histidine tags are typically produced using bacterial expression systems, with E. coli being the most common host . The process involves inserting the human MSTN gene sequence (specifically the mature domain, often residues 267-375) into an expression vector that incorporates an N-terminal or C-terminal histidine tag sequence . The His-tag consists of multiple histidine residues (usually six) that facilitate protein purification through metal affinity chromatography techniques. After expression in E. coli, the bacterial cells are lysed, and the His-tagged myostatin protein is purified using nickel or cobalt-based affinity resins . The purified protein is then typically formulated in a buffer system, such as Tris-HCl with glycerol, for stability during storage . This recombinant production method allows researchers to obtain relatively large quantities of purified myostatin protein for various experimental applications, including structural studies, binding assays, and cell-based functional tests .

What is the process of myostatin synthesis and activation in vivo?

The activation of this latent complex requires a second proteolytic event where members of the BMP-1/tolloid family of metalloproteases cleave the propeptide, releasing the active mature myostatin dimer . Only when free of these inhibitory proteins can myostatin signal by binding initially to type 2 activin receptors (ACVR2 and ACVR2B), followed by engagement of the type 1 receptors (ALK4 and ALK5) . This elaborate processing mechanism ensures tight regulation of myostatin activity, preventing uncontrolled inhibition of muscle growth .

How does the structure of pro-myostatin contribute to its latency?

Crystal structure analyses and small-angle X-ray scattering (SAXS) studies have revealed that pro-myostatin adopts a unique open, V-shaped structure with a domain-swapped arrangement, which is distinct from the structure of other TGF-β family members like TGF-β1 . The pro-domain of myostatin contains "forearm" helices that grasp the mature growth factor, and a globular "arm/shoulder" domain that sits atop the mature growth factor protomers .

Unlike the more closed conformation observed in pro-TGF-β1, pro-myostatin exhibits an "open-armed" conformation with no direct interaction between the arm/shoulder domains of the domain-swapped dimer . This structural arrangement stabilizes the interaction between the pro-domains and the mature growth factor, effectively preventing receptor binding and subsequent signaling . The pro-mature complex, after cleavage of the furin site, has significantly reduced activity compared to the mature growth factor and forms a stable complex that is resistant to natural antagonists like follistatin . This unique structural arrangement contributes to a regulated stepwise activation process that distinguishes myostatin from other TGF-β superfamily members .

What methodological approaches are used to study myostatin activation in research settings?

Researchers employ several methodological approaches to study myostatin activation:

  • Structural studies: X-ray crystallography and SAXS are used to determine the three-dimensional structure of pro-myostatin and its various processed forms, providing insights into the conformational changes associated with activation .

  • Proteolytic processing assays: In vitro assays using purified proteases (furin, BMP-1, TLL2) to cleave recombinant pro-myostatin, followed by SDS-PAGE and Western blotting to monitor the conversion from latent to active forms .

  • Cell-based reporter assays: Functional activation of myostatin can be measured using cells expressing myostatin receptors coupled to luciferase or other reporter genes that quantify signaling pathway activation .

  • In vivo activation models: Transgenic mouse models with modified myostatin processing sites or expression of myostatin inhibitors help elucidate the physiological importance of controlled activation .

  • Antibody-based detection methods: Development of antibodies that specifically recognize different forms of myostatin (pro-, latent, or active) enables researchers to track processing and activation in biological samples .

These methodological approaches collectively provide a comprehensive understanding of myostatin activation mechanisms, which is crucial for developing targeted therapeutic interventions .

What are the advantages of using His-tagged myostatin proteins in experimental research?

His-tagged myostatin proteins offer several significant advantages in experimental research:

  • Enhanced purification efficiency: The histidine tag forms strong coordination bonds with metal ions (typically Ni²⁺ or Co²⁺), allowing for single-step purification using immobilized metal affinity chromatography (IMAC). This results in high-purity protein preparations suitable for sensitive experimental applications .

  • Detection versatility: The His-tag can be recognized by anti-His antibodies, enabling straightforward detection in Western blotting, ELISA, and immunohistochemistry without requiring myostatin-specific antibodies .

  • Immobilization capability: His-tagged proteins can be immobilized on nickel-coated surfaces for interaction studies, including surface plasmon resonance (SPR) and protein microarrays, facilitating binding kinetics and partner protein identification studies .

  • Structural flexibility: The His-tag can be engineered at either the N-terminus or C-terminus depending on experimental requirements, and in some cases can be removed after purification via engineered protease sites .

  • Quantification simplicity: The consistent nature of the His-tag enables more standardized quantification across different preparations, improving experimental reproducibility when comparing different myostatin variants or mutants .

These advantages make His-tagged myostatin proteins valuable tools for studying protein-protein interactions, screening potential inhibitors, and conducting structural analyses that advance our understanding of myostatin biology .

What are optimal storage and handling conditions for His-tagged myostatin recombinant proteins?

Based on manufacturer recommendations and research protocols, His-tagged myostatin recombinant proteins should be handled and stored under the following conditions for optimal stability and activity:

Storage ParameterRecommended ConditionNotes
Short-term storage2-8°C (refrigerated)Stable for approximately one week
Long-term storage-20°C to -80°CStore in small aliquots to avoid repeated freeze-thaw cycles
Buffer composition20 mM Tris-HCl (pH 8.0) with 10% glycerolGlycerol helps prevent freeze-thaw damage
Freeze-thaw cyclesMinimize repeated cyclesEach cycle can reduce activity by 10-30%
Working concentrationExperiment-dependentTypically 10-100 ng/mL for cell-based assays
ReconstitutionFollow protein-specific instructionsSome preparations may require special reconstitution protocols

When handling the protein, it's important to work quickly and keep the protein cold to prevent degradation. Some researchers add protease inhibitors to prevent unwanted proteolytic processing during experimental procedures . For functional assays, it's essential to consider that commercially available His-tagged myostatin proteins may be in a denatured form, making them less suitable for functional studies but appropriate for applications like Western blotting or imaging assays .

How can researchers assess the biological activity of purified His-tagged myostatin proteins?

Researchers can employ several methodological approaches to assess the biological activity of purified His-tagged myostatin proteins:

  • Cell proliferation inhibition assays: Since myostatin inhibits myoblast proliferation, researchers can measure the ability of His-tagged myostatin to reduce proliferation rates in myoblast cell lines like C2C12. This is typically quantified using colorimetric assays (MTT, WST-1) or DNA synthesis measurements (BrdU incorporation) .

  • Smad phosphorylation analysis: Active myostatin triggers phosphorylation of Smad2/3 transcription factors in target cells. Western blotting for phospho-Smad2/3 provides a direct measure of myostatin signaling pathway activation .

  • Reporter gene assays: Cells transfected with Smad-responsive elements driving luciferase or other reporter genes can quantitatively measure myostatin signaling activity .

  • Myogenic differentiation inhibition: Myostatin inhibits the differentiation of myoblasts into myotubes. Researchers can assess this by measuring differentiation markers (myogenin, MyHC) or by quantifying fusion index in differentiating cultures exposed to the purified protein .

  • Receptor binding assays: Surface plasmon resonance (SPR) or other binding assays can determine whether His-tagged myostatin can bind to its cognate receptors (ActRIIA, ActRIIB) with appropriate affinity .

  • Comparative activity analysis: Comparing the activity of His-tagged myostatin with a reference standard (e.g., commercially available non-tagged myostatin) can provide a relative measure of biological potency .

For recombinant myostatin proteins that are supplied in a denatured state, refolding protocols may be necessary before conducting activity assays. The specific assay chosen should align with the research question and account for the protein's preparation method .

How do various myostatin inhibition strategies compare in research efficacy?

Research on myostatin inhibition has explored multiple strategies, each with distinct mechanisms and efficacy profiles:

Inhibition StrategyMechanismResearch EfficacySpecificityNotable Findings
Anti-myostatin antibodiesDirect binding to mature myostatinHighVariableGYM329 antibody shows superior efficacy by binding latent myostatin and "sweeping" it from circulation
Propeptide administrationBinds mature myostatin, preventing receptor interactionModerate-HighHighNaturally occurring inhibitory mechanism with high specificity but potential stability issues
Soluble receptor decoys (ActRIIB-Fc)Compete with cellular receptors for myostatin bindingVery HighLowAffects multiple TGF-β ligands beyond myostatin, leading to broader effects and potential side effects
Follistatin and derivativesBind and neutralize mature myostatinHighLowImpacts multiple TGF-β family members including activins
Small molecule inhibitorsVarious mechanisms including signaling pathway disruptionVariableVariableLess developed than biologic approaches but potentially more cost-effective
CRISPR/gene editingGenomic modification of MSTN geneVery HighVery HighPermanent effects; still primarily in preclinical research phases

What are the methodological challenges in studying myostatin's interaction with other regulatory proteins?

Researchers face several methodological challenges when investigating myostatin's interactions with its regulatory proteins:

  • Structural complexity: Myostatin exists in multiple forms (prepro-, pro-, latent, and active), each potentially interacting differently with regulatory proteins. Distinguishing these interactions requires specialized antibodies or tags that don't interfere with native binding interfaces .

  • In vitro versus in vivo relevance: Interactions observed in purified protein systems may not reflect the physiological context where multiple competing regulators are present simultaneously. Developing experimental systems that better mimic the in vivo environment remains challenging .

  • Temporal dynamics: Myostatin activation involves sequential processing steps with different regulatory proteins acting at different stages. Capturing these temporal dynamics requires sophisticated time-course experiments with appropriate detection sensitivity .

  • Tissue-specific regulation: Myostatin regulation differs across tissues, with muscle, adipose, and bone environments containing different complements of regulatory proteins. Replicating these tissue-specific contexts in research models requires specialized approaches .

  • Quantitative analysis limitations: Current methods often provide qualitative or semi-quantitative assessments of interactions rather than precise binding constants or stoichiometry in physiological conditions. Developing more quantitative approaches remains an active area of research .

To address these challenges, researchers are employing integrated approaches combining structural biology techniques (X-ray crystallography, cryo-EM) with functional assays and advanced imaging methods (FRET, BRET) to better characterize the complex network of interactions regulating myostatin activity .

How does myostatin contribute to crosstalk between muscle and other tissues in metabolic regulation?

Myostatin functions as a key mediator in inter-tissue communication, particularly between skeletal muscle and other metabolically active tissues:

  • Muscle-adipose tissue crosstalk: Myostatin secreted from muscle influences adipocyte differentiation and metabolism. Research indicates that myostatin promotes adipogenesis while inhibiting myogenesis, potentially contributing to the metabolic phenotype observed in conditions like sarcopenic obesity . In myostatin-deficient models, reduced adiposity accompanies increased muscle mass, suggesting a regulatory role in energy partitioning between these tissues .

  • Muscle-liver axis: Studies suggest that myostatin affects hepatic insulin sensitivity and glucose metabolism. Myostatin may indirectly influence hepatic glucose production through alterations in muscle metabolism and the release of myokines that target the liver . This interaction has implications for understanding insulin resistance and type 2 diabetes pathophysiology .

  • Muscle-bone communication: Myostatin regulates bone metabolism through direct effects on osteoblasts and osteoclasts, as well as indirect effects via altered mechanical loading from increased muscle mass. This crosstalk is particularly important during aging, where myostatin inhibition may simultaneously address sarcopenia and osteoporosis .

  • Cardiac-skeletal muscle interaction: Myostatin is expressed in cardiac tissue and may coordinate cardiac and skeletal muscle adaptation to stress. During heart failure, elevated myostatin levels contribute to skeletal muscle wasting, creating a pathological feedback loop .

  • Immune system interactions: Emerging research suggests myostatin modulates inflammatory responses and may influence muscle-immune system crosstalk during injury and repair processes .

Methodologically, studying these complex interactions requires integrated physiological models and tissue-specific genetic manipulations that can distinguish direct from indirect effects of myostatin signaling . Technologies like tissue-specific knockout models, parabiosis experiments, and multi-omics approaches are advancing our understanding of myostatin's role in inter-tissue communication and metabolic homeostasis .

What are the key differences between various expression systems for producing His-tagged myostatin?

Different expression systems offer distinct advantages and limitations for producing His-tagged myostatin:

Expression SystemAdvantagesLimitationsYieldPost-translational ModificationsApplications
E. coliHigh yield, fast growth, cost-effective, simplicityLacks eukaryotic PTMs, protein may form inclusion bodies requiring refolding, potential endotoxin contaminationHighMinimalStructural studies, antibody production, protein-protein interaction studies
Yeast (P. pastoris, S. cerevisiae)Higher eukaryote, secretion possible, proper folding of disulfide bondsHyperglycosylation may occur, lower yields than E. coliModerateBasic glycosylation, disulfide bondsFunctional studies requiring proper folding
Insect cells (Baculovirus)Advanced eukaryotic PTMs, proper folding, secretionMore expensive, longer production time, complex methodologyModerate-HighNear-mammalian glycosylation patternsCell-based assays, receptor binding studies
Mammalian cells (CHO, HEK293)Native-like protein with proper PTMs and foldingHighest cost, lowest yield, most complex methodologyLow-ModerateFull mammalian PTMs including complex glycosylationFunctional studies requiring authentic activity, therapeutic development

How can researchers optimize purification protocols for His-tagged myostatin proteins?

Optimizing purification protocols for His-tagged myostatin proteins requires attention to several key factors:

  • Lysis buffer optimization: Including low concentrations of imidazole (10-20 mM) in the lysis buffer reduces non-specific binding to the affinity resin while maintaining specific binding of the His-tagged protein. Additionally, incorporating reducing agents (DTT or β-mercaptoethanol) can help maintain proper folding of myostatin's cysteine-rich structure .

  • Affinity chromatography conditions: Using a step-wise elution gradient with increasing imidazole concentrations (typically 50, 100, 250, and 500 mM) can help separate His-tagged myostatin from contaminants with lower affinity for the resin. Researchers should collect and analyze fractions by SDS-PAGE to identify optimal elution conditions .

  • Secondary purification steps: Following initial IMAC purification, secondary purification steps such as size-exclusion chromatography (SEC) can significantly improve purity by separating monomers, dimers, and higher-order aggregates based on their molecular size. This is particularly important for myostatin, which forms homodimers .

  • Protein refolding strategies: For E. coli-expressed myostatin that forms inclusion bodies, optimized refolding protocols involving gradual dilution into refolding buffer containing redox pairs (reduced/oxidized glutathione) can improve the yield of correctly folded protein. Pulsed renaturation approaches may be particularly effective for myostatin's complex disulfide-bonded structure .

  • Tag removal considerations: When the His-tag needs to be removed for functional studies, incorporating a specific protease cleavage site (TEV, HRV-3C) between the tag and protein allows for efficient tag removal under mild conditions that preserve protein structure and activity .

  • Quality control metrics: Establishing rigorous quality control metrics, including SDS-PAGE with silver staining, Western blotting, mass spectrometry, and activity assays, helps ensure batch-to-batch consistency in purified His-tagged myostatin preparations .

By systematically optimizing these parameters, researchers can achieve higher purity, yield, and activity of His-tagged myostatin proteins for their specific experimental applications .

What analytical methods are most effective for confirming the identity and purity of His-tagged myostatin preparations?

Researchers should employ a combination of analytical methods to comprehensively characterize His-tagged myostatin preparations:

  • SDS-PAGE and Western blotting: Provides information on protein size, purity, and potential degradation products. Using both reducing and non-reducing conditions can reveal the extent of dimer formation through disulfide bonds. Western blotting with anti-His and anti-myostatin antibodies confirms protein identity .

  • Mass spectrometry:

    • MALDI-TOF MS: Confirms the molecular weight of the intact protein

    • LC-MS/MS: Provides peptide fingerprinting for definitive identification and sequence coverage

    • Intact protein MS: Can identify post-translational modifications and verify the intact mass of the His-tagged construct

  • Size-exclusion chromatography coupled with multi-angle light scattering (SEC-MALS): Determines the oligomeric state of the purified protein, critical for myostatin which functions as a dimer .

  • Circular dichroism (CD) spectroscopy: Assesses secondary structure content, particularly important for confirming proper folding after purification from E. coli inclusion bodies .

  • Dynamic light scattering (DLS): Evaluates protein homogeneity and detects aggregation, which can affect functional activity.

  • Endotoxin testing: Essential for preparations intended for cell-based assays to prevent misleading results from endotoxin contamination, particularly important for E. coli-derived proteins .

  • Bioactivity assays: Functional verification through cell-based assays measuring myostatin-specific activities, such as Smad2/3 phosphorylation or inhibition of myoblast differentiation .

The combined results from these analytical methods provide a comprehensive profile of the His-tagged myostatin preparation, ensuring that it meets the necessary quality standards for research applications .

What are emerging approaches for studying myostatin's tissue-specific effects beyond muscle?

Emerging research approaches are expanding our understanding of myostatin's roles beyond muscle:

  • Conditional tissue-specific knockout models: Advanced genetic tools using Cre-loxP systems allow researchers to delete myostatin or its receptors in specific tissues at defined time points, revealing direct versus indirect effects across tissues . These models are particularly valuable for understanding myostatin's impact on adipose tissue, liver, bone, and cardiac muscle without the developmental compensation that occurs in global knockout models .

  • Single-cell transcriptomics: This technique allows researchers to identify cell-specific responses to myostatin within heterogeneous tissues, revealing previously unrecognized target cell populations and signaling diversity . Applied to various metabolic tissues, this approach is uncovering how myostatin signals are integrated at the cellular level.

  • Tissue-specific secretome analysis: Mass spectrometry-based proteomics of tissue-specific secretomes in the presence or absence of myostatin is identifying novel downstream effectors and interconnected signaling networks . This systems biology approach helps map the complex cascade of effects triggered by myostatin across multiple tissues.

  • In vivo metabolic flux analysis: Isotope tracing combined with metabolomics provides a dynamic view of how myostatin alters metabolic pathways across tissues, particularly important for understanding its role in energy homeostasis and substrate utilization .

  • Organoid and multi-tissue-on-chip models: These emerging technologies allow for controlled study of myostatin's effects on tissue-tissue interactions in more physiologically relevant contexts than traditional cell culture, while maintaining experimental accessibility not possible in whole-animal studies .

These methodological advances are revealing myostatin as a multifunctional regulator with direct effects on adipogenesis, bone metabolism, cardiac function, and insulin sensitivity, significantly expanding its role beyond simply being a negative regulator of muscle mass .

How might structural insights from His-tagged myostatin research inform therapeutic development?

Structural studies using His-tagged myostatin proteins are providing crucial insights that can guide therapeutic development in several ways:

  • Targeted antibody design: Crystal structures of myostatin in its various forms (pro-, latent, and active) are enabling structure-based design of antibodies that target specific epitopes critical for activation or receptor binding . This approach has already yielded promising candidates like GYM329, which specifically targets the latent form of myostatin with improved efficacy over conventional antibodies .

  • Propeptide-based inhibitors: Structural understanding of how the propeptide interacts with mature myostatin is informing the development of optimized propeptide mimetics with enhanced stability and potency . These could provide highly specific myostatin inhibition with fewer off-target effects compared to broader TGF-β pathway inhibitors .

  • Small molecule binding site identification: Structural analysis reveals potential binding pockets in myostatin that could be targeted by small molecule inhibitors, offering advantages in terms of oral bioavailability and cost compared to biologic therapies . These studies help explain the molecular basis for the "open-armed" conformation of pro-myostatin and how it differs from other TGF-β family members .

  • Receptor interface targeting: Detailed structural information about myostatin's interaction with its type I and type II receptors enables the design of molecules that specifically disrupt these interactions without affecting other TGF-β family members . This approach could minimize side effects associated with broader receptor blockade.

  • Activation mechanism modulation: Understanding the structural changes during the transition from latent to active myostatin reveals potential intervention points to prevent activation, such as protease-resistant propeptide variants or molecules that stabilize the latent complex .

These structure-guided approaches are particularly valuable because they can enhance specificity for myostatin over other TGF-β family members, potentially reducing side effects while maintaining efficacy in conditions such as muscle wasting diseases, sarcopenia, and certain metabolic disorders .

What are the most promising methodological innovations for studying myostatin's role in aging and age-related diseases?

Several innovative methodological approaches are advancing research on myostatin's role in aging:

  • Longitudinal aging cohort studies with myostatin profiling: Comprehensive analysis of circulating myostatin levels and its various forms (free, bound, pro-domain) in well-characterized aging cohorts, correlating with functional outcomes, muscle quality measures, and other biomarkers of aging . These studies address conflicting data regarding serum myostatin changes during aging in humans by implementing standardized measurement protocols and distinguishing between different forms of the protein .

  • Single-cell analysis of aged tissues: Application of single-cell RNA sequencing and proteomics to investigate cell-type-specific changes in myostatin signaling pathways during aging across multiple tissues . This approach reveals how aging alters the cellular response to myostatin and identifies potential intervention points.

  • Senescence interaction models: In vitro and in vivo models examining how myostatin interacts with senescent cells, which accumulate with age and secrete pro-inflammatory factors (SASP). These studies are uncovering potential synergistic effects between myostatin and cellular senescence in age-related muscle wasting .

  • Multi-omics integration: Combining genomics, transcriptomics, proteomics, and metabolomics data to construct comprehensive networks of myostatin's influence on systemic aging processes . This systems biology approach helps identify key nodes where myostatin intersects with other aging pathways.

  • Novel animal models with accelerated or delayed aging: Utilizing models with altered aging trajectories to isolate myostatin's causal role in age-related pathologies from other age-associated changes . These include models with genetic modifications in longevity pathways or exposed to interventions known to extend lifespan.

  • Non-invasive imaging techniques: Development of specific tracers for PET/SPECT imaging or MRI techniques that can track myostatin activity in vivo, enabling longitudinal assessment of intervention effects without requiring tissue sampling .

These methodological innovations are particularly important because they address the complexity of aging as a multifactorial process and help distinguish myostatin's direct contributions to age-related muscle and metabolic decline from other concurrent pathological processes .

Product Science Overview

Structure and Function

Myostatin is synthesized as a large precursor molecule that undergoes proteolytic cleavage to generate an N-terminal propeptide and a disulfide-linked C-terminal dimer, which is the biologically active form of the protein . The circulating form of myostatin consists of a latent complex of the C-terminal dimer and other proteins, including its propeptide, which maintain the C-terminal dimer in an inactive state. Activation of myostatin requires additional cleavage of the prodomain by a tolloid-like metalloproteinase .

Biological Role

Myostatin acts as a negative regulator of skeletal muscle growth. It inhibits the differentiation of myoblasts into mature muscle fibers, thereby controlling muscle mass. Studies have shown that myostatin could also play an important role in cardiac development and physiology . The inhibition of myostatin activity has been explored as a potential therapeutic approach for muscle-wasting diseases and conditions characterized by muscle loss.

Recombinant Myostatin (Human, His Tag)

Recombinant human myostatin is produced using various expression systems, such as Escherichia coli. The recombinant protein typically includes a His tag, which is a sequence of histidine residues added to the protein to facilitate purification and detection . The His tag allows for easy purification of the protein using affinity chromatography techniques, which are based on the binding affinity between histidine residues and metal ions.

Recombinant myostatin is used in research to study its mechanisms of action, its role in muscle development, and its potential therapeutic applications. It is also used as a positive control in assays such as Western blots and ELISA .

Applications and Research

The study of myostatin has significant implications for understanding muscle biology and developing treatments for muscle-related diseases. Researchers use recombinant myostatin to investigate its effects on muscle cells, identify potential inhibitors, and explore its role in various physiological processes. The availability of high-purity recombinant myostatin with a His tag has facilitated these studies by providing a reliable and consistent source of the protein for experimental use .

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2024 Thebiotek. All Rights Reserved.