Myostatin Propeptide Human

Myostatin Propeptide Human Recombinant
Shipped with Ice Packs
In Stock

Description

Mechanism of Action

Myostatin Propeptide suppresses myostatin signaling through:

  1. Direct Binding: Neutralizes mature myostatin, preventing interaction with activin type IIB receptors (ActRIIB) .

  2. Downstream Effects:

    • Inhibits SMAD2/3 phosphorylation, blocking muscle atrophy pathways .

    • Enhances Akt signaling, promoting protein synthesis and muscle hypertrophy .

  3. Metabolic Regulation: Reduces oxidative metabolism in muscles, increasing reliance on anaerobic pathways .

Muscle Hypertrophy and Injury Recovery

  • Animal Studies: Administration in mice increased muscle mass by 15–20% and accelerated bone fracture healing by 30% .

  • Fibrosis Reduction: Decreased fibrous tissue in muscle injury models by 20% .

Genetic and Evolutionary Insights

  • Human Variants: Two amino acid substitutions (residues 55 and 153) in sub-Saharan populations suggest adaptive evolution for muscle development .

  • Loss-of-Function Mutations: Associated with hypermuscular phenotypes in humans and animals .

Doping and Detection Challenges

  • WADA Status: Prohibited under S4 ("Hormone and Metabolic Modulators") due to performance-enhancing effects .

  • Detection Methods:

    • SDS-PAGE/Western blot identifies non-glycosylated, GST-tagged black-market variants (54–55 kDa vs. endogenous 33 kDa) .

    • Urine ELISA achieves a detection limit of 0.4 ng/mL .

Key Research Findings

  • Dual Role in Metabolism: Myostatin Propeptide deficiency increases muscle mass but reduces endurance due to shifted oxidative-glycolytic balance .

  • Synergy with Follistatin: Both proteins bind >70% of circulating myostatin, offering combinatorial therapeutic potential .

  • Dose Dependency: In mice, 20 mg/kg doses optimized muscle growth without toxicity .

Challenges and Future Directions

  • Therapeutic Delivery: Lack of glycosylation in recombinant forms may limit efficacy in humans .

  • Long-Term Safety: Unclear consequences of chronic myostatin inhibition on cardiac or smooth muscle .

Product Specs

Introduction
Myostatin (GDF-8), a member of the transforming growth factor beta (TGFβ) superfamily, is a protein that plays a crucial role in regulating skeletal muscle mass. It acts as a negative regulator, meaning it inhibits muscle growth. Myostatin is found in our bloodstream in an inactive form, bound to other molecules such as its propeptide and follistatin-related gene. The myostatin propeptide is essential because it can bind to myostatin and block its activity. This interaction is significant in living organisms, as a majority (over 70%) of myostatin in the blood is bound to its propeptide, rendering it inactive. Therefore, the myostatin propeptide acts as a natural inhibitor of myostatin's muscle-suppressing effects.
Description
Recombinant Human Myostatin Propeptide is a protein that is artificially produced but structurally similar to the myostatin propeptide found in humans. It has a molecular weight of 27.8 kilodaltons (kDa) and consists of 244 amino acids, which are the building blocks of proteins.
Physical Appearance
The product is a white powder that has been sterilized by filtration and then freeze-dried to remove moisture.
Formulation
The product is freeze-dried without any additional substances.
Solubility
To use the lyophilized (freeze-dried) Myostatin Propeptide, it is recommended to dissolve it in a sterile solution of 20mM hydrochloric acid (HCl) at a concentration of 0.1mg/ml. This solution can be further diluted with other aqueous solutions as needed.
Stability
To ensure the longevity of the lyophilized protein, it is best to store it at -20°C. Once the protein is reconstituted, it is advisable to divide it into smaller portions to prevent repeated freezing and thawing, which can degrade the protein. The reconstituted protein can be stored at 4°C for a short period.
Purity
The purity of the protein is greater than 95%, as determined by SDS-PAGE, a technique used to separate proteins based on their size.
Biological Activity
The protein demonstrates full biological activity when compared to a standard sample. Its activity is assessed by its ability to inhibit the effects of 50 nanograms per milliliter (ng/ml) of Myostatin on MPC-11 cells, which are muscle cells. The typical effective concentration ranges from 0.13 to 0.2 micrograms per milliliter (µg/ml).
Synonyms
GDF-8, MSTN, Growth Differentiation Factor 8, MSTN Muscle Hypertrophy.
Source
Escherichia Coli.
Amino Acid Sequence
MNENSEQKE NVEKEGLCNA CTWRQNTKSS RIEAIKIQIL SKLRLETAPN ISKDVIRQLL PKAPPLRELI DQYDVQRDDS SDGSLEDDDY HATTETIITM PTESDFLMQV DGKPKCCFFK FSSKIQYNKV VKAQLWIYLR PVETPTTVFV QILRLIKPMK DGTRYTGIRS LKLDMNPGTG IWQSIDVKTV LQNWLKQPES NLGIEIKALD ENGHDLAVTF PGPGEDGLNP FLEVKVTDTP KRSRR.

Q&A

What is myostatin propeptide and how does it function in human physiology?

Myostatin propeptide is a 243-amino acid glycoprotein containing one N-glycosylation site that naturally suppresses myostatin signaling. In human serum, more than 70% of myostatin circulates bound to its propeptide in a latent complex . This binding prevents myostatin from interacting with its receptor (activin receptor IIB), thereby inhibiting myostatin's negative regulation of muscle growth . The propeptide is crucial for proper folding of the myostatin protein and serves as a key endogenous regulator of skeletal muscle mass . When myostatin propeptide binds to active myostatin, it prevents receptor activation, ultimately leading to increased muscle mass development and reduced body fat . This regulatory mechanism explains why experimental overexpression of the propeptide in animal models consistently produces muscle hypertrophy effects .

How does myostatin propeptide differ structurally and functionally from other myostatin inhibitors?

Myostatin propeptide is part of the native myostatin complex but differs from other inhibitors like follistatin in structure and binding mechanisms. Unlike follistatin, which is an extracellular cysteine-rich glycoprotein structurally dissimilar to TGF-β family members, the propeptide is derived from the same precursor protein as myostatin itself . Both molecules inhibit myostatin by preventing receptor binding, but through different molecular interactions . The myostatin propeptide binds non-covalently to the N-terminus of myostatin, forming a latent complex that requires proteolytic activation to release functional myostatin . In contrast, follistatin binds directly to mature myostatin in circulation. The FLRG (follistatin-related gene) protein represents another regulatory molecule that directly interacts with mature myostatin to inhibit its activity . These distinct inhibitory mechanisms provide multiple regulatory checkpoints for myostatin activity in vivo, with the propeptide serving as the primary physiological regulator (binding >70% of circulating myostatin) .

What signaling pathways are affected by myostatin propeptide administration?

Myostatin propeptide administration disrupts the normal Smad signaling pathway that mediates myostatin's growth-inhibitory effects. By binding myostatin, the propeptide prevents activation of the ActRIIB receptor and subsequent Smad2/3 phosphorylation . This interruption has cascading effects on several downstream targets important for myogenesis. Specifically, myostatin normally downregulates MyoD expression, but propeptide-mediated inhibition allows increased MyoD activity . Similarly, propeptide administration indirectly influences Pax3 expression, which is typically suppressed by myostatin but becomes upregulated when myostatin is inhibited . The propeptide's effects also interact with feedback control mechanisms involving Smad6 and Smad7, which normally compete for binding to activin type I receptors . When myostatin signaling is blocked by propeptide binding, this alters the expression of these antagonistic Smads, further modifying the cellular response to TGF-β family members. These signaling changes collectively promote myoblast proliferation and differentiation, driving muscle hypertrophy.

What are the most effective protocols for administering recombinant myostatin propeptide in animal models?

Based on rodent studies, the most effective administration protocol for recombinant myostatin propeptide involves periodic injections at a dosage of 20 mg/kg body weight. In mice, this dosage produced optimal muscle mass increases of more than 15% after three injections over a 15-day period . Higher dosages (50 mg/kg) did not yield additional benefits, suggesting a saturation effect . The recommended injection schedule involves treatments every 5 days (days 0, 5, and 10) to maintain effective propeptide levels . For fibula osteotomy models where both muscle and bone healing are assessed, this protocol significantly enhances tissue repair when compared to vehicle controls .

When designing administration studies, researchers should consider:

  • Species-specific dose adjustments (10 mg/kg was sufficient for serum detection in rats)

  • Timing of sample collection (24, 48, and 168 hours post-administration)

  • Route of administration (intraperitoneal or subcutaneous injections are most common)

  • Propeptide formulation (ensuring proper protein folding and stability)

  • Collection of both urine and serum samples to assess biodistribution and clearance

This protocol has been effective in multiple experimental contexts, though human applications would require additional safety and pharmacokinetic considerations.

How can researchers distinguish between endogenous and exogenous myostatin propeptide in biological samples?

The differentiation between endogenous and exogenous (particularly black market) myostatin propeptide relies on specific biochemical differences. The primary distinguishing feature is glycosylation - endogenous human myostatin propeptide contains characteristic glycosylation patterns at its N-glycosylation site, while exogenous forms produced in E. coli expression systems lack this post-translational modification . This difference creates detectable variations in both molecular mass and isoelectric point (pI).

The recommended analytical workflow for differentiation includes:

  • Immunoaffinity purification from serum or urine samples using specific antibodies against myostatin propeptide

  • Separation via electrophoretic techniques:

    • SDS-PAGE to detect molecular mass differences

    • IEF-PAGE to identify variations in isoelectric point

  • Detection through Western blotting using antibodies specific to myostatin propeptide

This methodology exploits the structural differences between glycosylated endogenous propeptide and non-glycosylated recombinant versions, providing a reliable approach for identifying exogenous administration. The technique has been validated in both animal models and analysis of black market products, making it suitable for both research and anti-doping applications .

What are the critical considerations when designing dose-response studies for myostatin propeptide?

When designing dose-response studies for myostatin propeptide, researchers must consider several critical factors to ensure meaningful results:

  • Dose Range Selection: Based on previous studies, an effective dose range should include at least three concentrations (e.g., 10 mg/kg, 20 mg/kg, and 50 mg/kg) to establish the response curve . The inclusion of a 0 mg/kg control group is essential for baseline comparisons.

  • Measurement Timepoints: Both acute responses (24-48 hours) and sustained effects (7+ days) should be assessed, as the pharmacokinetics of propeptide may differ from its physiological effects .

  • Sex-Specific Responses: Studies should include both male and female subjects, as previous research has noted potential differences in response magnitude between sexes .

  • Outcome Measurements:

    • Primary measurements: Changes in muscle mass (both absolute and relative to body weight)

    • Secondary measurements: Muscle fiber cross-sectional area, protein synthesis rates, fat mass changes

    • Functional outcomes: Strength, endurance, or recovery parameters

  • Sample Collection Protocol: Collection of multiple tissue types (blood, urine, muscle biopsies) allows for correlation between circulating propeptide levels and physiological responses .

  • Propeptide Formulation Control: Source verification and quality control testing of the propeptide are crucial, as variations in propeptide structure can significantly impact efficacy .

  • Statistical Considerations: Power analysis should account for expected effect sizes (~15% change in muscle mass at optimal dosing) to determine appropriate sample sizes .

Properly designed dose-response studies are essential for establishing therapeutic windows and avoiding potential side effects when translating findings toward clinical applications.

What are the current gold standard methods for detecting myostatin propeptide in human biological samples?

The gold standard for detecting myostatin propeptide in human biological samples involves a multi-step analytical approach combining immunoaffinity techniques with electrophoretic separation and immunochemical detection. The currently established protocol includes:

  • Sample Preparation:

    • For serum/plasma: Direct immunoaffinity purification using antibodies specific to myostatin propeptide

    • For urine: Concentration steps followed by immunoaffinity purification

  • Separation Techniques:

    • SDS-PAGE: Provides molecular weight-based separation, allowing detection of mass differences between glycosylated endogenous propeptide and non-glycosylated exogenous forms

    • IEF-PAGE: Separates proteins based on isoelectric point, providing an orthogonal identification method particularly useful for differentiating endogenous from E. coli-expressed propeptide

  • Detection Methods:

    • Western blotting using validated antibodies against myostatin propeptide

    • Visualization using chemiluminescence or fluorescence-based detection systems

  • Confirmation Strategies:

    • Mass spectrometry analysis of isolated propeptide to verify amino acid sequence and post-translational modifications

    • Functional assays to confirm biological activity of detected propeptide

This methodology effectively detects both endogenous and exogenous propeptide in biological matrices and can distinguish between them based on glycosylation status. The detection limits are typically in the ng/mL range for serum samples, consistent with physiological concentrations . Ongoing research continues to refine these methods, particularly for improving sensitivity in urine samples where concentrations may be lower.

How can researchers overcome analytical challenges in quantifying active versus latent myostatin propeptide complexes?

Quantifying active versus latent myostatin propeptide complexes presents significant analytical challenges that researchers can address through specialized techniques:

  • Differential Extraction Approaches:

    • Use of acid activation protocols to dissociate the latent complex prior to analysis, allowing measurement of total myostatin capacity

    • Parallel sample processing with and without acid treatment to determine the proportion of free versus complexed myostatin

  • Immunoprecipitation Strategies:

    • Sequential immunoprecipitation using antibodies specific to different complex components (propeptide, mature myostatin, FLRG)

    • Co-immunoprecipitation followed by Western blotting to identify interaction partners

  • Reporter Gene Assays:

    • Functional bioassays using cell lines expressing myostatin-responsive elements coupled to reporter genes

    • These assays can detect only biologically active myostatin, helping distinguish between active and latent forms

  • Chromatographic Separation:

    • Size-exclusion chromatography to separate free propeptide from propeptide-myostatin complexes

    • Affinity chromatography using immobilized ActRIIB receptor domains to capture only forms capable of receptor binding

  • Mass Spectrometry Applications:

    • Targeted mass spectrometry using selective reaction monitoring for specific peptides representing different complex states

    • Crosslinking mass spectrometry to characterize protein-protein interactions within the complex

  • Standardization Approaches:

    • Development of recombinant standards representing both free propeptide and propeptide-myostatin complexes

    • Use of isotopically labeled internal standards for absolute quantification

These methodological approaches collectively provide researchers with tools to distinguish between the different physiological states of myostatin propeptide, enabling more accurate interpretation of its biological activity in experimental contexts.

What are the limitations of current electrophoretic methods for myostatin propeptide detection?

Current electrophoretic methods for myostatin propeptide detection, while effective for many research applications, present several important limitations that researchers should consider:

  • Sensitivity Constraints:

    • Detection limits in the ng/mL range may be insufficient for detecting subtle changes in propeptide levels, particularly in urine samples

    • Limited sensitivity for detecting low concentrations that may still have biological significance

  • Specificity Challenges:

    • Cross-reactivity of antibodies used in Western blotting may lead to false positives, particularly with other TGF-β family members

    • Difficulty distinguishing between closely related propeptide variants or degradation products

  • Glycoform Heterogeneity:

    • Natural variation in glycosylation patterns of endogenous propeptide can complicate interpretation of mobility differences

    • Potential for misclassification of partially glycosylated endogenous forms as exogenous

  • Quantitative Limitations:

    • Semi-quantitative nature of Western blotting makes precise concentration determination challenging

    • Variability in transfer efficiency during blotting can affect quantitative accuracy

  • Throughput Restrictions:

    • Labor-intensive protocols limit sample processing capacity

    • Lengthy processing time (typically 1-2 days from sample preparation to results)

  • Sample Requirements:

    • Relatively large sample volumes needed for immunoaffinity purification

    • Limited stability of propeptide during extended processing steps

  • Method Standardization:

    • Lack of universally standardized protocols leading to inter-laboratory variability

    • Limited availability of certified reference materials for calibration

To address these limitations, researchers are increasingly exploring complementary techniques such as liquid chromatography coupled with tandem mass spectrometry (LC-MS/MS), which offers improved specificity and potential for absolute quantification, and developing enhanced immunoassay platforms with greater sensitivity and throughput capacity.

How does myostatin propeptide administration affect muscle regeneration following injury?

Myostatin propeptide administration significantly enhances muscle regeneration following injury through multiple mechanisms. In mouse models with fibula osteotomy and associated damage to lateral compartment muscles, administration of recombinant myostatin propeptide at 20 mg/kg substantially improved muscle healing outcomes . The propeptide's effects on muscle regeneration include:

  • Reduced Fibrosis Development: Myostatin normally promotes fibroblast proliferation and collagen deposition after injury. Propeptide administration inhibits this process, leading to decreased scar tissue formation at the injury site .

  • Enhanced Satellite Cell Activation: By blocking myostatin's inhibitory effects on satellite cell proliferation, the propeptide increases the pool of myogenic progenitor cells available for repair.

  • Accelerated Myofiber Regeneration: Treatment promotes faster formation of new myofibers with larger cross-sectional areas, facilitating functional recovery .

  • Improved Neuromuscular Junction Reinnervation: Evidence suggests propeptide treatment may support better reestablishment of neuromuscular connections in regenerating muscle.

  • Concurrent Enhancement of Bone Repair: In models with combined muscle and bone injury, propeptide administration shows benefits for both tissues, suggesting coordinated healing mechanisms .

The timing of administration appears critical, with early intervention (day of injury, followed by treatments at 5 and 10 days post-injury) showing optimal results . These findings indicate potential therapeutic applications for myostatin propeptide in clinical scenarios involving traumatic muscle injury, surgical recovery, or conditions requiring enhanced muscle regeneration.

What is the relationship between myostatin propeptide and other regulators of muscle growth in experimental models?

Myostatin propeptide functions within a complex regulatory network that controls muscle growth, interacting with multiple signaling pathways and growth factors. Key relationships include:

This interconnected regulatory network explains why experimental models often show variable responses to propeptide administration depending on the background expression of these other factors, highlighting the importance of considering the broader signaling context in experimental design.

What methodological approaches can distinguish therapeutic from performance-enhancing applications in myostatin propeptide research?

Distinguishing therapeutic from performance-enhancing applications in myostatin propeptide research requires careful methodological considerations that address both scientific and ethical dimensions:

  • Subject Selection Parameters:

    • Therapeutic research should focus on populations with muscle-wasting conditions, injuries, or age-related sarcopenia

    • Studies should establish clear inclusion criteria based on quantifiable deficits in muscle function or mass

    • Control groups should match pathological conditions rather than comparing to healthy baselines

  • Outcome Measure Differentiation:

    • Therapeutic endpoints should emphasize restoration of function rather than enhancement beyond normal ranges

    • Measures should include quality-of-life assessments, activities of daily living, and disease-specific functional metrics

    • Performance studies typically focus on strength, power, or athletic performance parameters in already healthy subjects

  • Dosing Strategy Considerations:

    • Therapeutic protocols should establish minimum effective doses to achieve clinical improvement

    • Dose-finding studies should include careful safety monitoring and stopping criteria

    • Assessment of dose-response relationships should focus on plateaus in therapeutic benefit rather than maximum possible effect

  • Mechanistic Investigation Approaches:

    • Therapeutic research should include molecular and cellular analyses connecting propeptide effects to specific disease mechanisms

    • Biomarker development to predict and monitor therapeutic responses

    • Comparative studies with approved therapies for the target condition

  • Ethical Research Framework:

    • Clear regulatory compliance with appropriate oversight for human studies

    • Transparent reporting of both positive and negative findings

    • Development of detection methods in parallel with therapeutic applications

  • Translational Research Design:

    • Step-wise progression from disease models to clinical applications

    • Early engagement with regulatory authorities regarding therapeutic classification

    • Development of administration protocols that minimize potential for misuse

These methodological approaches help create clear boundaries between legitimate therapeutic research and potentially problematic performance enhancement applications, supporting the responsible advancement of myostatin propeptide as a potential treatment while addressing concerns about misuse in sports or cosmetic bodybuilding contexts.

What detection windows exist for myostatin propeptide in anti-doping testing protocols?

The detection windows for myostatin propeptide in anti-doping contexts remain under investigation, with current evidence primarily derived from animal studies. Based on research conducted with rats administered black market myostatin propeptide, the following detection parameters have been established:

  • Serum Detection Window:

    • Exogenous propeptide is clearly detectable in serum for at least 24-48 hours following administration

    • Levels diminish significantly by 168 hours (7 days) post-administration

    • Detection sensitivity is influenced by the initial dosage (10 mg/kg body weight in rat studies)

  • Urine Detection Parameters:

    • Urinary excretion patterns are less well-characterized than serum profiles

    • Current research aims to determine if propeptide can be reliably detected in urine samples

    • The large molecular weight of propeptide (approximately 25 kDa) may limit urinary excretion

  • Analytical Sensitivity Factors:

    • Detection methods using electrophoresis (SDS-PAGE) and Western blotting can distinguish exogenous propeptide from endogenous forms based on glycosylation differences

    • These methods have sensitivity in the nanogram range for serum samples

  • Timing Considerations for Testing:

    • Optimal detection would occur within 48 hours of administration

    • Strategic testing during competition versus out-of-competition testing approaches must account for these windows

It's important to note that these parameters are based on animal models, and human pharmacokinetics may differ . The World Anti-Doping Agency (WADA) continues to refine testing approaches for myostatin propeptide as part of its prohibited substances program under category S4 (Hormone and Metabolic Modulators) .

How can researchers address ethical concerns when studying black market myostatin propeptide products?

Researchers face significant ethical challenges when studying black market myostatin propeptide products. A comprehensive ethical framework includes:

  • Alternative Model Systems:

    • Animal studies (particularly rat models) should be used instead of human administration of unregulated products

    • In vitro characterization of black market products before any in vivo testing

    • Development of humanized animal models where appropriate

  • Product Acquisition Protocol:

    • Establishment of transparent, documented procedures for obtaining black market products

    • Collaboration with regulatory authorities to ensure legal compliance

    • Avoidance of financial transactions that might support illegal markets

  • Analytical Characterization Requirements:

    • Comprehensive chemical analysis of product composition and purity

    • Assessment for contaminants, microbial contamination, and heavy metals

    • Comparison with pharmaceutical-grade reference standards

  • Risk Mitigation Strategies:

    • Implementation of strict laboratory safety protocols for handling unknown substances

    • Limited dosing studies designed to minimize animal numbers while obtaining necessary data

    • Clear stopping criteria based on adverse events

  • Transparency and Reporting Practices:

    • Complete disclosure of methods and findings in publications

    • Reporting of all safety concerns to appropriate regulatory bodies

    • Publication of negative or concerning results, not just positive findings

  • Educational Component:

    • Integration of research findings into educational materials about health risks

    • Communication strategies for disseminating safety information to target populations

    • Engagement with sports organizations and healthcare providers

This ethical framework allows researchers to generate valuable scientific knowledge about black market products while maintaining scientific integrity and minimizing potential harms. Such research is crucial for effective anti-doping efforts and protecting public health .

What documentation and controls are necessary for myostatin propeptide research to meet institutional and regulatory requirements?

Conducting compliant myostatin propeptide research requires comprehensive documentation and controls to satisfy institutional review boards, animal care committees, and regulatory agencies:

  • Regulatory Classification Documentation:

    • Confirmation of myostatin propeptide's status under WADA regulations (S4 category - Hormone and Metabolic Modulators)

    • Documentation of applicable national regulatory frameworks (e.g., controlled substance status)

    • Institutional position statements on research involving prohibited substances

  • Source and Quality Control Requirements:

    • Certificate of analysis for recombinant propeptide standards

    • Full characterization documentation (purity, activity, endotoxin levels)

    • Chain of custody records for all propeptide material

    • For black market products, documentation of analytical verification procedures

  • Experimental Protocol Controls:

    • Detailed administration protocols with justification for doses

    • Standard operating procedures for sample collection and processing

    • Blinding procedures to minimize bias in outcome assessments

    • Randomization protocols for subject/animal assignment

  • Ethics Committee Submissions:

    • Risk-benefit analysis specific to myostatin propeptide research

    • Justification for animal models with power calculations

    • Specific safety monitoring plans including stopping criteria

    • For human studies (when applicable), enhanced informed consent processes

  • Security and Accounting Measures:

    • Secure storage protocols for propeptide materials

    • Inventory management system with reconciliation procedures

    • Access restrictions to authorized personnel only

    • Disposal documentation for unused materials

  • Reporting Requirements:

    • Adverse event reporting procedures

    • Regular progress reports to oversight committees

    • Publication plans addressing negative and positive findings

    • Data sharing protocols in compliance with institutional policies

  • Laboratory Safety Documentation:

    • Risk assessments for handling procedures

    • Personnel training records

    • Emergency response procedures

    • Personal protective equipment specifications

These comprehensive documentation and control requirements ensure regulatory compliance while supporting scientific integrity and reproducibility in myostatin propeptide research. They also facilitate transparent communication with oversight bodies and the broader scientific community.

What methodological approaches can address inter-species variations in myostatin propeptide structure and function?

Addressing inter-species variations in myostatin propeptide research requires sophisticated methodological approaches to ensure translational relevance:

  • Comparative Sequence Analysis Techniques:

    • Phylogenetic analysis of propeptide sequences across species to identify conserved functional domains

    • Structure-function mapping using site-directed mutagenesis to determine critical regions

    • Computational prediction of species-specific post-translational modifications, particularly glycosylation patterns

  • Cross-Species Binding Affinity Studies:

    • Surface plasmon resonance (SPR) assays comparing propeptide-myostatin binding kinetics across species

    • Competition assays testing cross-reactivity of propeptides with myostatin from different species

    • Isothermal titration calorimetry to quantify thermodynamic binding parameters

  • Humanized Animal Model Development:

    • Generation of transgenic models expressing human myostatin and propeptide

    • Knockin strategies replacing endogenous propeptide with human sequence

    • Chimeric propeptide designs incorporating human domains into animal backbones

  • Scaling Approaches for Dosing Translation:

    • Allometric scaling calculations accounting for species-specific differences in metabolism

    • Pharmacokinetic/pharmacodynamic (PK/PD) modeling incorporating species variation

    • Physiologically-based pharmacokinetic (PBPK) modeling for human dose prediction

  • Comparative Cellular Response Assays:

    • Parallel testing in primary myoblasts from multiple species

    • Species-specific reporter systems for downstream signaling pathways

    • Cross-species transcriptomic analysis to identify conserved response patterns

  • Glycobiology-Focused Techniques:

    • Glycoproteomic analysis of propeptide from different species

    • Engineering of recombinant propeptides with species-appropriate glycosylation

    • Functional assessment of differentially glycosylated propeptide variants

These methodological approaches collectively provide a framework for translating findings between species while acknowledging inherent biological differences. Researchers should explicitly address these considerations when designing studies and interpreting results to enhance translational validity of myostatin propeptide research.

How can researchers accurately model the complex interactions between myostatin propeptide and other regulatory proteins?

Modeling the complex interactions between myostatin propeptide and its regulatory network requires multi-dimensional approaches that capture both structural relationships and functional consequences:

  • Integrated Structural Biology Approaches:

    • X-ray crystallography of propeptide-myostatin complexes and receptor interactions

    • Cryo-electron microscopy for visualization of larger multiprotein assemblies

    • NMR spectroscopy to analyze dynamic interactions in solution

    • Molecular dynamics simulations to predict conformational changes upon binding

  • Systems Biology Frameworks:

    • Mathematical modeling of the propeptide-myostatin-follistatin-FLRG network

    • Differential equation-based models incorporating binding kinetics and concentration-dependent effects

    • Sensitivity analysis to identify critical nodes in the regulatory network

    • Agent-based modeling of tissue-level responses to propeptide administration

  • Proteomics-Based Interaction Mapping:

    • Proximity labeling techniques (BioID, APEX) to identify novel interaction partners

    • Cross-linking mass spectrometry to capture transient interactions

    • Co-immunoprecipitation coupled with quantitative proteomics to measure binding stoichiometry

    • Protein microarrays to screen for additional binding partners

  • Live-Cell Imaging Techniques:

    • FRET/BRET sensors to monitor propeptide-myostatin interactions in real-time

    • Fluorescence correlation spectroscopy to measure binding dynamics in living cells

    • Photo-switchable fluorescent tags to track protein complex formation and dissociation

  • Functional Genomics Integration:

    • CRISPR-based screens to identify genes modulating propeptide function

    • RNA-seq analysis following propeptide administration to map transcriptional networks

    • ChIP-seq to identify downstream transcription factor binding events

  • Multi-Scale Modeling Approaches:

    • Integration of molecular, cellular, and tissue-level models

    • Pharmacokinetic/pharmacodynamic (PK/PD) modeling incorporating multiple interaction partners

    • Machine learning algorithms trained on experimental datasets to predict complex interactions

These complementary approaches allow researchers to construct comprehensive models of how myostatin propeptide functions within its broader regulatory context, enabling more accurate predictions of experimental outcomes and potential therapeutic effects across different physiological states.

What are the current limitations in translating animal model findings on myostatin propeptide to human applications?

Translating myostatin propeptide research from animal models to human applications faces several significant limitations that researchers must address:

  • Structural and Functional Differences:

    • Species-specific variations in myostatin propeptide amino acid sequence and glycosylation patterns

    • Differences in binding affinity between propeptide and myostatin across species

    • Potential variations in downstream signaling pathway sensitivity

  • Pharmacokinetic Challenges:

    • Limited human data on propeptide half-life, tissue distribution, and elimination

    • Uncertainty about appropriate dosing schedules based on animal studies

    • Potential immunogenicity of recombinant propeptides in humans

  • Regulatory and Production Hurdles:

    • Absence of pharmaceutical-grade propeptide products for human use

    • Complex regulatory pathway for approval of novel biologic agents

    • Challenges in cost-effective, large-scale production with consistent glycosylation

  • Methodological Limitations:

    • Ethical constraints on human administration studies, particularly with black market products

    • Difficulty establishing therapeutic index in humans based on animal data

    • Limited longitudinal data on long-term effects in any species

  • Biomarker and Monitoring Challenges:

    • Need for validated biomarkers to assess efficacy in human subjects

    • Technical challenges in detecting exogenous versus endogenous propeptide in humans

    • Complexity of measuring functional outcomes in clinical settings

  • Heterogeneity Factors:

    • Potential variations in response based on genetic background, age, and sex

    • Comorbidities in target human populations that are not represented in animal models

    • Environmental factors affecting myostatin regulation in humans

  • Ethical and Anti-Doping Considerations:

    • Dual-use potential for both therapeutic and performance-enhancing applications

    • Need for parallel development of detection methods for anti-doping purposes

    • Ethical frameworks for clinical trials involving substances with performance-enhancing potential

Addressing these limitations requires integrated research approaches that combine preclinical animal studies with careful human biomarker studies, development of humanized animal models, and targeted clinical investigations in appropriate patient populations rather than healthy subjects to maintain clear therapeutic focus.

What emerging technologies might enhance the specificity and sensitivity of myostatin propeptide detection?

Several emerging technologies hold promise for revolutionizing myostatin propeptide detection with improved specificity and sensitivity:

  • Advanced Mass Spectrometry Approaches:

    • High-resolution parallel reaction monitoring (PRM) for targeted propeptide detection

    • Ion mobility mass spectrometry to distinguish conformational isomers

    • MALDI imaging mass spectrometry for tissue localization studies

    • Nanopore-coupled mass spectrometry for real-time monitoring

  • Aptamer-Based Detection Systems:

    • SELEX (Systematic Evolution of Ligands by Exponential Enrichment) to develop propeptide-specific aptamers

    • Aptamer-based electrochemical sensors for rapid detection

    • Conformational aptamer beacons responsive to propeptide binding

    • Multiplexed aptamer arrays for simultaneous detection of propeptide and related proteins

  • Single-Molecule Detection Platforms:

    • Plasmonic nanobiosensors for label-free propeptide detection

    • Single-molecule FRET assays for propeptide-myostatin interactions

    • Nanopore sensing of individual propeptide molecules

    • Digital ELISA platforms with single-molecule sensitivity

  • Microfluidic Integration:

    • Lab-on-a-chip devices for automated sample preparation and analysis

    • Droplet microfluidics for high-throughput screening

    • Paper-based microfluidic systems for field testing

    • Organ-on-a-chip systems for functional propeptide assessments

  • Glycoproteomic-Specific Technologies:

    • Lectin arrays for glycosylation pattern profiling

    • Intact glycopeptide analysis workflows

    • Glycan-specific imaging agents for tissue distribution studies

    • Glycoform-specific antibodies for improved differentiation between endogenous and exogenous propeptide

  • Computational Enhancement:

    • Machine learning algorithms for pattern recognition in complex spectral data

    • Deep learning approaches for automated Western blot interpretation

    • Predictive models for propeptide metabolism and clearance

    • Network analysis tools for context-dependent propeptide activity assessment

These technologies promise to address current limitations in myostatin propeptide detection , potentially enabling detection at lower concentrations, improved differentiation between endogenous and exogenous forms, faster analysis times, and more comprehensive characterization of propeptide variants and their functional states.

What knowledge gaps remain in understanding the pharmacokinetics of myostatin propeptide in humans?

Despite significant research progress, several critical knowledge gaps persist in understanding myostatin propeptide pharmacokinetics in humans:

  • Absorption and Distribution Parameters:

    • Limited data on bioavailability following different administration routes

    • Incomplete understanding of tissue distribution patterns beyond circulation

    • Unknown extent of blood-brain barrier penetration

    • Lack of information on cellular uptake mechanisms in target tissues

  • Metabolism and Elimination Pathways:

    • Uncharacterized enzymatic processing mechanisms for exogenous propeptide

    • Limited understanding of clearance rates and half-life in humans

    • Insufficient data on renal versus hepatic elimination contributions

    • Unknown metabolite profiles and their potential biological activities

  • Temporal Dynamics:

    • Absence of human time-course data for both serum and tissue concentrations

    • Limited understanding of circadian variations in propeptide pharmacokinetics

    • Unknown duration of biological effects relative to detectable presence

  • Variability Factors:

    • Insufficient data on demographic factors affecting pharmacokinetics (age, sex, ethnicity)

    • Limited understanding of genetic polymorphisms affecting propeptide processing

    • Unknown impact of disease states on absorption, distribution, metabolism, and excretion

    • Limited data on potential drug-drug interactions

  • Formulation Effects:

    • Unknown impact of different delivery systems on bioavailability and tissue targeting

    • Limited data on stability of various formulations in vivo

    • Lack of information on how glycosylation differences affect pharmacokinetic parameters

  • Dose-Response Relationships:

    • Absence of human dose-proportionality data at therapeutically relevant doses

    • Unknown threshold concentrations for biological effects in various tissues

    • Limited understanding of receptor occupancy requirements for efficacy

  • Bioequivalence Parameters:

    • Lack of established bioequivalence criteria for different propeptide formulations

    • Insufficient comparative data between recombinant and black market products

These knowledge gaps highlight the need for carefully designed clinical pharmacokinetic studies in appropriate human populations, development of sensitive bioanalytical methods specific to human matrices, and more sophisticated modeling approaches that can integrate the limited available human data with more extensive animal findings.

How might combination therapies involving myostatin propeptide be designed for maximal efficacy and safety?

Designing optimal combination therapies involving myostatin propeptide requires systematic approaches to leverage complementary mechanisms while mitigating risks:

  • Rational Target Selection Strategies:

    • Combinations with IGF-1 pathway modulators to simultaneously activate anabolic signaling while removing catabolic inhibition

    • Pairing with anti-inflammatory agents to address the inflammatory component of muscle wasting

    • Combination with exercise mimetics to enhance nutrient delivery and satellite cell activation

    • Integration with metabolism modulators to optimize nutrient utilization for muscle growth

  • Temporal Optimization Approaches:

    • Sequential administration protocols based on signaling pathway dynamics

    • Pulsed dosing strategies to prevent receptor desensitization

    • Timed administration relative to exercise or rehabilitation interventions

    • Circadian timing considerations for maximum efficacy

  • Delivery System Integration:

    • Co-delivery platforms ensuring synchronized release of multiple agents

    • Tissue-targeted nanocarriers for localized combination effects

    • Stimuli-responsive systems activated by physiological triggers

    • Biodegradable implants for sustained release of combination therapies

  • Dosing Optimization Methodologies:

    • Response surface modeling to identify optimal dose ratios

    • Adaptive trial designs with pharmacokinetic/pharmacodynamic (PK/PD) feedback

    • Model-informed precision dosing accounting for patient-specific factors

    • Step-wise dose-finding protocols with safety monitoring

  • Safety Enhancement Strategies:

    • Selective tissue targeting to minimize systemic effects

    • Incorporation of molecular "brakes" to prevent excessive muscle growth

    • Real-time biomarker monitoring to detect early signs of adverse events

    • Development of specific antagonists as rescue agents if needed

  • Condition-Specific Combinations:

    • For cachexia: Propeptide + appetite stimulants + nutritional support

    • For sarcopenia: Propeptide + vitamin D + resistance exercise program

    • For muscular dystrophy: Propeptide + anti-fibrotic agents + gene therapy approaches

    • For injury recovery: Propeptide + growth factors for coordinated muscle-bone healing

  • Regulatory and Development Considerations:

    • Co-development pathways for novel combination products

    • Biomarker strategies to demonstrate synergistic effects

    • Clear differentiation from performance enhancement applications

    • Fixed-dose versus flexible combination approaches based on individual response

These design principles provide a framework for developing myostatin propeptide combination therapies that maximize therapeutic potential while maintaining acceptable safety profiles for specific patient populations and conditions.

What are the most promising near-term research directions for myostatin propeptide in human applications?

The most promising near-term research directions for myostatin propeptide focus on targeted therapeutic applications with clear clinical need, improved detection methodologies, and enhanced understanding of molecular mechanisms:

  • Clinical Applications in Muscle-Wasting Disorders:

    • Investigation of propeptide therapy for cancer cachexia, where rapid intervention is critical

    • Evaluation in age-related sarcopenia with functional endpoints like fall prevention

    • Application in disuse atrophy following prolonged hospitalization or immobilization

    • Combination approaches with standard-of-care treatments for muscular dystrophies

  • Detection Method Refinement:

    • Development of highly sensitive, high-throughput assays for clinical monitoring

    • Validation of biomarker panels predicting response to propeptide treatment

    • Improved differentiation between endogenous and exogenous propeptide in biological samples

    • Standardization of testing protocols for broader implementation

  • Targeted Delivery Systems:

    • Muscle-specific delivery platforms to minimize off-target effects

    • Extended-release formulations for reduced dosing frequency

    • Local delivery approaches for injury-specific applications

    • Novel propeptide derivatives with enhanced stability and bioavailability

  • Mechanism Elucidation:

    • Detailed mapping of propeptide-myostatin binding interfaces

    • Investigation of crosstalk between propeptide effects and other signaling pathways

    • Studies of propeptide's impact on muscle stem cell dynamics during regeneration

    • Exploration of non-muscle effects in metabolic regulation

  • Translational Model Development:

    • Establishment of humanized animal models for more predictive preclinical testing

    • Development of ex vivo human tissue models for efficacy and safety screening

    • Validation of imaging biomarkers for non-invasive monitoring of propeptide effects

    • Creation of patient-derived systems for personalized response prediction

  • Therapeutic Optimization:

    • Dose-finding studies establishing minimum effective doses for specific conditions

    • Identification of responder/non-responder characteristics through biomarker profiling

    • Development of companion diagnostics for patient selection

    • Exploration of propeptide variants with improved therapeutic indices

Product Science Overview

Introduction

Myostatin, also known as Growth Differentiation Factor 8 (GDF-8), is a member of the Transforming Growth Factor-beta (TGF-β) superfamily. It is a potent and specific negative regulator of skeletal muscle mass. Myostatin Propeptide (Human Recombinant) is a recombinant form of the propeptide that binds and inhibits myostatin, thereby regulating muscle growth.

Structure and Function

Recombinant human Myostatin Propeptide is a 27.8 kDa protein consisting of 244 amino acid residues . The propeptide binds to myostatin in vitro and in vivo, with over 70% of myostatin in serum bound to its propeptide . This binding inhibits myostatin’s activity, preventing it from exerting its muscle growth-inhibiting effects.

Production and Purification

The recombinant human Myostatin Propeptide is typically produced in E. coli and is available as a sterile filtered white lyophilized (freeze-dried) powder . The protein is purified using various techniques, including UV spectroscopy, RP-HPLC, and SDS-PAGE gels, to ensure high purity and activity .

Biological Activity

The biological activity of Myostatin Propeptide is determined by its ability to inhibit myostatin. It has been shown to inhibit 50 ng/mL of myostatin on MPC-11 cells at concentrations typically ranging from 0.01 to 0.04 μg/mL . This inhibition is crucial for research into muscle growth and muscle-wasting conditions.

Applications

Myostatin Propeptide (Human Recombinant) is used primarily in research settings to study muscle growth regulation and potential therapeutic applications for muscle-wasting diseases. It is not intended for human, animal, or diagnostic applications .

Storage and Stability

The lyophilized product is stable at -20°C. Once reconstituted, it should be aliquoted and frozen at -20°C with a carrier protein (0.1% HSA or BSA) for long-term storage . This ensures the protein remains active and stable for extended periods.

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.