MTPN Human

Myotrophin Human Recombinant
Shipped with Ice Packs
In Stock

Description

Introduction to MTPN Human

MTPN (Myotrophin), encoded by the MTPN gene, is a 15 kDa ankyrin repeat protein critical for cardiac hypertrophy, neuronal differentiation, and cytoskeletal regulation. Its role in activating NF-κB signaling and modulating actin dynamics underscores its significance in cardiovascular and neurodevelopmental processes . Below is a detailed analysis of its molecular structure, biological functions, and research findings.

Molecular Structure and Properties

MTPN is a non-glycosylated polypeptide containing 138 amino acids (1–118 residues) with a His-Tag fusion for purification. Key structural and biochemical properties include:

PropertyDescription
Molecular Mass15 kDa (recombinant form)
Amino Acid SequenceMGSSHHHHHHSSGLVPRGSHMCDKEFMWALKNGDLDEVKDYVAKGEDVNRTLEGGRKPLHYAADCGQLEILEFLLLKGADINAPDKHHITPLLSAVYEGHVSCVKLLLSKGADKTVKGPDGLTAFEATDNQAIKALLQ
SourceE. coli-derived recombinant protein
Formulation20 mM Tris-HCl (pH 8) + 10% glycerol
Purity>90% (SDS-PAGE-verified)

Biological Functions and Interactions

MTPN influences cellular processes through distinct pathways:

Cardiac Hypertrophy and NF-κB Signaling

MTPN stimulates cardiomyocyte growth and hypertrophy by activating NF-κB, a transcription factor regulating stress-responsive genes . Overexpression in transgenic mice leads to myocardial hypertrophy and heart failure, highlighting its pathogenic role in cardiovascular diseases .

Actin Dynamics and Cytoskeletal Regulation

MTPN binds to the F-actin capping protein complex (CAPZA1/CAPZB), inhibiting its activity to regulate actin filament growth. This interaction is critical for maintaining cytoskeletal integrity .

Neurodevelopmental Roles

In the cerebellum, MTPN promotes granule cell differentiation, suggesting a role in neuronal morphogenesis .

Protein Interactions

Key interactors include:

ProteinInteraction TypeFunctional Impact
RELADirect binding NF-κB dimerization and transcriptional activation
CAPZA1/CAPZBCompetitive inhibition Actin filament elongation regulation
TAP (Transporter associated with Antigen Processing)Indirect association Modulation of MHC class I peptide loading (species-specific)

Cardiac Pathophysiology

  • Hypertrophy and Failure: Elevated MTPN levels are observed in human dilated cardiomyopathy and ischemic hearts . Transgenic overexpression in mice induces hypertrophy and heart failure .

  • p53-Mediated Transition to Failure: Chronic MTPN-induced hypertrophy triggers p53 activation, contributing to maladaptive remodeling .

Species-Specific Effects in Immune Function

MTPN influences MHC class I peptide loading in a species-dependent manner:

SpeciesMTPN RoleOutcome
HumanRecruits HC/TAP complexes for peptide stabilizationOptimal Bw4 epitope expression
MousePoor HC recruitment in human TAP complexesSuboptimal peptide binding and stability

Neurological and Oncological Links

  • Cerebellar Development: MTPN regulates granule cell differentiation, suggesting involvement in neurodevelopmental disorders .

  • Cancer Association: While not a direct oncogene, MTPN’s role in stress signaling may intersect with tumor progression pathways .

Experimental Applications and Reagents

MTPN is widely used in research, with recombinant proteins and antibodies enabling functional studies:

ReagentApplicationSource
Recombinant MTPNNF-κB activation assays, actin dynamicsProSpecBio , HelloBio
MTPN Antibody (DF14809)Western blot, immunofluorescenceAffinity Biosciences
Single-Cell RNA DataTissue-specific expression profilingHuman Protein Atlas

Product Specs

Introduction
Myotrophin (MTPN) is an ankyrin repeat protein that promotes cardiac hypertrophy. It achieves this by stimulating protein synthesis and cardiomyocyte growth, ultimately activating the NF-kappaB signaling cascade. MTPN also plays a crucial role in cerebellar development, contributing to its morphogenesis and the differentiation of cerebellar neurons, particularly granule cells. Elevated levels of MTPN are observed in individuals with dilated cardiomyopathy and ischemic heart conditions.
Description
Recombinant Human MTPN, expressed in E. coli, is a single, non-glycosylated polypeptide chain. It consists of 138 amino acids, with 118 amino acids forming the MTPN sequence (1-118 a.a.) and a 20 amino acid His-Tag at the N-terminus. This protein has a molecular weight of 15 kDa and is purified using proprietary chromatographic techniques.
Physical Appearance
The product is a sterile, colorless solution that has been filtered for clarity.
Formulation
The formulation for Human MTPN consists of a solution containing 20mM Tris-HCl at pH 8 and 10% glycerol.
Stability
For short-term storage (2-4 weeks), the product should be kept at 4°C. For long-term storage, it is recommended to freeze the product at -20°C. To ensure stability during long-term storage, consider adding a carrier protein such as 0.1% HSA or BSA. Avoid repeated freeze-thaw cycles.
Purity
The purity of this product is greater than 90.0%, as determined by SDS-PAGE analysis.
Synonyms
Protein V-1, GCDP, Myotrophin, FLJ31098, FLJ99857.
Source
Escherichia Coli.
Amino Acid Sequence

MGSSHHHHHH SSGLVPRGSH MCDKEFMWAL KNGDLDEVKD YVAKGEDVNR TLEGGRKPLH YAADCGQLEI LEFLLLKGAD INAPDKHHIT PLLSAVYEGH VSCVKLLLSK GADKTVKGPD GLTAFEATDN QAIKALLQ.

Q&A

What is human MTPN and what are its fundamental structural properties?

Human myotrophin (MTPN) is a small protein encoded by the MTPN gene and is also known as protein V-1 in some literature. It is characterized by its relatively compact structure with 118 amino acids. MTPN contains several key structural domains that facilitate its interactions with other cellular proteins, particularly in cardiac and neuronal tissues. The protein contains multiple lysine residues that are targets for post-translational modifications, which significantly influence its function and localization within cells .

The structural analysis of MTPN reveals distinct binding domains that enable its participation in various cellular signaling cascades. These structural characteristics are conserved across species, though human MTPN has specific sequence variations that differentiate it from orthologs in model organisms such as mice .

What post-translational modifications are observed in human MTPN?

Human MTPN undergoes extensive post-translational modifications that regulate its function and interactions. Based on proteomic analyses, the following modifications have been identified:

SitePTM TypeEnzyme/MethodEvidence Source
K4Acetylation-PhosphoSitePlus, UniProt
K4Ubiquitination-PhosphoSitePlus
K11Acetylation-PhosphoSitePlus, UniProt
K11Ubiquitination-PhosphoSitePlus
K19Sumoylation-PhosphoSitePlus
K19Ubiquitination-PhosphoSitePlus
Y21Phosphorylation-PhosphoSitePlus
K24Acetylation-PhosphoSitePlus, UniProt
K24Ubiquitination-PhosphoSitePlus
T31Phosphorylation-PhosphoSitePlus, UniProt
K66Acetylation-PhosphoSitePlus
K66Ubiquitination-PhosphoSitePlus
C83Methylation-IEDB
S89Phosphorylation-PhosphoSitePlus
K90Ubiquitination-PhosphoSitePlus
K97Acetylation-PhosphoSitePlus
K97Ubiquitination-PhosphoSitePlus
K114Ubiquitination-PhosphoSitePlus

These modifications are critical for regulating MTPN's subcellular localization, protein-protein interactions, and biological activities. The extensive network of acetylation and ubiquitination suggests a complex regulatory system that may respond to different cellular conditions and stressors.

How does MTPN contribute to cardiac pathophysiology?

Research in transgenic mouse models demonstrates that cardiac overexpression of myotrophin triggers myocardial hypertrophy and heart failure . While these findings are from murine models, they suggest important implications for human cardiac health and disease. The mechanisms by which MTPN induces cardiac hypertrophy involve:

  • Activation of specific transcription factors including NF-κB

  • Modulation of hypertrophic signaling cascades

  • Alterations in cardiomyocyte calcium handling

  • Changes in extracellular matrix composition

In human research, analyses of cardiac tissue samples from patients with heart failure have shown correlations between MTPN expression levels and disease severity. Researchers investigating this relationship should employ a combination of proteomics and transcriptomics approaches to comprehensively assess MTPN's role in human cardiac pathologies .

What are the methodological considerations for studying MTPN in human neuronal development?

Based on studies in mouse models that demonstrate MTPN's involvement in cerebellar granule cell differentiation, researchers investigating its role in human neuronal development should consider the following methodological approaches:

  • Human iPSC-derived neuronal models that recapitulate developmental stages

  • Temporal analysis of MTPN expression during neuronal differentiation

  • CRISPR-Cas9 gene editing to create MTPN knockdown or knockout models

  • Co-immunoprecipitation studies to identify MTPN-interacting proteins in human neural precursors

  • High-resolution imaging to track MTPN localization during critical developmental windows

The potential role of MTPN in human neuronal differentiation may involve interactions with Notch signaling pathways, as studies in mice suggest that cerebellar development is regulated by components of this pathway . When designing experiments, researchers should incorporate multiple measurement timepoints to capture the dynamic changes in MTPN function throughout neuronal development.

What are optimal experimental designs for studying MTPN functions in human cells?

When designing experiments to investigate MTPN functions in human cells, researchers should consider the following approaches:

  • Loss-of-function studies: Employ RNA interference (siRNA/shRNA) or CRISPR-Cas9 technologies to downregulate or knock out MTPN expression. This approach should include appropriate controls to account for off-target effects.

  • Gain-of-function studies: Utilize overexpression systems with tagged versions of MTPN (e.g., FLAG, GFP) to track localization and identify interaction partners. Consider inducible expression systems to control timing and level of expression.

  • Post-translational modification analysis: Given the extensive modifications observed on MTPN , use site-directed mutagenesis to create variants at key modification sites (K4, K11, K24, etc.) and assess functional consequences.

  • Cell type considerations: Since MTPN functions may vary between tissues, experiments should be conducted in relevant cell types such as cardiomyocytes for cardiac studies and neuronal cells for studies of neuronal development and function.

  • Temporal dynamics: Include time-course analyses to capture dynamic changes in MTPN expression, localization, and interactions following relevant stimuli or during differentiation processes.

How should researchers approach phosphorylation site analysis of MTPN?

MTPN contains multiple phosphorylation sites including Y21, T31, and S89 . To effectively study these modifications:

  • Mass spectrometry-based approaches: Use phospho-enrichment techniques combined with high-resolution mass spectrometry to identify and quantify site-specific phosphorylation events. This should include both data-dependent and targeted approaches.

  • Phospho-specific antibodies: Develop or acquire antibodies that specifically recognize phosphorylated forms of MTPN at Y21, T31, and S89 for immunoblotting and immunofluorescence applications.

  • Phosphorylation site mutants: Generate phospho-mimetic (e.g., S→D or S→E) and phospho-deficient (e.g., S→A) mutants to assess functional consequences of phosphorylation.

  • Kinase prediction and validation: Use bioinformatic tools to predict potential kinases for each phosphorylation site, followed by in vitro kinase assays and cellular studies with specific kinase inhibitors to validate predictions.

  • Functional readouts: Establish clear functional assays to determine how phosphorylation at each site affects MTPN's activities, such as protein-protein interactions, subcellular localization, or target gene expression.

What human subjects research protocols are appropriate for studying MTPN in clinical contexts?

When designing human subjects research to investigate MTPN in clinical contexts, researchers must consider several key factors:

  • Study classification: Determine whether the research meets the NIH definition of a clinical trial by asking four critical questions: Does it involve human participants? Are participants prospectively assigned to interventions? Is it designed to evaluate intervention effects on participants? Is the evaluated effect a health-related biomedical or behavioral outcome?

  • Ethical considerations: Research involving human subjects requires appropriate IRB approval. For multi-site studies, the use of single IRBs is now required for NIH-supported research .

  • Tissue sampling strategies: For studies examining MTPN expression or modifications in human tissues:

    • Clearly define inclusion/exclusion criteria based on clinical characteristics

    • Standardize tissue collection protocols to minimize variability

    • Include appropriate controls (healthy tissue, adjacent non-affected tissue)

    • Document clinical parameters and medications that might influence MTPN expression

  • Data management and sharing: Establish comprehensive protocols for data collection, storage, and sharing that comply with all privacy regulations while maximizing research value.

  • Registration requirements: Studies meeting clinical trial definitions must be registered on ClinicalTrials.gov with appropriate reporting of results .

How can researchers effectively analyze contradictory findings about MTPN function?

When faced with contradictory findings regarding MTPN function across different studies:

  • Methodological analysis: Carefully evaluate experimental methods, including:

    • Cell types and conditions used

    • Species differences (human vs. model organisms)

    • Protein detection methods and antibody specificity

    • Expression systems and fusion tags that might affect function

    • Post-translational modification states of the studied protein

  • Contextual dependencies: Consider that MTPN function may be highly context-dependent, varying with:

    • Cell type and differentiation state

    • Presence of specific interacting partners

    • Metabolic state of the cell

    • Disease conditions vs. normal physiology

  • Meta-analysis approaches: When sufficient data exist, conduct formal meta-analyses to:

    • Identify patterns across studies

    • Determine effect sizes for specific MTPN functions

    • Identify moderating variables that explain discrepancies

  • Collaborative resolution: Establish collaborations between labs with contradictory findings to:

    • Exchange reagents and protocols

    • Perform side-by-side experiments under identical conditions

    • Jointly develop standardized assays for key MTPN functions

What are the optimal protein-protein interaction methods for studying MTPN complexes?

Given MTPN's involvement in multiple cellular pathways, understanding its protein interaction network is crucial. Researchers should consider these complementary approaches:

  • Proximity-based labeling: BioID or APEX2 fusion proteins can identify proteins in close proximity to MTPN in living cells, providing insights into the spatial organization of MTPN-containing complexes.

  • Co-immunoprecipitation with quantitative proteomics: Use stable isotope labeling approaches (SILAC, TMT) combined with immunoprecipitation to quantitatively assess MTPN interaction partners under different conditions.

  • Crosslinking mass spectrometry (XL-MS): Apply chemical crosslinking followed by mass spectrometry to identify direct binding interfaces between MTPN and its partners.

  • Fluorescence resonance energy transfer (FRET): Develop FRET-based assays to monitor MTPN interactions with key partners in living cells and assess how these interactions change dynamically.

  • Split protein complementation assays: Techniques such as bimolecular fluorescence complementation (BiFC) can validate specific interactions and determine their subcellular localization.

When analyzing interaction data, researchers should focus on distinguishing between direct binding partners and components of larger multiprotein complexes, as well as identifying interaction dependencies on post-translational modifications of MTPN, particularly at lysine residues that undergo multiple types of modifications .

How can researchers effectively study the impact of MTPN acetylation on its function?

MTPN undergoes acetylation at multiple lysine residues (K4, K11, K24, K66, K97) , which likely influences its function through multiple mechanisms. To study these effects:

  • Site-specific acetylation detection: Develop or utilize acetyl-lysine antibodies specific for each acetylation site in MTPN, complemented by mass spectrometry-based quantification.

  • Genetic code expansion technology: Incorporate acetyl-lysine directly during protein synthesis using amber codon suppression methods to generate MTPN with site-specific acetylation.

  • Acetyltransferase/deacetylase screening: Identify the enzymes responsible for adding and removing acetyl groups on MTPN through candidate approaches or systematic screening of HAT and HDAC libraries.

  • Acetylation mimetics: Generate lysine-to-glutamine (K→Q) mutations to mimic acetylation and lysine-to-arginine (K→R) mutations to prevent acetylation at specific sites.

  • Functional assays to assess acetylation effects:

    • Protein stability and turnover rates

    • Subcellular localization studies

    • DNA/RNA binding capabilities using EMSA or RNA-IP

    • Interaction partner preferences using quantitative proteomics

    • Transcriptional regulation activities if MTPN influences gene expression

Product Science Overview

Structure and Expression

Myotrophin is composed of 118 amino acids and has a molecular weight of approximately 15 kDa . The protein is typically expressed in Escherichia coli (E. coli) for recombinant production . The recombinant form of Myotrophin often includes a His-tag at the N-terminus to facilitate purification and detection .

Biological Functions
  1. Cardiac Hypertrophy: Myotrophin is known to stimulate protein synthesis and cardiomyocyte growth, which leads to cardiac hypertrophy. This process is mediated through the activation of the NF-kappaB signaling cascade . Elevated levels of Myotrophin have been observed in human dilated cardiomyopathic and ischemic hearts .

  2. Cerebellar Morphogenesis: Myotrophin plays a potential role in the development of the cerebellum, particularly in the differentiation of cerebellar neurons such as granule cells .

  3. Skeletal Muscle Growth: In addition to its role in cardiac and neural tissues, Myotrophin has been shown to promote skeletal muscle growth both in vitro and in vivo .

Applications and Research

Recombinant Myotrophin is widely used in research to study its various functions and potential therapeutic applications. It is utilized in experiments involving SDS-PAGE and mass spectrometry to analyze its purity and molecular weight . The protein is also used in studies focusing on cardiac hypertrophy, neurogenesis, and muscle differentiation.

Storage and Stability

Recombinant Myotrophin is typically shipped and stored at 4°C for short-term use (1-2 weeks). For long-term storage, it is aliquoted and kept at -20°C or -80°C to avoid freeze-thaw cycles, which can degrade the protein .

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.