TPA (311-562) Human

Tissue Plasminogen Activator (311-562 a.a.) Human Recombinant
Shipped with Ice Packs
In Stock

Description

TPA Human Recombinant produced in Sf9 Baculovirus cells is a single, glycosylated polypeptide chain containing 261 amino acids (311-562 a.a.) and having a molecular mass of 29.2kDa (Migrates at 28-40kDa on SDS-PAGE under reducing conditions).
TPA is expressed with a 6 amino acid His tag at C-Terminus and purified by proprietary chromatographic techniques.

Product Specs

Introduction
Tissue plasminogen activator (tPA or PLAT) is a serine protease that activates plasminogen into plasmin, a fibrinolytic enzyme responsible for breaking down blood clots. tPA is naturally occurring and plays a crucial role in dissolving blood clots, thereby aiding in the treatment of conditions such as stroke and heart attack. This enzyme is also involved in cell migration and tissue remodeling.
Description
This product consists of the recombinant form of human tissue plasminogen activator (tPA), specifically amino acids 311 to 562. It is produced using Sf9 insect cells and undergoes glycosylation, a process crucial for its biological activity. The protein, with a molecular weight of 29.2 kDa, is engineered to carry a 6-amino acid His tag at its C-terminus to facilitate purification. The final product is highly pure, exceeding 95% purity, as confirmed by SDS-PAGE analysis.
Physical Appearance
Clear, colorless solution, devoid of any particulate matter.
Formulation
The tPA protein is provided in a solution containing 50mM MES (pH 5.0), 5mM CaCl2, 1mM DTT, 0.5M NaCl, and 30% glycerol. This specific formulation helps maintain the stability and activity of the protein.
Stability
For short-term storage (up to 4 weeks), the product can be stored at 4°C. For extended storage, it is recommended to store the product at -20°C. To further enhance stability during long-term storage, the addition of a carrier protein such as HSA or BSA (0.1%) is advised. Repeated freezing and thawing should be avoided to maintain the integrity of the protein.
Purity
High purity exceeding 95%, determined by SDS-PAGE.
Synonyms
Tissue-type plasminogen activator, EC 3.4.21.68, tPA, t-PA, t-plasminogen activator, TPA, T-PA, DKFZp686I03148, PLAT and tPA.
Source
Sf9, Baculovirus cells.
Amino Acid Sequence
ADPIKGGLFA DIASHPWQAA IFAKHRRSPG ERFLCGGILI SSCWILSAAH CFQERFPPHH LTVILGRTYR VVPGEEEQKF EVEKYIVHKE FDDDTYDNDI ALLQLKSDSS RCAQESSVVR TVCLPPADLQ LPDWTECELS GYGKHEALSP FYSERLKEAH VRLYPSSRCT SQHLLNRTVT DNMLCAGDTR SGGPQANLHD ACQGDSGGPL VCLNDGRMTL VGIISWGLGC GQKDVPGVYT KVTNYLDWIR DNMRPHHHHH H.

Q&A

What is Human TPA (311-562) and how does it differ from full-length TPA?

Human Tissue-type Plasminogen Activator (311-562) represents a specific fragment of the full TPA protein, encompassing amino acids 311-562 of the 562-amino acid complete protein. This fragment contains the C-terminal serine protease domain, which is responsible for the catalytic activity of TPA. At 29.2 kDa, this fragment is significantly smaller than the full-length protein, which has a molecular weight of approximately 70 kDa when glycosylated .

The full-length TPA contains multiple domains including finger, growth factor, and kringle domains in its N-terminal region (amino acids 1-310), which are absent in the 311-562 fragment. These N-terminal domains are primarily involved in binding to fibrin and cell surfaces, while the C-terminal 311-562 region contains the catalytic domain responsible for the enzymatic conversion of plasminogen to plasmin .

What are the structural characteristics of the TPA (311-562) domain?

The TPA (311-562) fragment contains the serine protease domain of TPA, which belongs to the trypsin family of serine proteases. This domain contains the catalytic triad (His, Asp, Ser) responsible for the enzyme's proteolytic activity. The serine protease domain adopts a characteristic two β-barrel structure with the catalytic residues positioned at the interface between the barrels .

Crystal structure analysis has revealed that this domain contains multiple disulfide bonds that stabilize its tertiary structure. These structural features are critical for maintaining the proper conformation of the catalytic site and, consequently, the enzymatic activity of the protein .

What are the primary research applications for TPA (311-562)?

TPA (311-562) serves as a valuable molecular tool in various biochemical applications, particularly in studies focusing on:

  • Proteolytic mechanisms and enzyme kinetics

  • Structure-function relationships of serine proteases

  • Fibrinolysis pathway research

  • Development of novel thrombolytic agents

  • Complement system modulation studies

The isolated catalytic domain enables researchers to study the enzymatic properties of TPA without the influence of its binding domains, allowing for more focused investigations of its proteolytic activity and interactions with inhibitors or substrates .

How should I design experiments to study the influence of TPA (311-562) on the complement system?

When designing experiments to investigate TPA (311-562) interactions with the complement system, consider the following methodological approach:

  • Experimental models selection:

    • In vitro assays using purified complement components

    • Ex vivo models using human serum

    • Animal models (e.g., rabbits as mentioned in the research literature)

  • Experimental design framework:

    • Include appropriate controls (both positive and negative)

    • Design dose-response experiments to determine concentration-dependent effects

    • Compare TPA (311-562) with full-length TPA to assess domain-specific effects

    • Include time-course studies to evaluate kinetics of complement modulation

  • Key readouts to measure:

    • Complement activation markers (C3a, C5a, SC5b-9)

    • Complement-dependent cytotoxicity

    • Inflammatory mediator production

    • Cell surface deposition of complement components

Ensure that experimental conditions mimic physiological conditions relevant to the specific complement pathway being studied (classical, alternative, or lectin pathway) .

What experimental considerations are important when assessing TPA (311-562) activity in cancer research models?

When studying TPA (311-562) in cancer research, particularly in melanoma models where both TPA and glutathione transferase P1-1 (GST P1-1) expression have been observed, consider the following experimental design elements:

  • Model selection:

    • Human melanoma cell lines (correlating with the research on melanoma metastases)

    • Patient-derived xenografts

    • Primary tumor samples for ex vivo analysis

  • Critical parameters to analyze:

    • Gene expression analysis of TPA alongside N-RAS mutations

    • Correlation between TPA activity and GST P1-1 expression levels

    • Signal transduction pathways affected by TPA activity

    • Influence on tumor cell migration and invasion

  • Methodological approaches:

    • Immunohistochemistry for protein localization

    • Western blotting for expression level quantification

    • Genetic analysis for mutation identification

    • Functional assays to measure enzymatic activity

    • RNA interference to assess functional relationships

When designing such experiments, it is essential to include appropriate controls and to consider the heterogeneity of cancer cells within the same tumor .

What are the recommended storage and handling conditions for TPA (311-562)?

For optimal stability and activity of TPA (311-562), adhere to the following storage and handling guidelines:

  • Storage temperature: -20°C is recommended for long-term storage

  • Solution composition: Typically supplied in a buffer solution that maintains protein stability

  • Avoid repeated freeze-thaw cycles: Aliquot the protein upon receipt to minimize degradation

  • Handling precautions:

    • Use sterile, nuclease-free consumables and reagents

    • Work with the protein on ice when possible

    • Avoid prolonged exposure to room temperature

    • Minimize exposure to proteases and oxidizing agents

What assay systems are available for measuring TPA (311-562) activity?

Several assay systems can be utilized to measure the enzymatic activity of TPA (311-562):

  • Chromogenic substrate assays:

    • Utilize synthetic peptide substrates linked to chromogenic groups

    • Provide quantitative measurement of protease activity

    • Allow for real-time kinetic assessment of enzyme activity

  • Fluorogenic substrate assays:

    • More sensitive than chromogenic assays

    • Utilize peptide substrates linked to fluorescent reporters

    • Enable detection of lower enzyme concentrations

  • Plasminogen activation assays:

    • Measure the physiological function of TPA (conversion of plasminogen to plasmin)

    • Can be coupled with chromogenic or fluorogenic plasmin substrates

    • Allow for assessment of the complete enzymatic cascade

  • Clot lysis assays:

    • Measure the functional activity in fibrin clot dissolution

    • More physiologically relevant but less quantitative

    • Useful for comparing relative activities between different TPA variants

When selecting an assay system, consider the specific research question, required sensitivity, and available instrumentation.

How should I design human subject experiments involving TPA (311-562)?

When designing human subject experiments involving TPA (311-562), adhere to these methodological principles:

  • Study design considerations:

    • Clearly define evaluation constructs and measurements

    • Implement a controlled experimental design with appropriate randomization

    • Determine sample size through power analysis to ensure statistical validity

    • Select appropriate inclusion and exclusion criteria

    • Consider potential confounding variables

  • Ethical requirements:

    • Obtain proper institutional review board (IRB) approval

    • Develop comprehensive informed consent documentation

    • Ensure participant privacy and data security

    • Design protocols that minimize risks to participants

  • Methodological approach:

    • Select appropriate control groups (placebo, standard of care, etc.)

    • Implement blinding procedures where applicable

    • Establish clear primary and secondary endpoints

    • Develop a detailed statistical analysis plan before study initiation

Human studies should follow established guidelines for experimental design in clinical research, with particular attention to ethical considerations given TPA's biological activities .

How do post-translational modifications affect TPA (311-562) function?

Post-translational modifications (PTMs) can significantly impact TPA (311-562) function in several ways:

  • Glycosylation impacts:

    • While the full-length TPA contains multiple glycosylation sites, the 311-562 fragment may contain fewer glycosylation sites

    • Changes in glycosylation can affect protein stability, solubility, and resistance to proteolytic degradation

    • Differential glycosylation may influence interactions with inhibitors and substrates

  • Phosphorylation considerations:

    • Phosphorylation sites within the catalytic domain can modulate enzyme activity

    • Phosphorylation may affect protein-protein interactions and subcellular localization

    • Kinase-mediated regulation represents a potential mechanism for fine-tuning TPA activity

  • Disulfide bond formation:

    • Proper disulfide bond formation is critical for the structural integrity of the serine protease domain

    • Oxidative conditions can lead to aberrant disulfide bond formation, potentially inactivating the enzyme

    • Reduction of critical disulfide bonds will result in loss of tertiary structure and activity

Research methods to investigate PTMs include mass spectrometry, site-directed mutagenesis of modification sites, and comparative activity assays between differentially modified protein preparations.

What is known about structure-function relationships in the catalytic domain of TPA?

The structure-function relationship in the TPA catalytic domain (311-562) has been extensively characterized:

  • Catalytic triad:

    • His322, Asp371, and Ser478 form the canonical serine protease catalytic triad

    • These residues are essential for the hydrolysis of peptide bonds

    • Mutation of any triad residue results in profound activity reduction

  • Substrate binding pocket:

    • The S1 specificity pocket confers preference for arginine or lysine at the P1 position

    • Loops surrounding the active site determine substrate specificity beyond the P1 position

    • Surface loops create extended substrate binding sites (S2-S4 and S1'-S3')

  • Allosteric regulatory sites:

    • Regions distant from the active site can influence catalytic activity through conformational changes

    • Ligand binding to these sites can enhance or inhibit enzymatic activity

    • These sites represent potential targets for developing selective modulators

  • Structural determinants of zymogen activation:

    • TPA is produced as a single-chain form that undergoes proteolytic cleavage

    • The 311-562 fragment may represent the activated form depending on the specific preparation

    • The transition from zymogen to active enzyme involves substantial conformational changes

Understanding these structure-function relationships is crucial for rational design of inhibitors, engineered variants with altered specificity, and development of therapeutic proteins with improved properties.

How does TPA (311-562) interact with physiological inhibitors compared to full-length TPA?

The interaction between TPA (311-562) and physiological inhibitors differs from full-length TPA in several important aspects:

Experimental approaches to study these differences include:

  • Surface plasmon resonance to measure binding kinetics

  • Enzyme inhibition assays comparing IC50 and Ki values

  • Structural analysis of inhibitor-enzyme complexes using X-ray crystallography or cryo-EM

How can I troubleshoot inconsistent TPA (311-562) activity in my experiments?

When encountering inconsistent TPA (311-562) activity in experimental settings, systematically investigate these potential sources of variation:

  • Protein quality and integrity:

    • Verify protein concentration using multiple methods (Bradford, BCA, A280)

    • Assess protein integrity by SDS-PAGE to confirm absence of degradation

    • Consider lot-to-lot variability in commercial preparations

  • Experimental conditions:

    • Buffer composition: check pH, ionic strength, and presence of metal ions

    • Temperature fluctuations: maintain consistent temperature during assays

    • Substrate quality: use fresh substrates and verify their purity

    • Presence of inhibitors: check for inadvertent introduction of inhibitors

  • Instrument and measurement variations:

    • Calibrate instruments regularly

    • Use internal standards for normalization

    • Perform technical replicates to identify measurement variability

  • Statistical approaches to address variability:

    • Calculate coefficient of variation (%CV) for replicate measurements

    • Apply appropriate statistical tests considering data distribution

    • Consider nested experimental designs to separate sources of variation

If inconsistencies persist, consider performing a systematic evaluation of all experimental parameters through a design of experiments (DOE) approach to identify significant factors affecting enzyme activity.

What controls should be included when studying interactions between TPA (311-562) and complement components?

When investigating interactions between TPA (311-562) and complement components, include these essential controls:

  • Activity controls:

    • Positive control: Known activator of the specific complement pathway being studied

    • Negative control: Buffer alone or heat-inactivated enzyme

    • Full-length TPA: To compare with the 311-562 fragment and identify domain-specific effects

  • Specificity controls:

    • Enzymatically inactive TPA (311-562) mutant (e.g., catalytic serine to alanine mutation)

    • Related serine proteases to assess specificity of observed effects

    • Specific inhibitors of TPA activity to confirm enzyme-dependent effects

  • System validation controls:

    • Complement-deficient serum (for specific components)

    • Heat-inactivated serum (56°C for 30 minutes)

    • EDTA-treated samples (to block classical and alternative pathways)

  • Technical controls:

    • Internal standards for normalization across experiments

    • Concentration gradients to establish dose-dependent effects

    • Time-course samples to establish kinetics of interactions

Proper implementation of these controls will enhance data reliability and facilitate correct interpretation of results, particularly when studying complex biological systems like the complement cascade.

How do I interpret contradictory data regarding TPA (311-562) effects in different experimental models?

When faced with contradictory data regarding TPA (311-562) effects across different experimental models, employ the following analytical framework:

  • Systematic comparison of experimental conditions:

    ParameterModel AModel BModel C
    Cell/tissue type(specify)(specify)(specify)
    TPA concentrationX μg/mLY μg/mLZ μg/mL
    Incubation timeX hoursY hoursZ hours
    Medium composition(detail)(detail)(detail)
    Readout method(specify)(specify)(specify)
  • Biological context analysis:

    • Receptor expression profiles differ between models

    • Presence of endogenous inhibitors varies between systems

    • Signal transduction pathway components may differ

    • Cross-talk with other pathways may influence outcomes

  • Technical reconciliation approaches:

    • Standardize experimental conditions across models where possible

    • Use multiple readout methods to confirm observations

    • Perform intervention studies with specific inhibitors or genetic approaches

    • Consider temporal dynamics (early vs. late effects)

  • Integrated data interpretation:

    • Seemingly contradictory data may reveal context-dependent regulation

    • Build mechanistic models that can account for differential effects

    • Consider that effects may be direct in some systems and indirect in others

    • Evaluate the physiological relevance of each model system

Remember that contradictory results often provide valuable insights into context-dependent mechanisms and regulatory complexity rather than representing experimental failures.

How is TPA (311-562) being utilized in research on neurodegenerative diseases?

Current research is exploring the role of TPA (311-562) in neurodegenerative disease mechanisms:

  • Alzheimer's disease applications:

    • TPA's catalytic domain may influence amyloid-β aggregation and clearance

    • The 311-562 fragment allows study of proteolytic activity independent of binding domains

    • Researchers are investigating interactions with key Alzheimer's disease proteins

  • Parkinson's disease investigations:

    • Potential role in α-synuclein processing and clearance

    • Studies examining neuroprotective vs. neurotoxic effects of catalytic activity

    • Models using TPA (311-562) help disambiguate catalytic from binding effects

  • Experimental approaches:

    • Primary neuron cultures with controlled exposure to TPA (311-562)

    • Brain slice models to evaluate tissue-level effects

    • Transgenic animal models with modified TPA expression or activity

    • Patient-derived iPSC neurons to evaluate disease-specific effects

This research direction holds promise for understanding the dual roles of TPA in both neuroprotection and neurotoxicity in different disease contexts.

What are the emerging applications of TPA (311-562) in cancer research beyond melanoma?

Emerging research is expanding TPA (311-562) applications in multiple cancer types:

  • Breast cancer research:

    • Investigating relationships between TPA catalytic activity and tumor invasion

    • Studying interactions with matrix metalloproteinases in tumor microenvironment

    • Examining correlations between TPA expression and treatment resistance

  • Glioblastoma applications:

    • Exploring blood-brain barrier modulation by TPA catalytic domain

    • Investigating interactions with GST P1-1 in glioma cells

    • Studying potential roles in treatment delivery across the blood-brain barrier

  • Methodological approaches in cancer research:

    • 3D organoid models to study TPA effects on tumor architecture

    • Combination studies with chemotherapeutic agents

    • In vivo imaging of TPA activity using activity-based probes

    • Correlative studies between TPA activity and clinical outcomes

These studies leverage the catalytic function of TPA (311-562) to understand cancer progression mechanisms and potentially identify new therapeutic targets.

Product Science Overview

Introduction

Tissue Plasminogen Activator (tPA) is a serine protease enzyme that plays a crucial role in the breakdown of blood clots. It is primarily used in the treatment of ischemic stroke and other thrombotic conditions. The recombinant form of tPA, particularly the segment spanning amino acids 311-562, has been extensively studied and utilized in various therapeutic applications.

Structure and Function

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.