Recombinant Gloydius ussuriensis Zinc metalloproteinase/disintegrin ussurin

Shipped with Ice Packs
In Stock

Product Specs

Form
Lyophilized powder

Note: While we prioritize shipping the format currently in stock, please specify your format preference in order notes. We will accommodate your request to the best of our ability.

Lead Time
Delivery times vary depending on the purchase method and location. Please contact your local distributor for precise delivery estimates.

Note: Standard shipping includes blue ice packs. Dry ice shipping requires prior arrangement and incurs additional charges.

Notes
Avoid repeated freeze-thaw cycles. Store working aliquots at 4°C for up to one week.
Reconstitution
Centrifuge the vial briefly before opening to consolidate the contents. Reconstitute the protein in sterile, deionized water to a concentration of 0.1-1.0 mg/mL. For long-term storage, we recommend adding 5-50% glycerol (final concentration) and aliquoting at -20°C/-80°C. Our standard glycerol concentration is 50%, which may serve as a reference.
Shelf Life
Shelf life depends on various factors, including storage conditions, buffer composition, temperature, and protein stability. Generally, liquid formulations have a 6-month shelf life at -20°C/-80°C, while lyophilized formulations have a 12-month shelf life at -20°C/-80°C.
Storage Condition
Upon receipt, store at -20°C/-80°C. Aliquoting is essential for multiple uses. Avoid repeated freeze-thaw cycles.
Tag Info
Tag type is determined during the manufacturing process.

The tag type is determined during production. If you require a specific tag type, please inform us, and we will prioritize its inclusion.

Synonyms
Zinc metalloproteinase/disintegrin ussurin [Cleaved into: Snake venom metalloproteinase ussurin; SVMP; EC 3.4.24.-); Disintegrin ussurin]
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Expression Region
191-389
Protein Length
Full Length of Mature Protein
Purity
>85% (SDS-PAGE)
Species
Gloydius ussuriensis (Ussuri mamushi) (Gloydius blomhoffii ussuriensis)
Target Protein Sequence
YQRYVELVVV ADHRMVTKYN GNLIIIRTWV YEIFNTINEI YQRMNIHVAL VGLEIWSNGD KIIVQSSADV TLDLFGTWGE IDLLKRKSHD NAQLLTPTDF DGPTIGLAYV GTMCDPKRST GVVQDFSPIN LLVAVTMAHE IGHNLGMNHD ENYCSCGGFA CIMSPVISPQ PSKLFSYCSY IHYWTYINYR NPQCILNKP
Uniprot No.

Target Background

Function
This protein impairs hemostasis in envenomed animals by inhibiting platelet aggregation induced by ADP, thrombin, platelet-activating factor, and collagen. This inhibitory effect is achieved by blocking the interaction between fibrinogen and the platelet receptors GPIIb/GPIIIa (ITGA2B/ITGB3).
Protein Families
Venom metalloproteinase (M12B) family, P-II subfamily, P-IIa sub-subfamily
Subcellular Location
Secreted.
Tissue Specificity
Expressed by the venom gland.

Q&A

What is the molecular structure of ussurin and which functional domains does it contain?

Ussurin is a P-II class snake venom metalloproteinase with a molecular mass of approximately 53.2 kDa and an isoelectric point of 5.37. The full-length cDNA of ussurin contains a 51 bp 5'-UTR, an open reading frame of 1434 bp coding for 478 amino acids, and a 490 bp 3'-UTR .

The protein consists of four distinct regions:

  • A signal sequence of 18 amino acid residues

  • A zymogen pro-peptide of 171 amino acid residues containing a cysteine switch motif (PK-MCGVT)

  • A central metalloproteinase domain of 201 amino acid residues with a conserved zinc-chelating sequence (HEXXHXXGXXH) and a methionine-turn CIM involved in zinc binding

  • A disintegrin domain of 73 amino acid residues featuring the characteristic RGD sequence and six disulfide bonds

Between the metalloproteinase and disintegrin domains lies a spacer sequence of 15 amino acid residues with conserved T392, T397, and S400 residues, which are specific to P-II snake venom metalloproteinases .

How is ussurin classified within snake venom proteins and what is its relative abundance?

Ussurin belongs to the P-II SVMP class, which is characterized by having both metalloproteinase and disintegrin domains. Within venom composition studies, zinc metalloproteinase/disintegrin ussurin has been detected at relatively low abundance levels compared to other venom components.

As shown in transcriptomic studies, the relative abundance of ussurin in snake venom can be quantified as follows:

Protein Family/Protein SubtypeAccession No. (Species)Relative Abundance %
P-I SVMP categoryQ7SZD9 (Gloydius ussuriensis)0.03
P-II SVMP categoryQ7SZD9 (Gloydius ussuriensis)0.04

This suggests that although ussurin is a functionally important component, it comprises a small percentage of the total venom protein content in Gloydius ussuriensis .

What are the recommended expression systems for producing recombinant ussurin?

For successful recombinant expression of functional ussurin, researchers should consider several expression systems with specific methodological approaches:

  • Yeast expression systems: Particularly Pichia pastoris, which has been successfully used for the expression of snake venom metalloproteinases. This system allows for proper protein folding and post-translational modifications. The protocol typically involves:

    • Codon optimization of the ussurin gene for yeast expression

    • Cloning into a vector containing an inducible promoter (such as AOX1)

    • Transformation into competent P. pastoris cells

    • Selection of transformants on appropriate media

    • Induction of protein expression using methanol

    • Purification from culture supernatant

  • Bacterial expression systems: While potentially higher-yielding, these systems often result in inclusion body formation that requires additional refolding steps. If using E. coli:

    • Express the protein fused with solubility tags (e.g., MBP, SUMO, or Trx)

    • Perform careful optimization of induction conditions (temperature, IPTG concentration)

    • Include specific metal ions (particularly Zn²⁺) in the culture media to assist in proper folding

The choice between these systems should be guided by the specific research needs, balancing between yield, functional activity, and authenticity of post-translational modifications.

What purification strategies are most effective for isolating recombinant ussurin while maintaining its enzymatic activity?

Purification of recombinant ussurin requires special consideration to maintain the structural integrity and enzymatic activity of the protein:

Activity assessment should be performed at each purification step using fibrin plate assays or suitable chromogenic substrates to ensure that enzymatic function is maintained.

What are the established methods for assessing the proteolytic activity of recombinant ussurin?

Several methodologies can be employed to characterize the proteolytic activity of recombinant ussurin:

  • Fibrin plate assay: This is the gold standard for assessing fibrinolytic activity.

    • Prepare fibrin plates by mixing fibrinogen (16 mg/mL) with thrombin

    • Apply purified ussurin samples (starting from 0.2 mg/mL) to wells in the plate

    • Incubate at 37°C for 12-24 hours

    • Measure the diameter of clear zones of hydrolysis

  • Fibrinogen degradation analysis using SDS-PAGE:

    • Incubate fibrinogen (16 mg/mL) with ussurin (1.6 μg/mL) at 37°C

    • Collect samples at different time points (0, 15, 30, 60 min)

    • Analyze by SDS-PAGE to observe the degradation pattern of fibrinogen chains

    • Ussurin primarily hydrolyzes the α-chain of fibrinogen, which is characteristic of many P-II SVMPs

  • Inhibition studies: To confirm the metalloproteinase nature of ussurin, perform activity assays in the presence of various inhibitors:

    • EDTA and EGTA should completely inhibit activity

    • 1,10-phenanthroline should abolish activity

    • Thiol reducing agents (DTT, cysteine) may inhibit activity by disrupting disulfide bonds

    • Zn²⁺ typically enhances activity, while Fe²⁺ and Hg²⁺ often inhibit it

How can the disintegrin function of ussurin be evaluated in research settings?

The disintegrin domain of ussurin containing the RGD sequence mediates its interaction with platelet integrins. To assess this function:

  • Platelet aggregation assays:

    • Prepare platelet-rich plasma from fresh blood

    • Induce aggregation using agonists like ADP, collagen, or thrombin

    • Add various concentrations of purified ussurin

    • Monitor aggregation using a platelet aggregometer

    • Calculate IC₅₀ values for inhibition of aggregation

  • Cell adhesion inhibition assays:

    • Culture cells expressing relevant integrins (e.g., αIIbβ3 on platelets or αvβ3 on various cell types)

    • Coat plates with extracellular matrix proteins (fibronectin, vitronectin, etc.)

    • Pre-incubate cells with different concentrations of ussurin

    • Quantify cell adhesion inhibition

    • This approach helps identify the specific integrin targets of ussurin

  • Direct binding assays:

    • Purify integrins from appropriate cell sources

    • Perform solid-phase binding assays with immobilized integrins and labeled ussurin

    • Alternatively, use surface plasmon resonance (SPR) for real-time binding kinetics

    • Determine binding parameters (Kd, kon, koff) for different integrins

How does recombinant ussurin compare functionally to native ussurin from Gloydius ussuriensis venom?

Comparative analysis between recombinant and native ussurin is essential for validating expression systems and understanding structural determinants of function:

  • Enzymatic activity comparison:

    • Fibrinolytic activity between native and recombinant proteins often differs due to variations in post-translational modifications

    • Zn²⁺ supplementation is typically more critical for recombinant protein activity as the metal ion may be lost during expression/purification

    • Recombinant ussurin may show 60-80% of the activity of native protein, depending on the expression system

  • Structural analysis:

    • Circular dichroism spectroscopy to compare secondary structure elements

    • Mass spectrometry to identify differences in post-translational modifications

    • N-terminal sequencing to confirm proper processing of the signal peptide in recombinant systems

  • Stability differences:

    • Native ussurin often exhibits greater thermal stability than recombinant versions

    • Recombinant protein may show higher susceptibility to autoproteolysis

    • Different metal ion dependencies may be observed

How does ussurin compare to other P-II SVMPs from different snake species, and what are the implications for evolutionary studies?

Comparative analysis of P-II SVMPs across species reveals important evolutionary insights:

Snake speciesSVMPClassMass (kDa)Hemorrhagic activityFibrinogen degradationTripeptidePlatelet aggregation inhibition
Gloydius ussuriensisUssurinIIa53--RGDYes
Bothrops insularisInsularinase-AIIa53NoAlpha, BetaRGDYes
Crotalus atroxAtrolysin-EIIa53YesAlphaMVDYes
Protobothrops flavoviridisFlavoridinIIa54--RGDYes

Key observations from comparative studies:

  • Sequence conservation: Ussurin shows high sequence homology with other P-II SVMPs, particularly in the metalloproteinase domain and cysteine switch motif.

  • Functional diversity: Despite structural similarities, functional differences exist:

    • Some P-II SVMPs like Atrolysin-E are hemorrhagic, while others like Insularinase-A are not

    • The tripeptide motif in the disintegrin domain varies (RGD in ussurin vs. MVD in Atrolysin-E)

    • Fibrinogen degradation patterns differ between enzymes

  • Evolutionary implications:

    • Hypervariable regions in the disintegrin domain suggest adaptive evolution targeting different receptors

    • Conservation of the metalloproteinase domain and cysteine switch motif indicates functional constraints on enzyme activity

    • The selective pressure on disintegrin domains may reflect prey-specific adaptations in different snake species

What approaches can be used to create structure-function mutants of ussurin to examine specific protein domains?

Site-directed mutagenesis of recombinant ussurin provides powerful insights into structure-function relationships:

  • Metalloproteinase domain mutations:

    • Target the zinc-binding motif (HEXXHXXGXXH) by substituting histidine residues to abolish catalytic activity while maintaining structural integrity

    • Modify the methionine-turn (CIM) to investigate its role in zinc coordination

    • Create chimeric constructs with other SVMPs to explore determinants of substrate specificity

  • Disintegrin domain mutations:

    • Substitute the RGD sequence with KGD, MVD, or ECD to alter integrin specificity

    • Modify cysteine residues to disrupt disulfide bonding patterns

    • Truncate the C-terminal region to assess its role in integrin targeting

  • Domain deletion variants:

    • Express the metalloproteinase domain alone to study its activity independent of the disintegrin domain

    • Express the disintegrin domain separately to investigate its integrin-binding properties without proteolytic activity

    • Create progressive truncations to map functional regions

  • Expression and analysis strategies:

    • Use eukaryotic expression systems (preferably yeast or mammalian cells) for proper folding

    • Perform comprehensive biochemical characterization (proteolytic activity, integrin binding)

    • Conduct structural studies (X-ray crystallography or NMR) on wild-type and mutant proteins

How can researchers address the challenges of protein misfolding and autoprocessing when working with recombinant ussurin?

Recombinant expression of SVMPs like ussurin often encounters challenges with protein folding and autoprocessing:

  • Preventing autoprocessing:

    • Express the protein as a zymogen with intact pro-domain to maintain latency

    • Introduce mutations in the cysteine switch motif (PK-MCGVT) to prevent autolytic activation

    • Add specific metalloproteinase inhibitors (e.g., GM6001) to expression media

    • Lower the culture temperature to 16-20°C during expression to reduce enzyme activity

  • Promoting proper folding:

    • Co-express molecular chaperones (e.g., PDI, BiP) in eukaryotic systems

    • Supplement expression media with Zn²⁺ ions at 10-50 μM concentration

    • Include small molecule chemical chaperones like glycerol (5-10%) or L-arginine (50-200 mM) in culture media

    • Optimize oxidizing conditions for disulfide bond formation in the expression system

  • Refolding strategies from inclusion bodies:

    • Solubilize inclusion bodies using 6-8 M urea or 4-6 M guanidine-HCl

    • Perform step-wise dialysis to remove denaturant

    • Include a redox system (reduced/oxidized glutathione) for disulfide bond formation

    • Add Zn²⁺ ions during refolding to facilitate proper metalloproteinase domain folding

    • Monitor refolding by measuring recovery of enzymatic activity

What are the most promising research applications of recombinant ussurin beyond basic structural studies?

Recombinant ussurin has several potential applications in both basic and translational research:

  • Antiplatelet drug development:

    • The RGD-containing disintegrin domain provides a template for designing novel antiplatelet agents

    • Structure-based drug design using the disintegrin domain structure to create small molecule integrin antagonists

    • Development of peptide mimetics based on the RGD region and surrounding amino acids

  • Thrombolytic agent research:

    • Exploring the fibrinolytic activity for developing novel thrombolytic agents

    • Creating variants with enhanced fibrin specificity and reduced hemorrhagic potential

    • Investigating synergistic effects with established thrombolytics like tPA

  • Cell biology research tools:

    • Using the disintegrin domain as a probe for studying integrin-mediated cell adhesion

    • Developing fluorescently-labeled ussurin derivatives to visualize integrin distribution

    • Creating affinity reagents for purifying or detecting specific integrins

  • Cancer research applications:

    • Investigating the effects on tumor cell migration and invasion mediated by integrin inhibition

    • Exploring potential anti-angiogenic effects through disruption of endothelial cell interactions

    • Developing targeted delivery systems for anti-cancer drugs using the integrin-binding properties

How can systems biology approaches be applied to study the molecular interactions and signaling pathways affected by ussurin?

Advanced systems biology approaches provide comprehensive insights into ussurin's effects:

  • Interactome analysis:

    • Perform pull-down experiments with immobilized ussurin to identify binding partners

    • Use yeast two-hybrid or mammalian two-hybrid systems to screen for interacting proteins

    • Apply protein microarrays to identify novel molecular targets beyond integrins

    • Validate identified interactions using co-immunoprecipitation and SPR techniques

  • Phosphoproteomics:

    • Treat platelets or other target cells with ussurin and analyze changes in phosphorylation patterns

    • Map affected signaling pathways, particularly those downstream of integrin activation

    • Identify key nodes in signaling networks that could be targeted for therapeutic intervention

  • Transcriptomics and proteomics integration:

    • Perform RNA-seq on cells treated with ussurin to identify gene expression changes

    • Couple with proteomics data to create comprehensive network models

    • Apply pathway enrichment analysis to identify biological processes most affected

    • Develop predictive models of cellular responses to ussurin treatment

  • Single-cell analysis approaches:

    • Use single-cell RNA-seq to identify cell-specific responses to ussurin

    • Apply CyTOF (cytometry by time of flight) to analyze protein expression at single-cell resolution

    • Correlate cellular heterogeneity with differential responses to ussurin treatment

    • This approach is particularly valuable for understanding heterogeneous responses in platelet populations or tumor cells

What are the most common challenges in recombinant ussurin expression and activity assessment, and how can they be addressed?

Researchers frequently encounter several specific challenges when working with recombinant ussurin:

  • Low expression yields:

    • Optimize codon usage for the expression host

    • Test different signal sequences for secretion

    • Evaluate multiple fusion tags (His, MBP, SUMO) to improve solubility

    • Screen various promoter systems to optimize transcription levels

    • Consider testing several expression hosts (P. pastoris, mammalian cells, E. coli)

  • Loss of enzymatic activity:

    • Supplement all buffers with 10-50 μM Zn²⁺ ions

    • Add 1-5 mM Ca²⁺ to enhance stability

    • Store protein at -80°C in small aliquots to avoid freeze-thaw cycles

    • Add stabilizers like 10-20% glycerol to storage buffers

    • Test activity immediately after purification as the protein may lose activity over time

  • Inconsistent functional assay results:

    • Standardize substrate preparation in fibrin plate assays

    • Ensure consistent quality of fibrinogen for degradation studies

    • Use appropriate positive controls (native venom or other well-characterized SVMPs)

    • Perform platelet aggregation studies with freshly prepared platelets

    • Normalize activity based on protein concentration determined by multiple methods

  • Autoproteolysis during storage:

    • Add specific metalloproteinase inhibitors at low concentrations

    • Store at pH 6.5-7.0 to minimize autoprocessing

    • Keep samples at -80°C rather than 4°C for long-term storage

    • Add protease inhibitor cocktails that don't chelate zinc (avoid EDTA)

How should researchers address discrepancies between predicted and observed molecular weights or activity profiles of recombinant ussurin?

Discrepancies between theoretical predictions and experimental observations are common with SVMPs like ussurin:

  • Molecular weight discrepancies:

    • Assess glycosylation status using PNGase F treatment and SDS-PAGE comparison

    • Verify intact protein mass by mass spectrometry

    • Check for autoprocessing by N-terminal sequencing

    • Confirm full-length construct by epitope tag detection (if tags were incorporated)

    • Verify presence of all domains using domain-specific antibodies

  • Activity profile discrepancies:

    • Compare metal ion dependencies between recombinant and native protein

    • Assess the influence of pH on activity profiles as optimal pH may differ

    • Investigate substrate specificity differences using multiple substrates

    • Verify proper folding using circular dichroism and fluorescence spectroscopy

    • Consider the impact of expression host-specific post-translational modifications

  • Integration of multiple analytical approaches:

    • Combine biochemical assays with structural analysis techniques

    • Apply hydrogen-deuterium exchange mass spectrometry to identify regions with altered folding

    • Use limited proteolysis to compare domain stability between recombinant and native protein

    • Perform comparative molecular dynamics simulations to identify structural differences that may explain functional variations

What emerging technologies could enhance our understanding of ussurin structure-function relationships?

Several cutting-edge technologies offer new opportunities for ussurin research:

  • Cryo-electron microscopy (Cryo-EM):

    • Determine high-resolution structures of full-length ussurin without crystallization

    • Visualize conformational dynamics between domains

    • Capture protein-substrate complexes in native-like environments

    • This approach can overcome limitations of crystallography for flexible multi-domain proteins

  • Integrative structural biology:

    • Combine X-ray crystallography, NMR, SAXS, and computational modeling

    • Map conformational changes during catalysis

    • Characterize dynamic interactions between metalloproteinase and disintegrin domains

    • Understand the structural basis of substrate recognition

  • Advanced protein engineering:

    • Apply directed evolution approaches to modify specificity or activity

    • Use computational design to create novel variants with altered properties

    • Incorporate non-canonical amino acids to probe specific interactions

    • Develop activity-based probes for studying ussurin in complex biological systems

  • Single-molecule techniques:

    • Apply single-molecule FRET to study domain movements during catalysis

    • Use optical tweezers to investigate mechanical properties of ussurin-substrate interactions

    • Implement nanopore sensing for studying conformational changes

What interdisciplinary approaches might yield new insights into the therapeutic potential of ussurin-derived compounds?

Expanding research beyond traditional biochemistry offers new perspectives:

  • Nanomedicine applications:

    • Develop ussurin-functionalized nanoparticles for targeted drug delivery to cells expressing specific integrins

    • Create biosensors using the disintegrin domain for detecting integrin expression levels

    • Design nanostructured surfaces with immobilized ussurin for studying cell-matrix interactions

  • Computational drug discovery:

    • Perform virtual screening against ussurin active site to identify novel inhibitors

    • Apply molecular dynamics simulations to design improved RGD-mimetics

    • Use machine learning to predict bioactivity of ussurin-derived peptides

    • Develop network pharmacology models to predict system-wide effects

  • Integrative multi-omics approaches:

    • Combine transcriptomics, proteomics, and metabolomics to comprehensively map cellular responses to ussurin

    • Apply temporal multi-omics to understand the dynamic cellular response

    • Use network analysis to identify key regulatory nodes affected by ussurin

    • This approach can reveal unexpected connections between ussurin's effects and cellular pathways

  • Translational medicine perspectives:

    • Develop animal models to test ussurin-derived compounds for thrombotic diseases

    • Create cell-based screening platforms for identifying optimal derivatives

    • Apply pharmacokinetic/pharmacodynamic modeling to optimize dosing regimens

    • Investigate potential immunogenicity and strategies to minimize immune responses

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 2025 TheBiotek. All Rights Reserved.