Recombinant Tityus costatus Toxin-like TcoNTxP1

Shipped with Ice Packs
In Stock

Product Specs

Form
Lyophilized powder

Note: While we will prioritize shipping the format currently in stock, please specify your format preference in order notes for customized fulfillment.

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

Note: All proteins are shipped with standard 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 collect 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% and may serve as a guideline.
Shelf Life
Shelf life depends on various factors, including storage conditions, buffer components, 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 recommended for multiple uses. Avoid repeated freeze-thaw cycles.
Tag Info
The tag type is determined during the manufacturing process.

Tag type is determined during production. Please specify your required tag type for preferential development.

Synonyms
; Toxin-like TcoNTxP1; Insect-like toxic peptide Tco 38.32-2; PT-alpha* NaTx4.5
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Expression Region
20-81
Protein Length
Full Length of Mature Protein
Purity
>85% (SDS-PAGE)
Species
Tityus costatus (Brazilian scorpion)
Target Protein Sequence
G KEGYPADSKG CKVTCFLTAA GYCNTECKLQ KASSGYCAWP ACYCYGLPDS ASVWDSATNK C
Uniprot No.

Target Background

Function
This non-toxic protein elicits an immune response similar to that of whole venom. Consequently, polyclonal antibodies generated against this protein can neutralize venom effects.
Protein Families
Long (4 C-C) scorpion toxin superfamily, Sodium channel inhibitor family, Alpha subfamily
Subcellular Location
Secreted.
Tissue Specificity
Expressed by the venom gland.

Q&A

What is the biochemical composition of Tityus costatus venom and how does TcoNTxP1 fit into this profile?

Tityus costatus venom represents a complex mixture containing approximately 90 distinct components with molecular weights ranging from 413 to 45,482 atomic mass units . Generally, Tityus venoms primarily consist of neurotoxins (particularly those targeting sodium and potassium ion channels), metalloproteases, and peptides with hypotensive and antimicrobial properties . Within this composition, TcoNTxP1 would likely be categorized among the neurotoxins, which constitute the most abundant components of these venoms and are associated with autonomic nervous system stimulation through massive neurotransmitter release . The venom's complexity is further evidenced by high-performance liquid chromatography separation revealing at least 50 different components . When characterizing TcoNTxP1 or similar toxins, researchers should employ mass spectrometry for molecular weight determination, followed by N-terminal sequencing and cDNA library construction for complete structural elucidation.

What are the most effective methods for isolating and purifying native Tityus costatus toxin components prior to recombinant production?

A methodologically sound approach to isolating Tityus costatus toxin components involves a multi-step purification process:

  • Initial fractionation using size-exclusion chromatography to separate components based on molecular weight

  • Secondary purification via reversed-phase HPLC, which has demonstrated success in separating up to 50 different components from T. costatus venom

  • Confirmation of purified components through mass spectrometry analysis

  • Assessment of homogeneity using SDS-PAGE and isoelectric focusing

For subsequent recombinant production, researchers should consider:

  • Determining the complete amino acid sequence through Edman degradation or MS/MS analysis

  • Constructing a cDNA library from venom gland tissue for gene identification

  • Designing appropriate primers based on identified sequences

  • Optimizing codon usage for the selected expression system

When selecting an expression system, E. coli remains the most common choice for recombinant toxin production, though eukaryotic systems may be preferable for toxins requiring post-translational modifications, which account for approximately 80% of Tityus toxins .

What considerations should guide experimental design when evaluating the ion channel blocking activity of recombinant TcoNTxP1?

When designing experiments to evaluate the ion channel blocking activity of recombinant TcoNTxP1, researchers should implement a robust experimental framework that accounts for several critical factors:

  • Control for pH dependence: As demonstrated with TsTX-Kalpha from T. serrulatus, toxin activity can be significantly pH-dependent (with diminished effects below pH 7.0) . Design experiments with precisely controlled pH conditions, ideally testing activity across a pH range (6.0-8.0).

  • Channel specificity assessment: Include a comprehensive panel of ion channels, particularly focusing on:

    • Voltage-gated potassium channels (Kv1 family members)

    • Voltage-gated sodium channels (Nav subtypes)

    • Control channels from different families

  • Blocking mechanism characterization:

    • Implement electrophysiological techniques (patch-clamp) to determine whether the toxin affects channel kinetics, voltage-dependence, or pore blockage

    • Design protocols to distinguish between state-dependent binding (open vs. closed channels)

    • Consider site-directed mutagenesis of key residues to determine binding interface

  • Statistical considerations:

    • Use randomized block design to account for variability between cell preparations

    • Ensure adequate replication to detect statistically significant effects

    • Include positive controls (e.g., known Tityus toxins like TsTX-Kalpha) to validate experimental conditions

This systematic approach helps minimize experimental variability while maximizing the validity and reproducibility of results.

How should researchers optimize recombinant TcoNTxP1 expression systems to ensure proper folding and functionality?

Optimizing recombinant TcoNTxP1 expression requires addressing several critical parameters that influence proper protein folding and biological activity:

Expression SystemAdvantagesDisadvantagesOptimization Strategies
E. coliCost-effective, high yield, rapid growthLimited post-translational modifications, inclusion body formationUse periplasmic expression vectors with oxidizing environment; co-express chaperones; optimize codon usage; use fusion tags (e.g., thioredoxin)
Yeast (P. pastoris)Post-translational modifications, secretion capabilityLonger production time, glycosylation patterns differ from mammalsOptimize signal sequences; control methanol induction rate; implement temperature-shift strategies
Mammalian cellsNative-like post-translational modificationsHigher cost, lower yield, complex media requirementsUse transient expression systems; optimize transfection protocols; select appropriate cell lines (HEK293, CHO)
Insect cellsEfficient disulfide bond formation, moderate costModerate yield, glycosylation differencesOptimize viral titer; implement controlled infection strategies; regulate expression temperature

Toxin refolding strategies are particularly important given that TcoNTxP1 likely contains multiple disulfide bonds, similar to other Tityus toxins . A stepwise dialysis approach using a gradient of decreasing denaturant concentration combined with an optimized redox environment (GSH/GSSG ratio) often yields the best results. For validation, compare the recombinant toxin's activity with native forms using pharmacological assays and structural analyses (circular dichroism, NMR) to confirm proper folding .

What approaches should be used to investigate the structure-function relationship of TcoNTxP1 and its interaction with target ion channels?

Investigating structure-function relationships for TcoNTxP1 requires a comprehensive experimental strategy combining structural biology, computational approaches, and functional validation:

  • Structural characterization:

    • Determine the three-dimensional structure using NMR spectroscopy (preferred for small peptide toxins) or X-ray crystallography if possible

    • Compare structural features with other toxin-like peptides from Tityus species

    • Identify key structural elements (β-sheets, α-helices, disulfide bonding patterns)

  • Computational analyses:

    • Generate molecular models of TcoNTxP1-channel complexes based on available structures

    • Perform molecular dynamics simulations to predict critical interaction points

    • Use alanine scanning computational predictions to identify functionally important residues

  • Experimental validation:

    • Create a panel of site-directed mutants focusing on predicted functional residues (similar to K27 studies with TsTX-Kalpha)

    • Evaluate mutants using electrophysiological assays to quantify changes in binding affinity and kinetics

    • Implement complementary mutagenesis on target channels to confirm interaction sites

  • Advanced biophysical characterization:

    • Use surface plasmon resonance or isothermal titration calorimetry to determine binding kinetics and thermodynamics

    • Implement hydrogen-deuterium exchange mass spectrometry to map protein-protein interfaces

    • Consider fluorescence-based approaches (FRET) to study real-time binding dynamics

This multi-faceted approach would provide integrated insights into how specific structural elements of TcoNTxP1 contribute to its biological activity, potentially revealing unique features compared to other characterized Tityus toxins .

How can researchers address challenges in differentiating the effects of TcoNTxP1 from other components in complex experimental systems?

Differentiating the specific effects of TcoNTxP1 from other components in complex systems presents a significant challenge that requires meticulous experimental design. Researchers should implement:

  • Rigorous controls and comparisons:

    • Use highly purified recombinant TcoNTxP1 alongside native venom preparations

    • Include structurally similar but functionally distinct toxins as negative controls

    • Implement parallel experiments with known antagonists of specific channels/receptors

  • Selective neutralization strategies:

    • Develop specific antibodies against TcoNTxP1 for immunodepletion studies

    • Use RNA interference or CRISPR/Cas9 techniques to knock down expression of the putative target(s)

    • Implement competitive binding assays with known ligands

  • Channel-specific experimental platforms:

    • Utilize heterologous expression systems expressing only the channel of interest

    • Implement cell types with defined ion channel expression profiles

    • Consider reconstituted membrane systems with purified channels

  • Advanced analytical techniques:

    • Apply experimental blocking designs that can statistically separate the effects of multiple variables

    • Use multivariate analysis to identify distinct response signatures

    • Implement time-resolved measurements to differentiate kinetic profiles

These approaches minimize confounding variables while maximizing specificity for TcoNTxP1 effects, addressing a common challenge in toxin research. The methodology should account for the complex nature of scorpion venoms, which contain multiple toxins with potentially overlapping activities .

What are the most common pitfalls when characterizing recombinant Tityus toxins, and how can they be overcome?

Researchers frequently encounter several challenging issues when working with recombinant Tityus toxins:

  • Misfolded protein production:

    • Problem: Improper disulfide bond formation leading to inactive toxins

    • Solution: Implement oxidative folding conditions; consider periplasmic expression; use fusion partners that enhance disulfide bond formation; validate structural integrity through circular dichroism and activity assays

  • Low expression yields:

    • Problem: Poor expression of toxic peptides in host systems

    • Solution: Use inducible, tightly regulated expression systems; optimize codon usage for expression host; consider synthetic gene approaches; test multiple fusion tags for improved expression

  • Inconsistent activity measurements:

    • Problem: Variability in functional assays leading to contradictory results

    • Solution: Standardize experimental protocols; control for sample preparation variables; implement positive controls in each experiment; use multiple independent batches of recombinant protein

  • Non-specific binding artifacts:

    • Problem: False positives in binding or activity assays

    • Solution: Include appropriate negative controls; validate findings with multiple methodological approaches; implement dose-response studies; use competition assays with known ligands

  • Post-translational modification differences:

    • Problem: Recombinant toxins lacking native modifications affecting function

    • Solution: Characterize native toxin modifications; select expression systems capable of required modifications; implement analytical methods to confirm modification status; consider semi-synthetic approaches for specific modifications

By anticipating these common challenges, researchers can implement preventative strategies and contingency plans to ensure successful characterization of recombinant Tityus toxins.

How should researchers address data inconsistencies between recombinant TcoNTxP1 and native toxin studies?

When faced with data inconsistencies between studies using recombinant TcoNTxP1 and native toxin preparations, researchers should implement a systematic troubleshooting approach:

  • Verification of molecular identity:

    • Confirm sequence accuracy of the recombinant construct through DNA sequencing

    • Verify protein sequence through mass spectrometry and peptide mapping

    • Compare post-translational modifications between native and recombinant forms

  • Structural authentication:

    • Analyze secondary structure elements using circular dichroism

    • Compare disulfide bond patterns through non-reducing vs. reducing electrophoresis

    • Consider higher-resolution structural analysis when possible (NMR spectroscopy)

  • Methodological standardization:

    • Implement identical experimental conditions for both toxin forms

    • Use concentration-response curves rather than single-point measurements

    • Control for buffer composition, pH, and ionic strength which can significantly affect toxin activity

  • System-specific variables:

    • Evaluate the role of experimental models (cell type, expression system)

    • Consider channel subtype specificity and expression levels

    • Assess the impact of auxiliary proteins or subunits on toxin effects

  • Statistical approach:

    • Implement blocking in experimental design to reduce variability

    • Use power analysis to ensure adequate sample sizes

    • Consider meta-analytical approaches when comparing across studies

Documenting these systematic investigations helps identify whether discrepancies reflect actual biological differences between native and recombinant toxins or stem from methodological variability, advancing understanding of structure-function relationships.

What emerging methodologies could advance our understanding of TcoNTxP1 and related toxins beyond current approaches?

Several cutting-edge methodologies hold promise for deepening our understanding of TcoNTxP1 and related toxins:

  • Cryo-electron microscopy for toxin-channel complexes:

    • Recent advances in cryo-EM resolution now enable visualization of toxin-channel interactions at near-atomic resolution

    • This approach could reveal the precise binding interface of TcoNTxP1 with target channels

    • Sample preparation would involve purifying the channel-toxin complex in nanodiscs or detergent micelles

  • Single-molecule fluorescence techniques:

    • Implementing FRET-based approaches to monitor real-time binding events at the single-molecule level

    • This could reveal binding kinetics and potential conformational changes upon toxin binding

    • Requires strategic labeling of both toxin and channel proteins

  • Advanced transcriptomic and proteomic profiling:

    • Application of RNAseq to venom gland tissue under different conditions to understand toxin expression regulation

    • Quantitative proteomics to determine relative abundance and post-translational modifications

    • Integration of multiple -omics approaches for systems biology perspective

  • Computational toxicology advances:

    • Machine learning algorithms to predict toxin-channel interactions based on sequence and structural features

    • Molecular dynamics simulations with enhanced sampling techniques for more accurate binding predictions

    • Virtual screening to identify potential therapeutic applications or antidotes

  • Genome editing in model systems:

    • CRISPR/Cas9 modification of channels in model organisms to study toxin effects in vivo

    • Development of humanized animal models expressing relevant human ion channels

    • Creation of reporter systems for real-time visualization of toxin activity

These methodologies would complement existing approaches and potentially resolve current knowledge gaps regarding the mechanism of action, selectivity determinants, and physiological effects of TcoNTxP1 and related toxins .

What are the key experimental considerations for investigating potential therapeutic applications of TcoNTxP1?

Investigating potential therapeutic applications of TcoNTxP1 requires a comprehensive experimental framework addressing several critical aspects:

  • Target selectivity profiling:

    • Conduct comprehensive screening against a panel of ion channels, receptors, and transporters

    • Determine selectivity ratios for targets of interest versus off-targets

    • Implement cell-based functional assays alongside binding studies to confirm mechanism

  • Pharmacokinetic characterization:

    • Assess stability in biological fluids (plasma, cerebrospinal fluid)

    • Determine half-life and metabolism using in vitro and in vivo models

    • Evaluate tissue distribution and blood-brain barrier penetration if CNS targets are relevant

    • Consider strategies to enhance stability (cyclization, non-natural amino acids, PEGylation)

  • Safety assessment framework:

    • Design dose-escalation studies with comprehensive physiological monitoring

    • Implement focused assays for cardiovascular and neurological toxicity

    • Determine immunogenicity potential through in silico and in vitro approaches

    • Establish therapeutic index by comparing effective and toxic doses

  • Delivery system development:

    • Evaluate various administration routes (subcutaneous, intravenous, intrathecal)

    • Consider formulation approaches to enhance stability and targeting

    • Explore potential for peptide modifications to improve bioavailability

    • Implement controlled release systems for sustained activity

  • Appropriate disease models:

    • Select models that accurately reflect human pathophysiology

    • Include genetic models where appropriate (e.g., channelopathies)

    • Establish clear, quantifiable endpoints related to clinical outcomes

    • Design experiments with appropriate statistical power and controls

This comprehensive approach ensures rigorous evaluation of TcoNTxP1's therapeutic potential while addressing critical translational challenges that peptide therapeutics typically face. The experimental design should carefully balance initial preclinical characterization with downstream development considerations.

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.