CTSL Human

Cathepsin-L Human Recombinant
Shipped with Ice Packs
In Stock

Description

Introduction to CTSL Human

CTSL Human refers to the recombinant form of human Cathepsin L (CTSL), a lysosomal cysteine protease belonging to the peptidase C1 family. This enzyme is critical for intracellular protein degradation, extracellular matrix remodeling, and involvement in pathological processes such as viral entry, cancer metastasis, and cellular senescence . Produced via Escherichia coli expression systems, recombinant CTSL is widely used in biochemical research and therapeutic development due to its stability and functional activity .

Normal Physiological Functions

  • Protein Catabolism: Degrades intracellular proteins in lysosomes .

  • Extracellular Matrix Remodeling: Cleaves collagen and elastin, aiding tissue repair .

Disease Associations

  • Viral Infections: CTSL mediates SARS-CoV-2 entry by cleaving the viral spike protein . Elevated serum CTSL levels correlate with COVID-19 severity .

  • Metabolic Disorders: Hyperglycemia increases CTSL activity, exacerbating diabetic complications .

  • Cellular Senescence: Upregulates CUX1 and p16<sup>INK4a</sup>, driving vascular cell aging .

  • Neuroprotection: Rescues neurodegeneration in CTSB/CTSL double-deficient mice .

Engineered Mutants for Enhanced Function

  • CTSL-YLAL: A quadruple mutant (L69Y/M161L/G164A/A214L) with 30-fold increased specificity for proline-containing substrates, enabling collagen degradation in the presence of chondroitin sulfate .

  • CTSL-C4S: A collagenolytic mutant with modified surface residues (K9N/K195N/K196N) for glycosaminoglycan binding, enhancing collagenase activity .

Therapeutic Inhibitors

InhibitorTypeEfficacySource
Antibody-PropeptideHumanized antibody fusionPotent, selective inhibition (IC<sub>50</sub> < 1 nM)
PlumbaginNatural product>90% inhibition at 100 µM (uncompetitive)
Beta-LapachoneNatural product>90% inhibition at 100 µM (uncompetitive)
14a/14bDual-target inhibitorIC<sub>50</sub> = 3.34–347.6 nM (CTSL/CAPN1)

Clinical and Therapeutic Implications

  • COVID-19: Amantadine, an anti-influenza drug, inhibits CTSL activity and reduces SARS-CoV-2 pseudovirus infection in vitro and in vivo .

  • Diabetes: CTSL inhibitors like Plumbagin improve glucose tolerance and reduce insulin resistance .

  • Aging: Targeting CTSL-CUX1-p16<sup>INK4a</sup> axis may mitigate vascular senescence .

Expression Profiles and Tissue Distribution

  • High Expression: Lung, liver, placenta, and spleen .

  • mRNA Levels: 53.1 NX (normal lungs), 237.0 NX (placenta) .

  • Protein Levels: Medium in lungs and liver; undetectable in 31 tissues (e.g., colon) .

Future Directions

  • Drug Development: Optimize dual inhibitors targeting CTSL and related proteases (e.g., CAPN1) .

  • Gene Therapy: Explore CTSL overexpression/knockdown models for neurodegenerative diseases .

  • Diagnostic Biomarkers: Correlate circulating CTSL levels with disease progression in metabolic and viral disorders .

Product Specs

Introduction
Cathepsin-L, also known as CTSL, is a member of the peptidase C1 family. CTSL is a dimer composed of disulfide-linked heavy and light chains, both formed from a single protein precursor. It is a lysosomal cysteine proteinase that plays a major role in intracellular protein catabolism. Substrates of CTSL include collagen, elastin, and alpha-1 protease inhibitor, which is a key regulator of neutrophil elastase activity. CTSL has been implicated in several pathological processes, including myofibril necrosis in myopathies, myocardial ischemia, and the renal tubular response to proteinuria. Multiple alternatively spliced transcript variants of CTSL have been identified.
Description
Recombinant human CTSL, produced in E. coli, is a single, non-glycosylated polypeptide chain containing 339 amino acids (residues 18-333). It has a molecular weight of 38.3 kDa. The CTSL protein is fused to a 23-amino acid His-tag at the N-terminus and purified using proprietary chromatographic techniques.
Physical Appearance
A clear, sterile-filtered solution.
Formulation
The CTSL protein solution has a concentration of 1 mg/ml and contains 20 mM Tris-HCl (pH 8.0) and 10% glycerol.
Stability
For short-term storage (2-4 weeks), store the vial at 4°C. For long-term storage, freeze the product at -20°C. It is recommended to add a carrier protein (0.1% HSA or BSA) for extended storage. Avoid repeated freeze-thaw cycles.
Purity
Purity is greater than 90% as determined by SDS-PAGE analysis.
Synonyms
Cathepsin L, CTSL1, Cathepsin L1, Major Excreted Protein, MEP, EC 3.4.22.15, CATL, EC 3.4.22.
Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MGSTLTFDHS LEAQWTKWKA MHNRLYGMNE EGWRRAVWEK NMKMIELHNQ EYREGKHSFT MAMNAFGDMT SEEFRQVMNG FQNRKPRKGK VFQEPLFYEA PRSVDWREKG YVTPVKNQGQ CGSCWAFSAT GALEGQMFRK TGRLISLSEQ NLVDCSGPQG NEGCNGGLMD YAFQYVQDNG GLDSEESYPY EATEESCKYN PKYSVANDTG FVDIPKQEKA LMKAVATVGP ISVAIDAGHE SFLFYKEGIY FEPDCSSEDM DHGVLVVGYG FESTESDNNK YWLVKNSWGE EWGMGGYVKM AKDRRNHCGI ASAASYPTV.

Q&A

What is Cathepsin L (CTSL) and what distinguishes human CTSL from other species?

Human Cathepsin L is a widely expressed cysteine protease that predominantly resides within lysosomes. The key distinguishing characteristic between humans and other mammalian models is that humans possess two CTSL-like homologs (CTSL and CTSL2), whereas mice have only one CTSL enzyme. This difference creates important considerations when translating experimental findings from animal models to human applications . The human CTSL gene is located on chromosome 9q21.33, and the protein plays crucial roles in protein degradation within lysosomes, antigen processing, and extracellular matrix remodeling.

What are the essential molecular characteristics of human CTSL?

Human CTSL (also known as CATL, CTSL1, or MEP) is identified by the UniProt ID P07711 and the GenBank accession number NM_001912 . The protein is synthesized as a preproenzyme and undergoes several processing steps to become the mature, active enzyme. The mature CTSL is a single-chain protein approximately 220 amino acids in length with a molecular weight of about 30 kDa. Its structure includes a catalytic triad (Cys25, His159, and Asn175) that is essential for its proteolytic activity. CTSL functions optimally in slightly acidic environments (pH 5.0-5.5), reflecting its primary lysosomal localization.

How do human CTSL isoforms differ in their expression patterns and functions?

Human CTSL exists in multiple isoforms resulting from alternative splicing and post-translational modifications. The canonical isoform is widely expressed in most tissues, with particularly high levels in the kidney, liver, and immune cells. A notable characteristic of human CTSL is its dual localization – while primarily found in lysosomes, a significant portion can be secreted into the extracellular space or translocated to the nucleus under specific conditions . Nuclear CTSL has been implicated in histone H3 processing and chromatin remodeling, suggesting important roles beyond protein degradation. Different isoforms may possess varying substrate specificities and cellular localizations, which contributes to the diverse functions of CTSL in different physiological contexts.

What are the recommended methodologies for quantifying CTSL expression in human samples?

For quantifying CTSL expression at the transcriptional level, quantitative PCR (qPCR) remains the gold standard. When performing qPCR analysis of human CTSL, researchers should use properly validated qPCR template standards such as those based on the NM_001912 sequence . For protein-level quantification, Western blotting with specific anti-CTSL antibodies remains widely used, though careful validation is necessary to distinguish between proenzyme and mature forms of the protein.

For more comprehensive expression analysis, RNA-Seq provides insights into differential splicing patterns. When working with tissue samples, immunohistochemistry can reveal spatial distribution patterns, particularly important when studying CTSL in heterogeneous tissues. Activity-based assays using selective CTSL substrates or activity-based probes offer complementary functional data beyond mere expression levels. These methodological approaches should be selected based on specific research questions and available sample types.

What experimental models are most appropriate for studying human CTSL function?

Several experimental models have been developed for studying human CTSL function:

  • Cell culture systems: Human cell lines with either endogenous CTSL expression or engineered to overexpress/knock down CTSL provide accessible models. These systems allow for controlled manipulation of CTSL activity and observation of cellular consequences.

  • Transgenic mouse models: As demonstrated in the literature, transgenic mice expressing human CTSL in CTSL-deficient backgrounds offer valuable insights into functional rescue capabilities . These models are particularly useful for studying neurodegeneration and other systemic effects of CTSL modulation.

  • Patient-derived samples: Biospecimens from patients with conditions associated with CTSL dysregulation provide clinically relevant material, though variability between samples can present challenges.

  • In vitro enzymatic assays: Purified recombinant human CTSL can be used in biochemical assays to assess enzymatic properties, substrate specificities, and inhibitor profiles.

Each model system has strengths and limitations that researchers should consider when designing experiments to address specific questions about human CTSL function.

What challenges exist in designing selective inhibitors for human CTSL?

Designing selective inhibitors for human CTSL presents several significant challenges:

  • Structural homology: CTSL shares substantial structural similarity with other cathepsin family members, particularly cathepsin B, K, and S, making selective targeting difficult.

  • Dynamic active site: The active site of CTSL demonstrates conformational flexibility, which complicates structure-based inhibitor design.

  • Physiological redundancy: The potential functional redundancy between CTSL and other proteases means that inhibition of CTSL alone may result in compensatory mechanisms.

  • Clinical translation: While many inhibitors show promising results in vitro, achieving appropriate pharmacokinetic properties and tissue distribution in vivo remains challenging .

Recent advances using machine learning approaches coupled with structure-based virtual screening have shown promise in identifying natural CTSL inhibitors with improved selectivity profiles. These computational methods can screen large compound libraries to identify molecules with optimal binding characteristics for the CTSL active site while minimizing interactions with related cathepsins .

How is CTSL implicated in cancer progression and metastasis?

CTSL plays multiple roles in cancer progression and metastasis, with dysregulated expression observed across various cancer types . The mechanisms by which CTSL contributes to cancer pathophysiology include:

  • Extracellular matrix degradation: Secreted CTSL can degrade components of the basement membrane and extracellular matrix, facilitating tumor cell invasion and metastatic spread.

  • Cell signaling modulation: CTSL has been shown to process growth factors, cytokines, and cell surface receptors, potentially altering signaling pathways that promote tumor growth.

  • Angiogenesis promotion: CTSL contributes to the formation of new blood vessels within tumors by processing angiogenic factors and their inhibitors.

  • Treatment resistance: Elevated CTSL expression has been associated with resistance to various cancer therapies, including radiation and some chemotherapeutic agents .

  • Immune evasion: CTSL may help cancer cells evade immune surveillance by degrading components of the antigen presentation machinery.

These diverse mechanisms make CTSL an attractive target for anti-cancer therapeutic development, particularly for aggressive or treatment-resistant malignancies.

What is the current evidence for CTSL's role in neurodegenerative disorders?

Evidence from mouse models indicates that CTSL plays a neuroprotective role, particularly when CTSB is also deficient. CTSB/CTSL double-deficient mice exhibit severe neurodegeneration, including loss of Purkinje cells in the cerebellum and neurons in the cerebral cortex, resulting in pronounced motor deficits and premature death. Remarkably, transgenic expression of human CTSL in these double-deficient mice rescues the neurodegenerative phenotype and prevents lethality .

In human neurodegenerative conditions, the evidence is still emerging, but dysregulated lysosomal function—including altered cathepsin activity—has been implicated in conditions like Alzheimer's and Parkinson's diseases. CTSL may participate in the processing of proteins associated with neurodegenerative diseases, potentially influencing their aggregation properties. Additionally, the neuroprotective effects observed in mouse models suggest that enhancing CTSL activity might represent a therapeutic approach for certain neurodegenerative conditions, though careful consideration of context-specific effects is necessary.

How does human CTSL contribute to viral infections, including SARS-CoV-2?

Human CTSL has emerged as an important host factor in multiple viral infections. For SARS-CoV-2 specifically, CTSL has been identified as a potential facilitator of viral entry through its role in processing the viral spike protein. The enzyme may cleave the S protein at specific sites, promoting fusion of the viral envelope with host cell membranes.

Beyond SARS-CoV-2, CTSL has been implicated in the life cycles of other viruses:

  • Ebola virus: CTSL can process the Ebola virus glycoprotein, facilitating viral entry.

  • Hendra and Nipah viruses: CTSL contributes to the activation of these paramyxovirus fusion proteins.

  • Reovirus and SARS-CoV-1: CTSL-mediated proteolysis has been shown to enhance infection with these viruses.

How do post-translational modifications regulate human CTSL activity and localization?

Human CTSL undergoes several post-translational modifications that significantly impact its activity, stability, and subcellular localization:

  • Glycosylation: N-linked glycosylation at multiple sites affects CTSL folding, stability, and trafficking. Alterations in glycosylation patterns can redirect CTSL from lysosomes to secretory pathways.

  • Proteolytic processing: CTSL is synthesized as an inactive proenzyme that requires proteolytic removal of the propeptide for activation. This can occur autocatalytically at acidic pH or through the action of other proteases.

  • Phosphorylation: Phosphorylation at specific serine and threonine residues has been reported to modulate CTSL activity and influence its interactions with regulatory partners.

  • Oxidation: The active site cysteine is susceptible to oxidation, which can reversibly or irreversibly inactivate the enzyme, representing an important regulatory mechanism in oxidative stress conditions.

Understanding these modifications is crucial for interpreting CTSL behavior in different cellular contexts and for developing therapeutic strategies that might selectively target specific modified forms of the enzyme.

What are the key similarities and differences in substrate specificity between human and mouse CTSL?

Human and mouse CTSL share approximately 75% sequence identity, with higher conservation in the catalytic domains. Despite this similarity, several notable differences in substrate specificity have been observed:

FeatureHuman CTSLMouse CTSLImplications for Research
Collagen degradationHigher activityLower activityMay affect interpretation of fibrosis models
MHC class II processingSpecific cleavage patternsDifferent cleavage preferencesImmune response studies may not directly translate
Inhibitor sensitivityDistinctive profileDifferent affinities for some inhibitorsRequires human-specific validation of inhibitors
pH optimumSlightly broader rangeNarrower pH optimumAffects activity in different subcellular compartments
Nuclear substratesProcesses specific histonesDifferent histone preferencesEpigenetic regulation studies need careful interpretation

These differences underline the importance of validating findings from mouse models using human CTSL, as demonstrated by transgenic approaches where human CTSL can functionally replace mouse CTSL in vivo .

How can machine learning approaches advance the discovery of selective CTSL inhibitors?

Machine learning (ML) approaches offer powerful tools for accelerating the discovery of selective CTSL inhibitors. Recent research has employed random forest classification models trained on compound activity data from the CHEMBL database to identify potential natural CTSL inhibitors . The methodology involves several sophisticated steps:

  • Feature extraction and representation: Morgan fingerprints (1024 bits) can be calculated for active and inactive molecules, providing a mathematical representation of chemical structures suitable for ML analysis.

  • Model training and validation: Random forest models trained on these fingerprints can achieve high predictive performance, with area under the ROC curve (AUC) values of approximately 0.91 in cross-validation tests .

  • Virtual screening: Trained models can rapidly screen large libraries of natural compounds to prioritize candidates for experimental testing.

  • Molecular dynamics simulations: Top candidates from ML screening can be further evaluated using detailed simulations to assess binding stability and selectivity.

  • Structure-activity relationship analysis: ML models can identify key structural features that contribute to CTSL inhibitory activity, guiding further optimization efforts.

This integrated approach has identified promising compounds like ZINC4098355, which demonstrates stable binding to CTSL in molecular dynamics simulations . The advantage of ML-based methods is their ability to efficiently navigate large chemical spaces while learning complex patterns of structure-activity relationships that might not be apparent through traditional medicinal chemistry approaches.

What are the best practices for measuring CTSL enzymatic activity in complex biological samples?

Measuring CTSL enzymatic activity in complex biological samples requires careful consideration of several methodological aspects:

  • Selective substrates: Use substrates with high selectivity for CTSL, such as Z-Phe-Arg-AMC, but include controls with specific inhibitors (e.g., E-64, cathepsin B inhibitors) to distinguish CTSL activity from other cathepsins.

  • pH optimization: Perform assays at pH 5.5 (optimal for CTSL) and compare with pH 7.4 to distinguish between lysosomal and extracellular/cytosolic activity.

  • Sample preparation: Careful homogenization and fractionation techniques preserve enzyme activity and allow analysis of CTSL in different subcellular compartments.

  • Activity-based probes: Consider using activity-based probes that covalently bind to active CTSL, allowing visualization and quantification of active enzyme rather than total protein.

  • Normalization methods: Normalize activity to total protein content, cell number, or tissue weight depending on the experimental context.

  • Native vs. recombinant standards: Include purified human recombinant CTSL as a positive control and for generating standard curves.

These methodological considerations ensure reliable quantification of CTSL activity and facilitate meaningful comparisons between different experimental conditions or clinical samples.

How should researchers approach CTSL knockout or knockdown studies in human cell models?

When designing CTSL knockout or knockdown studies in human cell models, researchers should consider:

  • Choice of technique:

    • CRISPR-Cas9 for complete knockout

    • shRNA for stable knockdown

    • siRNA for transient knockdown

    • Each approach has different advantages depending on the research question

  • Target specificity: Design targeting sequences that specifically affect CTSL without cross-reactivity with CTSL2 or other cathepsins.

  • Validation approach: Validate knockdown/knockout at multiple levels:

    • mRNA (qPCR)

    • Protein (Western blot)

    • Enzymatic activity (fluorogenic substrate assay)

  • Compensatory mechanisms: Assess potential upregulation of other cathepsins (especially CTSB and CTSL2) that might compensate for CTSL loss.

  • Phenotypic analyses: Include comprehensive phenotypic analyses:

    • Lysosomal morphology and function

    • Cell viability and proliferation

    • Specific CTSL-dependent processes relevant to research question

  • Rescue experiments: Include rescue experiments with wild-type human CTSL to confirm specificity of observed phenotypes.

What considerations are important when translating CTSL research findings from in vitro to in vivo settings?

Translating CTSL research findings from in vitro to in vivo settings requires careful consideration of several factors:

  • Physiological relevance: Cell culture conditions rarely recapitulate the complex environment of intact tissues. Consider the influence of extracellular matrix, neighboring cell types, and systemic factors on CTSL activity and function.

  • Species differences: As highlighted in transgenic studies, human and mouse CTSL have distinct properties . Consider species-specific differences when designing animal studies or interpreting results for human relevance.

  • Tissue distribution: CTSL expression and activity vary significantly across tissues. Findings in one tissue or cell type may not generalize to others.

  • Pharmacodynamics and pharmacokinetics: For CTSL inhibitor studies, consider bioavailability, tissue penetration, half-life, and potential off-target effects in vivo that might not be apparent in vitro.

  • Disease context: The role of CTSL may differ substantially between normal physiological conditions and disease states, particularly in cancer and inflammatory conditions where the microenvironment is dramatically altered .

  • Genetic redundancy: The presence of multiple cathepsins with overlapping functions means that compensatory mechanisms may mask phenotypes in vivo that are apparent in simplified in vitro systems.

Researchers can address these challenges by using multiple complementary approaches, including human tissue samples, primary cells, organoids, and carefully designed animal models expressing human CTSL .

What role might CTSL play in emerging therapeutic approaches for neurodegenerative diseases?

Based on the neuroprotective effects demonstrated in mouse models , CTSL modulation represents a promising avenue for neurodegenerative disease therapeutics. Several potential approaches are being explored:

  • CTSL enhancement strategies: Given that human CTSL expression can rescue neurodegeneration in mouse models lacking endogenous CTSL , therapeutic approaches might include:

    • Gene therapy to deliver CTSL to affected brain regions

    • Small molecules that enhance CTSL expression or activity

    • Agents that promote proper CTSL trafficking to lysosomes

  • Targeting specific substrates: Rather than modulating CTSL itself, therapeutic approaches might target specific CTSL substrates implicated in neurodegeneration:

    • Preventing cleavage of protective factors

    • Promoting degradation of neurotoxic proteins

  • Combinatorial approaches: Since neurodegeneration often involves multiple proteolytic systems, combination therapies targeting CTSL alongside other proteases may prove more effective than single-target approaches.

  • Cell-type specific interventions: Given the importance of CTSL in specific neuronal populations like Purkinje cells , developing targeted delivery systems for these cell types could maximize therapeutic benefits while minimizing off-target effects.

These approaches require further validation in preclinical models, but the ability of human CTSL to rescue neurodegeneration in mice suggests significant therapeutic potential.

How might advances in structural biology and computational methods accelerate CTSL inhibitor development?

Recent advances in structural biology and computational methods are transforming CTSL inhibitor development:

  • Cryo-electron microscopy: High-resolution structures of CTSL in complex with various ligands can reveal subtle binding interactions and conformational changes that might be exploited for inhibitor design.

  • Fragment-based drug design: Identifying small molecular fragments that bind to different regions of CTSL can lead to novel inhibitor scaffolds with improved properties.

  • Molecular dynamics simulations: Extended simulations of CTSL-inhibitor interactions can reveal binding mechanisms and conformational flexibility not apparent in static crystal structures .

  • Quantum mechanical calculations: More accurate modeling of electronic interactions between CTSL and potential inhibitors can improve prediction of binding affinities and reaction mechanisms.

  • Deep learning approaches: Beyond traditional ML models, deep learning architectures can identify complex patterns in structure-activity relationships that guide inhibitor optimization .

  • In silico ADMET prediction: Computational prediction of absorption, distribution, metabolism, excretion, and toxicity properties can prioritize compounds with favorable drug-like characteristics early in development.

The integration of these computational methods with experimental validation creates a powerful iterative approach to CTSL inhibitor discovery, potentially accelerating the development of clinically viable compounds.

What are the most promising translational applications of CTSL research in precision medicine?

CTSL research holds significant promise for several precision medicine applications:

  • Cancer therapeutics and diagnostics:

    • CTSL expression profiles may serve as biomarkers for cancer aggressiveness and treatment response

    • CTSL inhibitors might selectively target cancer cells with elevated CTSL activity

    • Combination therapies targeting CTSL alongside standard treatments may overcome resistance mechanisms

  • Neurodegenerative disease interventions:

    • Genotyping CTSL variants could identify patients at higher risk for specific neurodegenerative conditions

    • CTSL enhancement strategies might be personalized based on individual protease activity profiles

    • Monitoring CTSL activity could serve as a biomarker for treatment response

  • Infectious disease applications:

    • CTSL inhibitors might serve as broad-spectrum antiviral agents for susceptible viruses

    • Individual variations in CTSL activity could explain differential susceptibility to certain infections

    • Targeting CTSL might complement standard antimicrobial therapies

  • Autoimmune condition management:

    • Given CTSL's role in antigen processing, personalized modulation of CTSL activity might help manage specific autoimmune conditions

    • CTSL activity profiles could predict response to immunomodulatory therapies

These translational applications highlight the potential impact of CTSL research across multiple disease domains and underscore the importance of continued investigation into this multifunctional protease.

Product Science Overview

Structure and Function

Cathepsin-L is synthesized as an inactive preproenzyme. The cleavage of its 96-residue proregion is necessary to generate the fully active 221-residue mature enzyme . The enzyme is potent in degrading collagen, laminin, elastin, and other structural proteins of basement membranes . It also hydrolyzes a number of proteins, including the proform of urokinase-type plasminogen activator, which is activated by Cathepsin-L cleavage .

Biological Activity

Cathepsin-L is involved in numerous physiological processes and pathological conditions. It participates in apoptosis, antigen processing, and extracellular matrix remodeling. These functions are implicated in various diseases, including tumor invasion and metastasis, inflammatory conditions, atherosclerosis, renal disease, diabetes, bone diseases, and viral infections .

Recombinant Human Cathepsin-L

Recombinant human Cathepsin-L is produced using various expression systems, including mouse myeloma cell lines and HEK293 cells . The recombinant protein is typically purified to high levels of purity, often exceeding 90% as determined by SDS-PAGE and HPLC . It is available in different formulations, including carrier-free versions that do not contain bovine serum albumin (BSA), which can interfere with certain applications .

Applications

Recombinant human Cathepsin-L is used in various research applications, including studies on protein degradation, cell cycle regulation, and disease mechanisms. It is also used in assays to measure its activity, such as the ability to cleave fluorogenic peptide substrates .

Storage and Stability

Recombinant human Cathepsin-L is typically shipped with dry ice or equivalent and should be stored at -20 to -70°C to maintain its stability. It is recommended to avoid repeated freeze-thaw cycles to preserve its activity .

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.