KLK2 Human

Kallikrein-2 Human Recombinant
Shipped with Ice Packs
In Stock

Description

KLK2 is overexpressed in prostate cancer and correlates with tumor aggressiveness:

  • Biomarker Potential:

    • KLK2 mRNA levels in blood and tissues show 92% accuracy for prostate cancer detection (sensitivity: 59%, specificity: 82%) .

    • In metastatic castration-resistant prostate cancer (mCRPC), KLK2 expression is linked to androgen receptor (AR) signaling, with 80% of bone lesions exhibiting AR-driven KLK2/KLK3 co-expression .

  • Therapeutic Target:

    • A Phase 0 study demonstrated selective accumulation of anti-KLK2 antibody h11B6 in mCRPC metastases, supporting its use for imaging and targeted therapy .

Recombinant KLK2 Production

Recombinant KLK2 is used in research to study protease mechanisms and inhibitor development. Key products include:

ProductSourceMolecular WeightTagCatalog Numbers
LS-G2393 (LSBio)E. coli28.5 kDaN-terminal HisLS-G2393-20, LS-G2393-100
ENZ-719 (Prospec)E. coli28.5 kDaN-terminal HisENZ-719

Both variants retain enzymatic activity and are stabilized in Tris-HCl buffer (pH 8.0) with glycerol .

Research Applications

  • Mechanistic Studies: Structural analyses (e.g., PDB entries 4NFE, 4NFF) reveal Zn²⁺ binding at the 99-loop/active site interface, informing inhibitor design .

  • Disease Modeling: KLK2 knockdown reduces AR-mediated proliferation in castration-resistant cell lines .

Future Directions

Current research focuses on:

  1. Developing KLK2-targeted radioligands for mCRPC imaging .

  2. Optimizing 99-loop mutants to enhance enzymatic stability for industrial applications .

Product Specs

Introduction
Kallikrein-related peptidase 2 (KLK2) belongs to the glandular kallikrein family, a group of serine proteases with diverse biological roles. These proteases cluster on chromosome 19. KLK2 displays potent trypsin-like activity, preferentially cleaving proteins at arginine residues. Primarily found in the prostate, it's responsible for converting inactive prostate-specific antigen (PSA) into its active form. KLK2's elevated expression in prostate tumor cells suggests its potential as a prognostic marker for prostate cancer risk.
Description
Recombinant human KLK2, expressed in E. coli, is a single polypeptide chain with 260 amino acids (residues 25-261). It possesses a molecular weight of 28.5kDa. A 23 amino acid His-tag is fused to the N-terminus to facilitate purification, achieved through proprietary chromatographic techniques.
Physical Appearance
Clear, colorless solution, sterile-filtered.
Formulation
KLK2 is supplied in a solution containing 20mM Tris-HCl buffer at pH 8.0, 0.4M urea, and 10% glycerol. The protein concentration is 1mg/ml.
Stability
For short-term storage (up to 4 weeks), keep at 4°C. For extended storage, freeze at -20°C. Adding a carrier protein like HSA or BSA (0.1%) is advisable for long-term storage. Minimize repeated freezing and thawing.
Purity
Purity exceeds 90% based on SDS-PAGE analysis.
Synonyms
hK2, KLK2A2, Kallikrein-2, Glandular kallikrein-1, hGK-1, issue kallikrein-2, KLK2.
Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MGSIVGGWEC EKHSQPWQVA VYSHGWAHCG GVLVHPQWVL TAAHCLKKNS QVWLGRHNLF EPEDTGQRVP VSHSFPHPLY NMSLLKHQSL RPDEDSSHDL MLLRLSEPAK ITDVVKVLGL PTQEPALGTT CYASGWGSIE PEEFLRPRSL QCVSLHLLSN DMCARAYSEK VTEFMLCAGL WTGGKDTCGG DSGGPLVCNG VLQGITSWGP EPCALPEKPA VYTKVVHYRK WIKDTIAANP.

Q&A

What is human kallikrein 2 (hK2) and how does it relate to the kallikrein family?

Human kallikrein 2 (hK2) is a serine protease encoded by the KLK2 gene, belonging to the kallikrein family of proteases. It is a trypsin-like enzyme with highly tissue-specific expression, predominantly found in the luminal epithelium of the prostate. The kallikrein family consists of 15 members (KLK1-15) with varying substrate specificity and tissue expression patterns. hK2 is characterized by its abundant expression in the prostate, where its levels are many orders of magnitude higher than in any other tissue .

hK2 is synthesized as a preproenzyme requiring multiple post-translational modifications to become catalytically active. This includes proteolytic cleavage of the signal sequence by a signal peptidase, followed by release of a short N-terminal activation peptide by trypsin-like peptidases, resulting in an enzymatically active 237-amino acid single-chain form . The kallikrein family shares significant sequence homology but differs in substrate specificity and physiological functions.

How is KLK2 expression regulated at the molecular level?

KLK2 expression is primarily regulated by androgens via the androgen receptor (AR) signaling pathway. The KLK2 gene contains androgen response elements (AREs) in its promoter region that bind activated AR, leading to transcriptional activation. This explains why KLK2 expression reflects the functional status and activity of the nuclear androgen receptor and its response to testosterone or other androgens .

This androgen-driven expression profile makes KLK2 a valuable biological indicator of prostate epithelium activity. The regulation mechanism involves:

  • Testosterone binding to AR

  • Translocation of the AR-androgen complex to the nucleus

  • Binding to AREs in the KLK2 promoter

  • Recruitment of co-activators and RNA polymerase II

  • Initiation of KLK2 transcription

Unlike some kallikreins, KLK2 expression is highly restricted to prostatic tissue, with minimal expression elsewhere, suggesting tissue-specific regulatory mechanisms beyond androgen regulation.

What is the functional relationship between hK2 and prostate-specific antigen (PSA)?

Their functional relationship includes:

  • Enzymatic activation: hK2 is capable of activating pro-PSA by cleaving its propeptide, converting it from inactive zymogen to catalytically active PSA .

  • Semen liquefaction: Both enzymes participate in the liquefaction of semen, with PSA cleaving semenogelin I and II proteins to release motile sperm, potentially activated by hK2 .

  • Biomarker complementarity: In clinical settings, both proteins serve as biomarkers for prostatic diseases, with evidence suggesting that combined measurements may improve diagnostic accuracy.

  • Genetic correlation: Variants in KLK2 have been associated with both hK2 and PSA levels, indicating shared genetic determinants of expression and activity .

Both proteins have similar secretion patterns, being released primarily into seminal fluid but also detectable at much lower concentrations in serum, particularly in cases of prostatic disease.

What are the current gold standard methods for measuring hK2 in clinical and research samples?

The gold standard for measuring hK2 in clinical and research samples is immunofluorometric assay (IFMA) with specific monoclonal antibodies. This methodology provides high sensitivity and specificity, which is crucial given that hK2 levels in serum are typically 100-fold lower than PSA levels.

A refined IFMA method for hK2 measurement includes:

  • Capture antibody: Biotinylated monoclonal antibody (Mab) 6H10 specific to hK2

  • Detection antibody: Mab 7G1-Eu labeled with europium for fluorometric detection

  • PSA blocking: Three PSA-specific anti-PSA monoclonal antibodies (2E9, 5F7, and 5H6) that do not cross-react with hK2, ensuring specificity

  • Sensitivity enhancement: Increased sample volume and enhanced labeling of the tracer antibody

Using this methodology, researchers have achieved a coefficient of variation (CV) of approximately 12% at a mean concentration of 0.008 mg/mL in seminal plasma . For serum measurements, the assays typically have detection limits in the ng/L range.

When measuring hK2 in complex matrices like seminal plasma or serum, careful sample handling and processing are essential to prevent degradation or inactivation of the protein.

How should researchers design experiments to study KLK2 genetic variants and their functional effects?

When designing experiments to study KLK2 genetic variants and their functional effects, researchers should employ a comprehensive approach incorporating genomic, proteomic, and functional analyses:

  • SNP selection strategy:

    • Focus on SNPs with known association to hK2 levels or disease risk

    • Include SNPs across the entire KLK2 gene region including regulatory elements

    • Consider linkage disequilibrium patterns to select tag SNPs

    • Include SNPs rs198972, rs198977, rs198978, and rs80050017, which have shown strong associations with hK2 levels

  • Genotyping methodology:

    • Use high-throughput methods such as Sequenom MassARRAY MALDI-TOF analysis

    • Ensure assay design with appropriate software (e.g., MassARRAY Assay Design 2.0)

    • Include quality control samples and check for Hardy-Weinberg equilibrium

  • Population selection:

    • Use well-characterized cohorts with detailed clinical information

    • Consider age-related effects (young healthy individuals versus older individuals with potential prostatic conditions)

    • Include sufficient sample size for adequate statistical power

  • Functional validation:

    • Employ reporter gene assays to assess effects on transcriptional activity

    • Use site-directed mutagenesis to introduce specific variants

    • Assess effects on protein expression, secretion, and enzymatic activity

    • Consider in vivo models for physiological relevance

  • Statistical analysis:

    • Test for genotype-phenotype associations using appropriate statistical tests (e.g., Kruskal-Wallis test)

    • Calculate linkage disequilibrium parameters (D′) using software like Haploview

    • Adjust for multiple testing and potential confounding factors

What techniques enable accurate differentiation between hK2 and PSA in experimental protocols?

Accurate differentiation between hK2 and PSA in experimental protocols is crucial due to their high sequence homology (approximately 80%). The following techniques enable reliable discrimination:

  • Immunological methods:

    • Use of highly specific monoclonal antibodies that recognize unique epitopes on each protein

    • Implementation of sandwich immunoassays with capture and detection antibodies validated for non-cross-reactivity

    • Incorporation of blocking antibodies against the non-target protein (e.g., anti-PSA antibodies in hK2 assays)

  • Mass spectrometry-based approaches:

    • Peptide mapping targeting unique sequence regions

    • Multiple reaction monitoring (MRM) of specific peptides unique to each protein

    • Isotope dilution mass spectrometry using labeled internal standards

  • Enzymatic activity differentiation:

    • Exploitation of differential substrate specificity (hK2 has trypsin-like activity while PSA has chymotrypsin-like activity)

    • Use of specific chromogenic or fluorogenic substrates

    • Implementation of specific inhibitors for either enzyme

  • Genetic analyses:

    • PCR-based methods targeting specific regions of KLK2 and KLK3 genes

    • Allele-specific PCR for variants unique to each gene

    • Digital PCR for absolute quantification of transcript levels

When combining these approaches, researchers can achieve reliable differentiation between these closely related proteins even in complex biological samples where both may be present simultaneously.

How can hK2 be utilized as a complementary biomarker to PSA in prostate cancer research?

hK2 offers significant potential as a complementary biomarker to PSA in prostate cancer research through several mechanisms:

  • Enhanced specificity in diagnosis:

    • The ratio of hK2 to free PSA (%fPSA) may improve discrimination between benign prostatic hyperplasia and prostate cancer

    • Certain genetic variants in KLK2 are associated with both hK2 levels and %fPSA in serum, potentially improving risk stratification

    • hK2 may show differential expression patterns in aggressive versus indolent tumors

  • Combined marker panels:

    • Incorporation of hK2 into multi-marker panels with PSA isoforms and other biomarkers

    • Development of algorithms integrating hK2, total PSA, free PSA, and clinical parameters

    • Statistical models incorporating genetic information from both KLK2 and KLK3 variants

  • Androgen-axis monitoring:

    • Both hK2 and PSA reflect androgen receptor activity in prostate tissue

    • Changes in hK2:PSA ratios may indicate alterations in androgen signaling during progression to castration-resistant prostate cancer

    • Monitoring both markers may provide insight into treatment resistance mechanisms

  • Imaging applications:

    • Development of hK2-targeted imaging agents such as [111In]-DOTA-h11B6 monoclonal antibody

    • Visualization of metastatic lesions through selective accumulation in tumor tissue

    • Potential for theranostic applications combining diagnostic imaging and targeted therapy

For optimal implementation, researchers should consider genetic background of patients, as genetic variants in KLK2 and KLK3 significantly impact baseline levels of these biomarkers in both healthy individuals and cancer patients .

What are the emerging therapeutic approaches targeting hK2 in cancer treatment?

Emerging therapeutic approaches targeting hK2 in cancer treatment represent a promising frontier in prostate cancer research, with several strategies under investigation:

  • Targeted radiopharmaceuticals:

    • Development of radiolabeled anti-hK2 monoclonal antibodies such as [111In]-DOTA-h11B6

    • First-in-human phase 0 trials demonstrating selective accumulation in metastatic castration-resistant prostate cancer (mCRPC) lesions

    • Potential progression to therapeutic radioisotopes for targeted radiotherapy

  • Antibody-drug conjugates (ADCs):

    • Engineering of anti-hK2 antibodies conjugated to cytotoxic payloads

    • Selective delivery of potent anti-cancer agents to hK2-expressing tumor cells

    • Potential to overcome resistance mechanisms to current therapies

  • Immune-based approaches:

    • Development of chimeric antigen receptor (CAR) T-cells targeting hK2

    • Bispecific antibodies engaging immune effector cells with hK2-expressing tumor cells

    • Therapeutic vaccines inducing immune responses against hK2-expressing cells

  • Enzymatic inhibition strategies:

    • Design of small molecule inhibitors of hK2 enzymatic activity

    • Disruption of hK2's role in tumor growth, invasion, and metastasis

    • Combination with androgen-targeted therapies for synergistic effects

The first-in-human phase 0 trial with [111In]-DOTA-h11B6 demonstrated that:

  • The antibody visibly accumulated in known mCRPC lesions within 6-8 days of administration

  • Limited uptake was observed in normal organs

  • Serum clearance and tumor targeting were independent of total antibody mass (2 or 10 mg)

  • The approach had a favorable safety profile with no treatment-related adverse events

These findings credential hK2 as a promising target for prostate cancer treatment, particularly in advanced disease settings where new therapeutic options are urgently needed.

How do genetic variations in KLK2 influence hK2 levels and what are the implications for personalized medicine?

Genetic variations in KLK2 have substantial impacts on hK2 levels with significant implications for personalized medicine approaches:

  • Strong genotype-phenotype correlations:

    • Four KLK2 SNPs (rs198972, rs198977, rs198978, and rs80050017) exhibit robust associations with hK2 levels

    • Individuals homozygous for major alleles demonstrate 3- to 7-fold higher hK2 levels compared to minor allele homozygotes

    • Heterozygotes display intermediate hK2 levels, suggesting an additive genetic effect

  • Impact on both seminal plasma and serum levels:

    • Genetic associations are consistent across different biological compartments

    • Suggests that genetic variation influences basal expression and secretion of hK2 rather than disease-specific alterations

    • Provides evidence for germline genetic determinants of hK2 production

  • Implications for biomarker interpretation:

    • Genotype-dependent reference ranges may be necessary for accurate interpretation of hK2 as a biomarker

    • SNP genotyping could enhance the specificity of hK2-based diagnostic approaches

    • Combined genetic and protein biomarker panels may improve risk stratification

  • Personalized screening strategies:

    • Incorporation of KLK2 genetics into risk prediction models

    • Tailored screening intervals and cut-off values based on genetic profile

    • Modified interpretation of changes in hK2 levels over time

  • Therapeutic targeting considerations:

    • Genetic variants may influence response to hK2-targeted therapies

    • Potential impact on antibody binding or enzymatic inhibition

    • Genotyping may help select patients most likely to benefit from hK2-targeted approaches

Table 1: Association of selected KLK2 SNPs with hK2 levels in young healthy men

SNP IDMajor/Minor AlleleEffect on hK2 in Seminal PlasmaEffect on hK2 in SerumP-value
rs198972C/T3-7 fold higher in major allele homozygotesSignificant positive association<0.001
rs198977T/C3-7 fold higher in major allele homozygotesSignificant positive association<0.001
rs198978A/G3-7 fold higher in major allele homozygotesSignificant positive association<0.001
rs80050017A/T3-7 fold higher in major allele homozygotesSignificant positive association<0.001

These findings highlight the importance of considering genetic background when developing precision medicine approaches targeting hK2 or utilizing it as a biomarker .

How should researchers interpret age-dependent variations in hK2 expression patterns?

Interpreting age-dependent variations in hK2 expression patterns requires careful consideration of multiple physiological and pathological factors:

  • Baseline age-related changes:

    • Young healthy men show stable hK2 levels with low intra-individual variability (less than 10%)

    • Older men may demonstrate increased hK2 levels independent of disease

    • Age-related changes in prostatic morphology and androgen sensitivity influence hK2 expression

  • Methodological considerations for age-comparative studies:

    • Separate cohorts by age decades for meaningful comparisons

    • Adjust for prostate volume and PSA levels as potential confounders

    • Consider genetic variants known to influence hK2 baseline levels

    • Account for subclinical prostatic conditions in older populations

  • Distinguishing pathological from physiological changes:

    • Young cohorts (e.g., median age 18.1 years) serve as optimal reference populations due to minimal confounding by prostatic conditions

    • In older men, hK2 alterations must be interpreted in the context of potential benign prostatic hyperplasia, prostatitis, or cancer

    • Longitudinal studies are preferred over cross-sectional approaches to track individual-level changes over time

  • Research design recommendations:

    • Include age-matched controls in all studies of hK2 as a disease biomarker

    • Establish age-specific reference ranges stratified by genetic variants

    • Consider the ratio of hK2 to PSA rather than absolute hK2 levels alone

    • Integrate imaging and biopsy data to correlate hK2 levels with histopathological findings

Researchers should recognize that the physiological role of hK2 may evolve with age, potentially reflecting changing reproductive biology and adaptive responses to environmental factors throughout the lifespan.

What statistical approaches are recommended for analyzing genetic association data for KLK2 variants?

Analyzing genetic association data for KLK2 variants requires robust statistical methodologies to ensure valid and reproducible findings:

  • Preliminary data quality assessment:

    • Test for Hardy-Weinberg equilibrium using Fisher's exact test

    • Remove SNPs showing significant deviation (e.g., rs11670728 in prior studies)

    • Assess linkage disequilibrium (LD) between SNPs using D′ calculations

    • Consider pruning redundant SNPs in high LD to minimize multiple testing burden

  • Association testing methodology:

    • For continuous phenotypes (hK2, PSA levels): Use non-parametric Kruskal-Wallis test to avoid assumptions of normal distribution

    • For categorical outcomes: Implement logistic regression with appropriate covariates

    • Consider genotypic (comparing three genotypes), allelic (comparing two alleles), and dominant/recessive models

    • Adjust for relevant clinical covariates including age, BMI, and prostate volume

  • Multiple testing correction:

    • Apply Bonferroni correction for independent tests

    • Use false discovery rate (FDR) approaches for correlated tests

    • Consider permutation-based methods to establish empirical significance thresholds

    • Report both nominal and adjusted P-values for transparency

  • Advanced analytical approaches:

    • Employ haplotype analysis across the KLK2-KLK3 region

    • Implement sliding window approaches to identify critical regulatory regions

    • Consider gene-gene interaction models between KLK2 and related genes

    • Utilize polygenic risk scores incorporating multiple variants

  • Validation strategies:

    • Split discovery and validation cohorts when sample size permits

    • Perform cross-validation procedures for smaller sample sizes

    • Validate findings in independent populations with different ethnic backgrounds

    • Conduct meta-analysis when multiple datasets are available

Software recommendations include Haploview for LD analysis, PLINK for association testing, and R for advanced statistical modeling and visualization of results .

What are the challenges in developing hK2-targeted therapies and how can they be addressed?

Developing hK2-targeted therapies presents several challenges that require systematic approaches to overcome:

  • Target accessibility challenges:

    • hK2 exists in multiple forms (pro-enzyme, active enzyme, complexed with inhibitors)

    • Intracellular versus extracellular localization affects drug delivery

    • Solutions: Development of highly specific antibodies that recognize distinct epitopes; design of small molecules that can penetrate cellular membranes

  • Specificity and cross-reactivity concerns:

    • High homology between hK2 and PSA (80% amino acid sequence identity)

    • Risk of off-target effects on related kallikreins

    • Solutions: Extensive screening against related proteins; structure-guided design targeting unique surface regions; validation in models expressing multiple kallikreins

  • Pharmacokinetic optimization:

    • Radiolabeled antibodies like [111In]-DOTA-h11B6 require optimization of antibody mass

    • Balancing tumor penetration versus systemic clearance

    • Solutions: Dose-escalation studies; modification of antibody fragments (Fab, scFv); PEGylation strategies

  • Heterogeneous target expression:

    • Variable hK2 expression based on genetic background

    • Changes in expression during disease progression and treatment

    • Solutions: Patient selection based on genetic profiling; development of companion diagnostics; combination therapy approaches

  • Clinical development pathway:

    • Need for appropriate patient selection in early-phase trials

    • Challenges in measuring response to targeted therapies

    • Solutions: Phase 0 trials with imaging endpoints; enrichment of trial populations based on biomarker profiles; novel endpoint designs

The recent first-in-human phase 0 trial with [111In]-DOTA-h11B6 demonstrates a successful approach to addressing these challenges:

  • Confirmed selective accumulation in mCRPC lesions

  • Established safety profile

  • Determined optimal antibody mass

  • Validated hK2 as a viable therapeutic target

Future development should continue this translational pathway from imaging to therapeutic applications, potentially incorporating alpha- or beta-emitting radioisotopes for therapeutic effect.

How can single-cell sequencing technologies advance our understanding of KLK2 expression heterogeneity?

Single-cell sequencing technologies offer unprecedented opportunities to elucidate KLK2 expression heterogeneity across cell types, disease states, and treatment conditions:

  • Cellular resolution of expression patterns:

    • Identification of specific prostatic epithelial cell subtypes expressing KLK2

    • Mapping KLK2 expression to distinct zones of the prostate (peripheral, transition, central)

    • Correlation with androgen receptor signaling at single-cell level

    • Detection of rare KLK2-expressing cells in non-prostatic tissues

  • Disease-related heterogeneity assessment:

    • Comparison of KLK2 expression patterns between normal, hyperplastic, and malignant prostatic cells

    • Characterization of expression dynamics during cancer progression

    • Identification of tumor cell subpopulations with differential KLK2 expression

    • Correlation with other markers of aggressiveness or treatment resistance

  • Methodological approaches:

    • Single-cell RNA sequencing (scRNA-seq) to quantify transcript levels

    • Spatial transcriptomics to preserve tissue architecture information

    • Single-cell ATAC-seq to assess chromatin accessibility at KLK2 locus

    • Single-cell proteomics to detect hK2 protein at cellular resolution

  • Integration with genetic information:

    • Single-cell genotyping to correlate KLK2 variants with expression levels

    • Allele-specific expression analysis in heterozygous individuals

    • Epigenetic profiling to identify regulatory mechanisms

    • Combined genetic and transcriptomic analysis at single-cell resolution

This approach would substantially advance our understanding of the biological roles of KLK2 in both normal physiology and disease states, potentially revealing new therapeutic opportunities and biomarker applications.

What novel technological platforms could enhance the sensitivity and specificity of hK2 detection?

Emerging technological platforms offer promising approaches to enhance hK2 detection sensitivity and specificity:

  • Digital immunoassay platforms:

    • Single molecule array (Simoa) technology enabling femtomolar detection

    • Digital ELISA with isolation of individual immunocomplexes

    • Microfluidic compartmentalization for absolute quantification

    • Application to ultra-low concentration detection in serum and other biofluids

  • Nanomaterial-enhanced detection:

    • Plasmonic biosensors utilizing gold nanoparticles

    • Quantum dot-based fluorescence amplification

    • Carbon nanomaterial-based electrochemical detection

    • Magnetic nanoparticle-facilitated capture and concentration

  • CRISPR-based detection systems:

    • Cas12/13-based molecular diagnostics

    • DNA-encoded antibody libraries

    • Aptamer-based recognition elements

    • Programmable biosensors with nucleic acid amplification

  • Multiplexed detection platforms:

    • Simultaneous measurement of multiple kallikreins and related proteins

    • Microarray and suspension array technologies

    • Mass cytometry approaches for complex sample analysis

    • Integrated proteogenomic platforms combining genetic and protein detection

  • Point-of-care adaptations:

    • Lateral flow immunoassays with enhanced sensitivity

    • Smartphone-integrated optical detection systems

    • Paper-based microfluidic devices

    • Wearable biosensors for continuous monitoring

These technologies would address current limitations in hK2 measurement, potentially enabling earlier detection of disease, more precise monitoring of treatment response, and expanded applications in research and clinical settings.

How might integrating proteomics and genomics advance KLK2-related personalized medicine approaches?

Integrating proteomics and genomics creates powerful opportunities for advancing KLK2-related personalized medicine:

  • Multi-omics characterization of KLK2 biology:

    • Correlation of KLK2 genetic variants with hK2 protein expression, processing, and activity

    • Identification of post-translational modifications affecting hK2 function

    • Mapping of protein-protein interaction networks involving hK2

    • Integration with transcriptomic data to elucidate regulatory mechanisms

  • Comprehensive biomarker development:

    • Combined genetic risk scores based on KLK2/KLK3 variants

    • Protein panels incorporating multiple kallikrein forms and related markers

    • Integration of genetic and protein data into unified risk prediction models

    • Development of algorithms accounting for genetic influence on protein levels

  • Precision medicine applications:

    • Patient stratification based on integrated genetic and protein profiles

    • Tailored screening protocols based on genetic risk and protein biomarkers

    • Selection of optimal therapeutic strategies guided by molecular profiles

    • Monitoring approaches accounting for genetic background

  • Research methodology advances:

    • Mendelian randomization studies using KLK2 variants as instrumental variables

    • Proteogenomic mapping of disease mechanisms

    • Systems biology approaches to model kallikrein pathway dynamics

    • Functional characterization of genetic variants through proteomic analysis

Table 2: Integration of genetic and protein markers for personalized hK2-based approaches

ApplicationGenetic ComponentProtein ComponentIntegrated Approach
Risk StratificationSNP-based risk scoreshK2, PSA, %fPSA levelsCombined algorithm accounting for genetic influence on protein levels
Therapeutic SelectionGenetic variants affecting target expressionTarget protein quantificationSelection of patients with favorable genetic/protein profile
Response MonitoringPharmacogenomic markersDynamic protein changesIntegrated response assessment accounting for genetic background
ScreeningRisk-associated variantsBaseline and longitudinal protein levelsPersonalized screening intervals and thresholds

This integrative approach represents the future of precision medicine in prostate cancer and other KLK2-related conditions, enabling truly personalized approaches to prevention, diagnosis, and treatment.

What are the most promising future directions for KLK2 research?

The field of KLK2 research is poised for significant advances in several promising directions that will likely shape future investigations:

  • Therapeutic target development:

    • Progression from imaging applications using radiolabeled antibodies like [111In]-DOTA-h11B6 to therapeutic radiopharmaceuticals

    • Development of antibody-drug conjugates targeting hK2

    • Exploration of small molecule inhibitors of hK2 enzymatic activity

    • Integration with immunotherapy approaches for enhanced efficacy

  • Improved biomarker implementation:

    • Development of multi-marker panels incorporating hK2, various PSA forms, and novel markers

    • Integration of genetic information to refine biomarker interpretation

    • Application across the disease spectrum from early detection to treatment monitoring

    • Expansion to non-invasive sampling methods (urine, exosomes)

  • Biological role elucidation:

    • Further characterization of hK2's role in prostatic biology beyond semen liquefaction

    • Investigation of potential roles in vascular events and extracellular matrix remodeling

    • Exploration of interactions with other proteases and inhibitors in the tumor microenvironment

    • Study of hK2's role in treatment resistance mechanisms

  • Technological innovations:

    • Application of advanced imaging techniques for visualization of hK2 expression in vivo

    • Development of ultra-sensitive detection methods for early disease detection

    • Implementation of artificial intelligence for integrated data analysis

    • Creation of organoid and other 3D models for functional studies

Product Science Overview

Structure and Function

KLK2 is a secreted enzyme that is highly specific for cleavage after arginine residues . The enzyme is synthesized as an inactive precursor and undergoes proteolytic cleavage to become active. The active form of KLK2 can activate the urokinase-type plasminogen activator, which is involved in the breakdown of blood clots . Additionally, KLK2 is structurally related to KLK3, also known as prostate-specific antigen (PSA), and both are considered biomarkers for prostate cancer .

Industrial Production

Recombinant human KLK2 is produced using various expression systems, including bacterial, yeast, and mammalian cells. One common method involves the use of mouse myeloma cell lines (NS0) to produce KLK2 with a C-terminal His-tag for easy purification . The recombinant protein is typically purified using chromatographic techniques to achieve high purity levels, often greater than 85% .

Applications

KLK2 is used in various research and clinical applications. It is employed as a biomarker for prostate cancer diagnosis and monitoring. Additionally, KLK2 is used in enzymatic assays to study its activity and interactions with other proteins. The enzyme’s ability to cleave specific peptide substrates makes it valuable in biochemical research .

Stability and Storage

Recombinant KLK2 is supplied as a filtered solution in sodium citrate and NaCl. It is shipped with dry ice to maintain stability and should be stored at -70°C to prevent degradation. The protein remains stable for up to six months when stored under these conditions .

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.