Kallikrein-1 Human Recombinant produced in E.coli is a single, non-glycosylated polypeptide chain containing 259 amino acids (25-262) and having a molecular mass of 28.7kDa.
KLK1 is fused to a 21 amino acid His-tag at N-terminus & purified by proprietary chromatographic techniques.
Human Kallikrein 1 (KLK1), also known as tissue kallikrein, is a serine protease belonging to the S1 serine protease superfamily. It is a member of the human tissue kallikrein family. The primary physiological function of KLK1 is the enzymatic cleavage of low molecular weight kininogen (LMWK) to release vasoactive kinin peptides, specifically lysyl-bradykinin (kallidin) or bradykinin . These kinin peptides regulate numerous physiological processes including blood pressure reduction, vasodilation, smooth muscle relaxation and contraction, pain induction, and inflammation. Additionally, KLK1 plays roles in angiogenesis and tumorigenesis processes .
Human KLK1 precursor contains three distinct regions:
A signal peptide (residues 1 to 18)
A short pro-peptide (residues 19 to 24)
A mature chain (residues 25 to 262)
In recombinant protein formulations, KLK1 is often expressed with tags such as C-terminal 6His or 10His tags for purification purposes, and may include propeptides from other kallikreins (e.g., KLK5 propeptide) to stabilize the protein . The mature protein has a molecular mass of approximately 28.2 kDa, though the apparent molecular mass after post-translational modifications is around 38 kDa .
KLK1 is one of 15 members of the human kallikrein gene family, all located in a gene cluster on chromosome 19q13.4. What distinguishes KLK1 from other family members is its true kininogenase activity. While other kallikreins like KLK2 and KLK3 (prostate-specific antigen or PSA) share structural similarities and chromosomal location with KLK1, they possess either very low (KLK2) or no (KLK3) kininogenase activity . KLK1 is functionally conserved across species in its capacity to cleave low molecular weight kininogen, though its evolutionary relationship with rodent kallikreins shows divergence after the separation of primate and rodent lineages .
Recombinant human KLK1 protein requires careful handling to maintain activity. Based on commercial specifications:
Storage temperature: Store at -20°C or below for long-term stability
Stability: Stable for approximately 6 months when properly stored
Formulation: Typically supplied in buffer solutions containing TrisHCl, NaCl, and CaCl2 (e.g., 20mM TrisHCl, 150mM NaCl, 2mM CaCl2, pH 8.0)
Shipping: Usually shipped frozen with blue ice/gel packs
Handling: Minimize freeze-thaw cycles to preserve enzymatic activity
Filtration: Solutions are typically 0.2 μm filtered for sterility
To verify KLK1 enzymatic activity, fluorogenic peptide substrate assays are commonly employed. A standardized protocol includes:
Activation step (if using KLK1 with propeptide):
Incubate KLK1 with thermolysin (1:10 ratio thermolysin:KLK1) in assay buffer
Incubate at 37°C for 1 hour
Stop reaction with 20 mM 1,10 Phenanthroline (final concentration 10 mM)
Activity assay:
Dilute activated KLK1 to 0.4 ng/μL in assay buffer
Prepare fluorogenic substrate at 200 μM in assay buffer
Combine 50 μL of diluted KLK1 with 50 μL substrate
Include substrate blank (50 μL buffer + 50 μL substrate)
Monitor fluorescence at excitation/emission wavelengths of 380 nm/460 nm in kinetic mode
Several methodological approaches are employed to study KLK1 inhibition:
Human monoclonal antibody inhibition assays:
Specific antibodies have been developed that selectively inhibit KLK1
These can be used in enzyme assays to determine inhibition constants
Surface plasmon resonance (SPR) techniques quantify binding kinetics and affinity
Serine protease inhibitor studies:
Natural inhibitors like SPINK6 (serine protease inhibitor of Kazal-type 6) can be tested
Inhibition constants (Ki) determination using concentration-dependent inhibition curves
Pre-incubation of KLK1 with potential inhibitors before substrate addition
Small molecule inhibitor screening:
KLK1 has emerged as a promising therapeutic target, particularly for cardiovascular, cerebrovascular, and renal disorders. Current research approaches include:
Gene therapy approaches:
Delivery of KLK1 gene to target tissues using viral vectors
Stem cell-based delivery systems incorporating KLK1 genetic modifications
Evaluation of tissue-specific expression systems for localized effects
Therapeutic mechanisms under investigation:
Enhancement of nitric oxide (NO) production via bradykinin receptor activation
Promotion of angiogenesis in ischemic tissues
Anti-inflammatory effects in vascular and renal tissues
Modulation of blood pressure regulation pathways
Preclinical and clinical evidence:
Several significant challenges exist in developing KLK1-based therapeutics:
Delivery challenges:
Protein stability issues affecting half-life in circulation
Targeted delivery to affected tissues to minimize systemic effects
Optimization of gene therapy vectors for safe, long-term expression
Functional concerns:
Potential immunogenicity of recombinant or modified KLK1
Balancing therapeutic effects with potential pro-inflammatory actions
Risk of hypotension from excessive vasodilation
Regulatory and development challenges:
Need for tissue-specific or controllable expression systems
Development of appropriate biomarkers to monitor efficacy
Integration with existing treatment modalities
Research methodology limitations:
Post-translational modifications (PTMs) significantly impact KLK1 functionality, and analyzing these modifications requires sophisticated approaches:
When investigating KLK1's involvement in angiogenesis and tumor biology, researchers should consider:
Model system selection:
Cell models: Endothelial cells (HUVECs, HMVECs) for angiogenesis studies
3D models: Matrigel tube formation assays, spheroid models
In vivo models: Appropriate tumor xenograft or genetic models
Patient-derived samples: Primary cultures or tissue sections
Experimental variables:
KLK1 concentration ranges physiologically relevant to target tissues
Time-dependent effects (acute vs. chronic exposure)
Interaction with extracellular matrix components
Cross-talk with other proteases and inhibitors in the microenvironment
Readout selection:
Angiogenesis markers: VEGF expression, CD31 staining, tube formation
Tumor progression indicators: Proliferation, invasion, metastasis
Signaling pathway activation: Bradykinin receptor signaling, NO production
Protease activation cascades: Downstream protease activation
Control considerations:
Resolving contradictory data about KLK1 effects requires systematic approaches:
Source of contradictions:
Species differences in KLK1 function and regulation
Variations in experimental conditions (pH, ionic strength, substrate availability)
Different cell types or tissues exhibiting context-dependent responses
Methodological differences in activity measurement or protein preparation
Resolution strategies:
Head-to-head comparisons using standardized protocols
Comprehensive characterization of KLK1 preparations (activity, purity, modifications)
Inclusion of multiple readouts to capture diverse biological effects
Detailed reporting of experimental conditions to facilitate replication
Mechanistic investigations:
Identification of cell-specific receptor and signaling pathway variations
Examination of proteolytic processing differences between systems
Analysis of local inhibitor concentrations affecting net activity
Investigation of compensatory mechanisms in different models
Integration approaches:
To effectively study interactions between KLK1 and other kallikrein family members:
Enzymatic activation cascades:
Sequential incubation experiments to detect cross-activation
Activity-based protein profiling with selective probes
Time-course analysis of activation patterns
Mass spectrometry identification of cleavage products
Protein-protein interaction studies:
Co-immunoprecipitation experiments with tagged kallikreins
Surface plasmon resonance (SPR) for binding kinetics
Proximity ligation assay (PLA) for in situ interaction detection
Yeast two-hybrid or mammalian two-hybrid screening
Co-expression analysis:
qPCR for co-expression patterns in tissues
Single-cell RNA sequencing to identify co-expressing cells
Immunohistochemistry co-localization studies
Conditional expression systems to manipulate expression ratios
Functional consequence investigation:
Researchers working with recombinant KLK1 often encounter several technical challenges:
Activity loss during storage/handling:
Solution: Aliquot protein upon receipt to minimize freeze-thaw cycles
Add stabilizing agents like glycerol (10-20%) for storage
Include calcium in buffers (e.g., 2mM CaCl₂) to maintain structural integrity
Monitor activity regularly with standardized assays
Variable activation efficiency:
Solution: Optimize thermolysin:KLK1 ratios empirically for each lot
Carefully control incubation time and temperature
Use activity assays to confirm complete activation
Consider alternative activation methods if inconsistency persists
Background/non-specific activity:
Solution: Include appropriate controls (substrate-only, heat-inactivated enzyme)
Use highly specific substrates for KLK1
Pre-clear solutions by high-speed centrifugation
Test for cross-reactivity with other proteases in your system
Buffer compatibility issues:
Distinguishing direct KLK1 effects from bradykinin-mediated effects requires:
Pharmacological approaches:
Use selective bradykinin B1 and B2 receptor antagonists (e.g., HOE-140 for B2)
Compare native KLK1 effects with direct bradykinin administration
Employ selective inhibitors of downstream bradykinin signaling (NOS inhibitors, cyclooxygenase inhibitors)
Use engineered KLK1 mutants with altered kininogenase activity but preserved protein structure
Genetic approaches:
Utilize bradykinin receptor knockout models or cells
Employ siRNA/shRNA knockdown of bradykinin receptors
Use CRISPR/Cas9 to create receptor-deficient cell lines
Compare phenotypes between KLK1 knockout and bradykinin receptor knockout models
Biochemical strategies:
For accurate assessment of KLK1 activity in complex biological samples:
Selective substrate approaches:
Fluorogenic substrates with high selectivity for KLK1
Internally quenched FRET peptide substrates for improved signal-to-noise
Comparison of cleavage patterns with multiple substrates to confirm specificity
Spike-in of known amounts of active KLK1 as internal calibrators
Immunocapture-activity assays:
Capture KLK1 using specific antibodies immobilized on plates or beads
Wash to remove interfering substances
Add specific substrate to measure activity of captured enzyme
Include controls with KLK1-specific inhibitors
Mass spectrometry approaches:
Multiple reaction monitoring (MRM) of specific KLK1 cleavage products
MALDI-TOF analysis of substrate cleavage patterns
Activity-based protein profiling with KLK1-selective probes
Parallel reaction monitoring for simultaneous quantification of multiple kallikreins
Data analysis considerations:
KLK1 is emerging as a target in stem cell-based therapeutic approaches:
Genetic modification strategies:
Transduction of mesenchymal stem cells with KLK1-expressing vectors
Conditional expression systems allowing regulated KLK1 production
CRISPR/Cas9 modification of stem cells for stable KLK1 expression
Development of inducible expression systems for temporal control
Therapeutic applications under investigation:
Cardiovascular disorders: Improving tissue perfusion and reducing damage
Cerebrovascular conditions: Neuroprotection in stroke models
Renal disorders: Protection against fibrosis and promoting regeneration
Wound healing: Enhancing angiogenesis and tissue repair
Delivery approaches:
Direct implantation of modified stem cells into affected tissues
Systemic administration with homing to sites of injury
Encapsulation technologies for sustained release
Combined approaches with scaffolds for tissue engineering
Monitoring methodologies:
KLK1's role in inflammatory diseases presents both challenges and opportunities:
Dual roles in inflammation:
Pro-inflammatory effects: Bradykinin production leading to vasodilation, increased vascular permeability, and pain
Anti-inflammatory potential: Activation of regulatory pathways, resolution of inflammation in certain contexts
Tissue-specific effects varying by local microenvironment
Temporal dynamics with different roles in acute versus chronic inflammation
Disease-specific findings:
Airway diseases: Potential involvement in asthma and COPD pathophysiology
Inflammatory bowel diseases: Altered expression in affected tissues
Neuroinflammatory conditions: Modulation of microglial responses
Dermatological disorders: Role in inflammatory skin conditions
Therapeutic strategies under investigation:
Selective inhibition in pro-inflammatory contexts
Augmentation in settings where anti-inflammatory effects predominate
Targeted delivery to specific tissue microenvironments
Temporal modulation based on disease stage
Biomarker development:
Advances in understanding KLK1 structure-function relationships are driving new drug development approaches:
Structural insights guiding inhibitor design:
Crystal structures revealing unique features of KLK1's active site
Identification of specific binding pockets for selective targeting
Understanding conformational changes upon substrate binding
Comparative analysis with other kallikreins to enhance selectivity
Rational design approaches:
Structure-based virtual screening for novel inhibitor scaffolds
Fragment-based drug discovery targeting specific KLK1 domains
Peptide-mimetic approaches based on natural substrates and inhibitors
Allosteric modulator development targeting regulatory sites
Antibody-based strategies:
Epitope mapping of function-blocking antibodies
Single-domain antibody (nanobody) development for enhanced tissue penetration
Bi-specific antibodies targeting KLK1 and disease-specific markers
Antibody-drug conjugates for targeted delivery
Alternative modulation strategies:
Human KLK1 shows important differences from orthologs in other species:
Structural and functional comparisons:
Species | Key Differences | Implications for Research |
---|---|---|
Mouse | Multiple gene copies vs. single human gene | Potentially confounding compensation mechanisms |
Rat | Different substrate specificity | May affect translational relevance of findings |
Non-human primates | Higher homology to human KLK1 | Better translational models for inhibitor testing |
Other mammals | Variable glycosylation patterns | Altered pharmacokinetics of recombinant proteins |
Evolutionary considerations:
Human and rodent kallikrein families diverged after separation of primate and rodent lineages
Expanded gene family in rodents (up to 20 genes) compared to humans (15 genes)
Human KLK2 and KLK3 have no known orthologs in rodents
Conservation of key catalytic residues across species despite sequence variations
Research strategy adaptations:
Functional relationships between KLK1 and other proteases vary considerably across systems:
Species-specific interaction networks:
Different complement of potential activators and substrates
Variable expression patterns of interacting proteins
Altered regulatory mechanisms for activation cascades
Different inhibitor profiles affecting net proteolytic activity
Experimental system considerations:
Cell culture systems may lack key components of activation cascades
In vitro biochemical assays fail to capture cellular compartmentalization
Animal models reflect species-specific networks rather than human biology
Patient samples capture disease-specific alterations in protease networks
Methodological approaches for network mapping:
Degradomics approaches to identify comprehensive substrate profiles
Interactome analysis using affinity purification-mass spectrometry
Functional screening using siRNA/CRISPR libraries
Computational modeling of protease cascades with system-specific parameters
Translational implications:
Genomic and transcriptomic studies have revealed important aspects of KLK1:
Evolutionary insights:
The human KLK locus shows high synteny with corresponding regions in other mammals
Human kallikrein genes are transcribed in the same direction and share 70-90% sequence homology
All kallikrein genes maintain five coding exons with conserved exon-intron splice sites
TATA box variants and polyadenylation signals are conserved across species
Expression regulation mechanisms:
Tissue-specific promoter elements controlling expression patterns
Hormone-responsive elements in promoter regions (androgens, estrogens)
Epigenetic regulation through DNA methylation and histone modifications
Alternative splicing patterns affecting function and regulation
Comparative expression profiling:
Species-specific expression patterns in different tissues
Developmental regulation differences between species
Disease-associated expression changes with variable conservation
Sex-specific expression patterns with implications for disease susceptibility
Regulatory network evolution:
Promising future directions for KLK1-related applications include:
Advanced diagnostic approaches:
Development of highly specific KLK1 activity assays for clinical samples
Multi-marker panels combining KLK1 with related biomarkers
Point-of-care testing for rapid KLK1 activity assessment
Imaging agents targeting KLK1 for non-invasive visualization
Therapeutic strategies:
Gene therapy approaches using tissue-specific promoters
mRNA-based therapeutics for transient KLK1 modulation
Development of bispecific antibodies with enhanced targeting
Small molecule allosteric modulators with improved selectivity
Precision medicine applications:
Genetic profiling to identify patients likely to respond to KLK1-targeted therapies
Companion diagnostics measuring KLK1 activity to guide treatment
Combination approaches targeting multiple kallikrein family members
Patient stratification based on KLK1 pathway activation status
Novel delivery technologies:
To advance our understanding of KLK1 functions, methodological improvements are needed:
Advanced imaging technologies:
Live-cell imaging of KLK1 activity using selective reporters
Super-resolution microscopy to localize KLK1 in cellular compartments
Intravital microscopy for in vivo visualization of KLK1 activity
Correlative light and electron microscopy for ultrastructural context
Systems biology approaches:
Multi-omics integration (proteomics, transcriptomics, metabolomics)
Network analysis of KLK1-dependent processes
Agent-based modeling of KLK1 functions in tissue microenvironments
Machine learning algorithms to identify patterns in complex datasets
Advanced genetic models:
Conditional and inducible KLK1 knockout/knockin systems
Tissue-specific expression models with controllable levels
Humanized animal models expressing human KLK1
Patient-derived organoids for disease-specific studies
Improved biochemical tools:
Emerging computational and structural approaches offer significant potential:
Advanced computational techniques:
Molecular dynamics simulations of KLK1-substrate/inhibitor interactions
Machine learning for prediction of KLK1 substrates and inhibitors
Systems pharmacology modeling of KLK1 in disease networks
Quantum mechanics/molecular mechanics for transition state analysis
Structural biology advances:
Cryo-electron microscopy of KLK1 complexes
Time-resolved crystallography to capture enzymatic intermediates
NMR studies of KLK1 dynamics and conformational changes
Hydrogen-deuterium exchange mass spectrometry for protein dynamics
Integration of structural and functional data:
Structure-based pharmacophore modeling for inhibitor design
Correlation of structural features with functional outcomes
In silico mutagenesis to predict functional consequences
Virtual screening against diverse KLK1 conformational states
Novel computational drug design strategies:
The Human Recombinant Kallikrein-1 (His Tag) is produced using an expression system in HEK293 cells . This recombinant protein includes a C-terminal His-tag, which facilitates its purification and detection. The amino acid sequence of this recombinant protein corresponds to the human KLK1 precursor, spanning from Met1 to Ser262 .
Kallikrein-1 is involved in the proteolysis of bradykininogen to bradykinin, a peptide that plays a crucial role in various physiological processes, including blood pressure regulation, inflammation, and pain . The recombinant form of this protein is often used in research to study these processes and to explore its potential as a biomarker for diseases .
Growing evidence suggests that many kallikreins, including KLK1, are implicated in carcinogenesis and may serve as novel biomarkers for cancer and other diseases . The ability to produce recombinant forms of these proteins allows researchers to study their functions in detail and to develop potential therapeutic applications.
In summary, Kallikrein-1 (Human Recombinant, His Tag) is a valuable tool in biomedical research, providing insights into the physiological roles of kallikreins and their potential implications in disease.