LL-37

LL-37
Shipped with Ice Packs
In Stock

Description

Introduction

LL-37, the sole human cathelicidin antimicrobial peptide, is a 37-amino-acid, cationic α-helical peptide derived from the C-terminal cleavage of the human cationic antimicrobial protein hCAP18 . It plays critical roles in innate immunity, exhibiting broad-spectrum antimicrobial activity against bacteria, fungi, and viruses, while also modulating immune responses and inflammation . Despite its therapeutic potential, challenges such as stability, toxicity, and bacterial resistance hinder clinical translation .

Direct Antimicrobial Activity

LL-37 disrupts microbial membranes via:

  1. Membrane Permeabilization: Binds to bacterial lipids (e.g., LPS, LTA) via electrostatic interactions, forming transmembrane pores and inducing lysis .

  2. Intracellular Targeting: Penetrates membranes to inhibit DNA replication, protein synthesis, and biofilm formation .

Key Data:

PathogenMIC (μM)Citation
Staphylococcus aureus0.62
Pseudomonas aeruginosa<10
Escherichia coli<10

Immunomodulatory Functions

LL-37 exhibits dual pro- and anti-inflammatory roles:

  • Pro-inflammatory:

    • Chemotaxis for neutrophils, monocytes, and T cells via formyl-peptide receptor 2 (FPR2) .

    • Synergizes with IL-1β to enhance NF-κB activation and cytokine production (e.g., IL-6, TNF-α) .

  • Anti-inflammatory:

    • Neutralizes LPS and suppresses TLR2/4 signaling, reducing IL-6 and TNF-α in macrophages .

    • Enhances LPS clearance by liver sinusoidal endothelial cells (LSECs) via lysosomal degradation .

Antimicrobial Spectrum and Efficacy

LL-37 demonstrates activity against:

  • Gram-positive bacteria: Staphylococcus epidermidis, methicillin-resistant S. aureus (MRSA) .

  • Gram-negative bacteria: Salmonella typhimurium, Acinetobacter baumannii .

  • Fungi: Candida albicans (via fragment RK-31) .

  • Biofilms: Inhibits S. aureus biofilm formation on titanium alloy surfaces at 0.62 μM .

Limitations: Reduced efficacy in physiological salt concentrations (e.g., 100 mM NaCl) against Proteus mirabilis and Candida albicans .

Immunomodulatory Roles in Disease

  • COVID-19: Modulates neutrophil NETosis and cytokine storms by degrading neutrophil extracellular traps (NETs) and suppressing IL-8 .

  • Sepsis: Reduces IL-6, TNF-α, and macrophage pyroptosis in murine models .

  • Cancer: Paradoxically promotes lung cancer progression via Wnt/β-catenin signaling, correlating with poor prognosis in NSCLC patients .

Clinical Implications and Therapeutic Potential

  • Anti-infective Therapy:

    • Topical formulations for chronic wounds and P. aeruginosa infections in cystic fibrosis .

    • Synergy with conventional antibiotics (e.g., ciprofloxacin) against biofilms .

  • Immunotherapy: Modulates dendritic cell maturation and macrophage polarization .

Challenges and Limitations

  1. Stability: Susceptible to proteolytic degradation in vivo .

  2. Toxicity: Cytotoxic to human cells at high concentrations (>50 μM) .

  3. Cost: High production costs due to peptide synthesis challenges .

  4. Resistance: Bacterial adaptations include membrane modification and efflux pumps .

Future Directions

  • Peptide Engineering: Fragment optimization (e.g., LL-37(17–29)) to enhance stability and reduce toxicity .

  • Delivery Systems: Nanoparticle encapsulation for targeted delivery .

  • Dual-Action Therapies: Combining LL-37 with checkpoint inhibitors in oncology .

Product Specs

Description
LL-37 Synthetic is a single, non-glycosylated polypeptide chain containing 37 amino acids, with a molecular mass of 4493 Daltons and a molecular formula of C205H340N60O53.
Physical Appearance
Sterile Filtered White lyophilized (freeze-dried) powder.
Formulation
The protein was lyophilized with no additives.
Solubility
It is recommended to reconstitute the lyophilized LL-37 in sterile 18MΩ-cm H2O at a concentration not less than 100 µg/ml, which can then be further diluted to other aqueous solutions.
Stability
Lyophilized LL-37, while stable at room temperature for 3 weeks, should be stored desiccated below -18°C. Upon reconstitution, LL-37 should be stored at 4°C for between 2-7 days and below -18°C for future use. For long term storage, it is recommended to add a carrier protein (0.1% HSA or BSA). Please prevent freeze-thaw cycles.
Purity
Greater than 97.0% as determined by analysis by RP-HPLC.
Synonyms

LL37, antibacterial protein LL-37, cathelicidin LL 37, camp.

Amino Acid Sequence

H-Leu-Leu-Gly-Asp-Phe-Phe-Arg-Lys-Ser-Lys-Glu-Lys-Ile-Gly-Lys-Glu-Phe-Lys-Arg-Ile-Val-Gln-Arg-Ile-Lys-Asp-Phe-Leu-Arg-Asn-Leu-Val-Pro-Arg-Thr-Glu-Ser-OH.

Q&A

What is the structural composition of LL-37?

LL-37 is a linear 37-amino acid peptide derived from the C-terminus of the hCAP18 precursor protein through proteolytic cleavage. It is named for its two N-terminal leucine residues. The peptide has a cationic charge of +6 at physiological pH, with a high content of basic and hydrophobic amino acids. While relatively disordered in aqueous solution, LL-37 adopts an amphipathic α-helical structure when in contact with lipid membranes, which is crucial for its antimicrobial activity. The peptide's helical conformation is particularly stable in its core region, sometimes referred to as KR-12, which maintains structural integrity even under varying environmental conditions .

How does LL-37 interact differently with bacterial versus mammalian membranes?

LL-37 demonstrates markedly different interactions with bacterial and mammalian membranes due to their distinct phospholipid compositions. Molecular dynamics simulations reveal that LL-37 rapidly adsorbs onto bacterial-mimicking POPG (negatively charged) bilayers, maintaining its helical conformation and positioning parallel to the membrane surface. This interaction causes significant membrane deformation. In contrast, LL-37 interacts more slowly with mammalian-mimicking POPC (neutral) bilayers, loses much of its helical structure during interaction, and only partially embeds into the membrane without causing significant deformation. This selective interaction explains LL-37's ability to target bacterial cells while sparing host cells and is primarily driven by electrostatic attractions between the positively charged peptide and negatively charged bacterial membranes .

What analytical techniques are most effective for studying LL-37's structural properties?

For comprehensive structural analysis of LL-37, researchers should employ multiple complementary techniques:

TechniqueApplicationKey Insights
Circular Dichroism (CD)Secondary structure analysisConfirms α-helical content in different environments
Nuclear Magnetic Resonance (NMR)Atomic-level structure determinationProvides detailed conformational information
Molecular Dynamics (MD) SimulationsDynamic interactions with membranesReveals mechanism of membrane interaction and deformation
Differential Scanning Calorimetry (DSC)Thermodynamic stabilityMeasures phase transitions upon membrane interaction
Solid-state NMROrientation in membranesDetermines peptide alignment relative to membrane

When studying membrane interactions specifically, combining MD simulations with experimental techniques such as solid-state NMR provides the most complete picture of LL-37's behavior at membrane interfaces .

Where is LL-37 primarily expressed in the human body?

LL-37 is produced by multiple cell types throughout the body, with expression patterns that support its role in host defense. Primary expression sites include phagocytic leukocytes (particularly neutrophils, where it is stored in azurophilic granules), epithelial cells of mucosal surfaces (respiratory, intestinal, and urogenital tracts), and keratinocytes in the skin. Additionally, LL-37 is present in various bodily secretions including mucosal fluids, sweat, and at low concentrations in plasma. Expression is often constitutive in these tissues but can be significantly upregulated during infection, inflammation, or wound healing processes .

How is LL-37 expression regulated during infection and inflammation?

LL-37 expression is regulated through multiple mechanisms that respond to pathogen recognition and inflammatory signals. Research has identified several key regulatory pathways:

  • Pattern recognition receptor (PRR) activation: Bacterial components like LPS can trigger TLR-dependent pathways leading to increased hCAP18/LL-37 expression.

  • Cytokine signaling: Pro-inflammatory cytokines including IL-1β, IL-6, and TNF-α can enhance LL-37 production through activation of transcription factors like NF-κB.

  • Vitamin D pathway: 1,25-dihydroxyvitamin D3 strongly induces LL-37 expression via the vitamin D receptor (VDR), which binds to vitamin D response elements in the CAMP gene promoter.

  • Histone modifications: Epigenetic regulation through histone deacetylase inhibitors can increase LL-37 expression by enhancing chromatin accessibility at the CAMP gene locus.

Methodologically, investigating these regulatory mechanisms requires a combination of techniques including real-time PCR, chromatin immunoprecipitation (ChIP), reporter gene assays, and Western blotting to track both transcriptional and post-transcriptional regulation .

What is the primary mechanism through which LL-37 exerts its antimicrobial activity?

LL-37 employs multiple mechanisms to combat pathogens, with membrane disruption being the primary mode of action against bacteria. The peptide's amphipathic helical structure allows it to interact with bacterial membranes through a sequential process: initial electrostatic attraction between the cationic peptide and anionic bacterial surface, followed by insertion into the membrane and disruption of membrane integrity. Molecular dynamics simulations have visualized how LL-37 causes significant deformation of bacterial-mimicking POPG bilayers while having minimal effect on mammalian-mimicking POPC membranes .

The specific membrane disruption mechanism appears to follow a two-step model: LL-37 first binds to the outer bacterial membrane as monomers and oligomers, then dissociates into monomers that cover the membrane in a carpet-like fashion. Some studies suggest LL-37 subsequently induces membrane leakage through the formation of toroidal pores. Beyond direct membrane disruption, LL-37 also inhibits biofilm formation by downregulating genes essential for biofilm development in pathogens like Pseudomonas aeruginosa .

How does bacterial resistance to LL-37 develop, and what methodologies are used to study resistance mechanisms?

Bacterial resistance to LL-37 develops through several sophisticated mechanisms that researchers can investigate using specific methodological approaches:

Resistance MechanismMethodological ApproachKey Markers
Surface charge modificationZeta potential measurementsChanges in LPS structure or membrane composition
Proteolytic degradationProtease activity assays, Mass spectrometryDetection of specific proteases that cleave LL-37
Efflux pump upregulationReal-time efflux assays, Gene expression analysisExpression of membrane transporters
Biofilm formationCrystal violet staining, Confocal microscopyExopolysaccharide production
Capsule productionNegative staining, Uronic acid assaysPolysaccharide capsule visualization

To comprehensively study resistance, researchers should employ sequential passaging experiments exposing bacteria to sub-inhibitory concentrations of LL-37, followed by whole-genome sequencing to identify genetic adaptations. Complementary approaches include transcriptomics and proteomics to detect changes in gene expression patterns and protein levels that correlate with decreased LL-37 susceptibility .

How does LL-37 bridge innate and adaptive immunity?

LL-37 serves as a critical link between innate and adaptive immune responses through multiple mechanisms. After infection, LL-37 facilitates this immunological bridging by:

The methodological approach to studying these bridging functions requires a combination of chemotaxis assays, calcium flux measurements, co-culture systems with dendritic cells and T cells, and in vivo immunization models with appropriate cytokine profiling .

What is the dual role of LL-37 in inflammation, and how can researchers investigate this dichotomy?

LL-37 exhibits a remarkable dual functionality in inflammatory processes, acting as both pro-inflammatory and anti-inflammatory mediator depending on the microenvironment and cellular context. This dichotomy represents one of the most intriguing aspects of LL-37 biology .

Pro-inflammatory activities:

  • Stimulates phagocytosis and reactive oxygen species production in neutrophils

  • Induces chemokine production in monocytes and epithelial cells

  • Promotes IL-1β processing and release from LPS-primed monocytes via P2X7 receptor activation

  • Acts synergistically with GM-CSF and IL-1β to amplify inflammatory responses

Anti-inflammatory activities:

  • Suppresses LPS-induced production of inflammatory cytokines through direct LPS binding

  • Modulates TLR and NF-κB signaling pathways to selectively downregulate pro-inflammatory gene expression

  • Protects against endotoxemia in mouse models

  • Inhibits responses to lipoteichoic acid

To investigate this dichotomy methodologically, researchers should:

  • Design contextual experiments that vary cell types, inflammatory stimuli, and timing of LL-37 administration

  • Employ phospho-specific antibodies to track activation states of key signaling molecules in inflammatory pathways

  • Use gene expression profiling to identify selectively regulated subsets of inflammatory genes

  • Develop tissue-specific knockout or transgenic mouse models to evaluate context-dependent effects in vivo

  • Conduct time-course experiments to distinguish between early and late effects of LL-37 on inflammatory processes

How does LL-37 interact with other immune mediators in complex inflammatory environments?

LL-37 participates in a sophisticated network of interactions with other immune mediators, creating complex signaling patterns that shape inflammatory responses. Key synergistic and antagonistic relationships include:

  • Synergy with IL-1β: LL-37 acts synergistically with IL-1β to activate AKT, CREB, and NF-κB pathways, resulting in enhanced induction of cytokines and chemokines. This synergy likely relates to the shared MyD88-dependent signaling pathways utilized by IL-1β receptor and most TLRs .

  • Synergy with GM-CSF: LL-37 and GM-CSF cooperatively enhance IL-8 production by monocytes, amplifying neutrophil recruitment signals .

  • Positive feedback with leukotriene B4: A functional interaction between LL-37 and leukotriene B4 in neutrophils creates a positive feedback loop in which each mediator enhances the expression and release of the other, both in vitro and in vivo .

  • Differential modulation of TLR responses: LL-37 inhibits responses to LPS and lipoteichoic acid but enhances some responses to CpG oligonucleotides, demonstrating complex, ligand-specific modulation of TLR signaling .

To methodologically investigate these interactions, researchers should:

  • Design factorial experiments that test LL-37 in combination with other immune mediators at various concentrations

  • Employ phosphoproteomics to map signaling pathway convergence points

  • Use transcriptomics to identify synergistically regulated gene sets

  • Apply systems biology approaches to model interaction networks

  • Develop in vitro tissue models that incorporate multiple cell types to better reflect in vivo complexity

What are the optimal methods for studying LL-37 interactions with microbial and host cell membranes?

Studying LL-37's interactions with membranes requires a multi-technique approach to capture both structural and functional aspects:

TechniqueApplicationMethodological Considerations
Molecular Dynamics SimulationsAtomic-level interaction detailsRequires appropriate force fields (e.g., OPLS-2005); minimum 100ns simulation time; explicit solvent models (TIP3P water model); neutralized systems with counter ions
Fluorescence SpectroscopyMembrane binding kineticsUse of fluorescently labeled LL-37 or membrane-embedded probes; careful control for photobleaching
Surface Plasmon ResonanceBinding affinity measurementsRequires lipid bilayer deposition on sensor chips; multiple regeneration cycles
Solid-State NMROrientation and insertion depth15N or 13C labeled peptides; specialized equipment and expertise
Confocal MicroscopyVisualization of membrane disruptionFluorescently labeled membranes and/or peptides; live cell imaging capabilities
ElectrophysiologyPore formation assessmentPlanar lipid bilayer or patch-clamp techniques; precise control of membrane composition

What are the most reliable in vitro and in vivo models for studying LL-37's biological functions?

Selecting appropriate models for studying LL-37 functions requires careful consideration of their advantages and limitations:

In vitro models:

  • Bacterial killing assays: Use standardized protocols with appropriate controls for medium composition, as divalent cations and serum proteins can significantly affect LL-37 activity

  • Artificial membrane systems: Liposomes with defined lipid compositions (POPG for bacterial, POPC for mammalian membrane mimics) allow controlled study of membrane interactions

  • Cell culture systems: Primary cells are preferred over cell lines for immunomodulatory studies; monocytes, neutrophils, keratinocytes, and epithelial cells are particularly relevant

  • 3D organoid cultures: Provide more physiologically relevant tissue architecture than monolayer cultures for studying epithelial responses to LL-37

  • Ex vivo tissue models: Human skin explants or airway epithelial cultures maintain tissue complexity while allowing experimental manipulation

In vivo models:

  • CRAMP-deficient mice: Mouse cathelicidin (CRAMP) is functionally analogous to human LL-37; Camp-/- mice provide valuable insights into cathelicidin functions in infection and inflammation

  • Humanized models: Mice expressing human LL-37 under tissue-specific promoters better reflect human biology

  • Infection models: Challenge with relevant pathogens followed by local or systemic LL-37 administration can assess therapeutic potential

  • Wound healing models: Excisional or burn wound models evaluate LL-37's role in tissue repair

  • Disease-specific models: Models of inflammatory conditions (e.g., psoriasis, inflammatory bowel disease) help assess context-specific functions

When designing experiments, researchers should consider that no single model fully recapitulates human LL-37 biology, and complementary approaches are typically required for comprehensive understanding .

How can contradictory findings about LL-37's effects on different cell types be reconciled?

Contradictory findings regarding LL-37's effects on different cell types represent a significant challenge in the field. These apparent discrepancies can be methodologically addressed through several approaches:

  • Standardization of experimental conditions: Variations in peptide concentration, purity, storage, and experimental conditions (pH, temperature, medium composition) can dramatically affect results. Researchers should establish standardized protocols and reporting requirements.

  • Comprehensive concentration-response studies: LL-37 often exhibits biphasic effects, with low concentrations (1-5 μg/ml) typically promoting cell survival and immune functions, while high concentrations (>20 μg/ml) may be cytotoxic. Complete concentration-response curves should be generated for each cell type.

  • Accounting for temporal dynamics: LL-37's effects may change over time, with initial activation of certain pathways followed by compensatory inhibition. Time-course experiments are essential for capturing these dynamic effects.

  • Consideration of cellular microenvironment: The presence of serum proteins, extracellular matrix components, and soluble mediators can significantly modulate LL-37's activities. For example, LL-37 binds to DNA, which can alter its immunomodulatory properties.

  • Cell-specific receptor expression profiling: Different cell types express varying levels of LL-37 receptors (FPRL1, P2X7, EGFR). Quantifying receptor expression and using selective antagonists can help explain differential responses .

What is the significance of LL-37 oligomerization in its various biological functions?

LL-37 oligomerization plays a crucial role in modulating its biological activities, with important implications for both antimicrobial and immunomodulatory functions:

Antimicrobial activity:

  • LL-37 has been proposed to function through a two-step model where it initially binds to bacterial membranes as monomers and oligomers, followed by dissociation into monomers that cover the membrane in a carpet-like fashion

  • Oligomerization state affects the mechanism of membrane permeabilization, with different structures (toroidal pores vs. carpet-like disruption) depending on concentration and aggregation state

  • The critical concentration for oligomerization may determine the minimum inhibitory concentration against various pathogens

Immunomodulatory functions:

  • Oligomerization may protect LL-37 from proteolytic degradation in inflammatory environments

  • Different oligomeric states may present distinct binding interfaces for immune receptors, potentially explaining some context-dependent effects

  • Receptor clustering induced by LL-37 oligomers may trigger specific signaling pathways that monomers cannot activate

To methodologically investigate oligomerization, researchers should employ:

  • Analytical ultracentrifugation to determine oligomerization states in solution

  • Size-exclusion chromatography to separate different oligomeric forms

  • Dynamic light scattering to monitor concentration-dependent aggregation

  • Cross-linking studies followed by SDS-PAGE to capture transient oligomeric states

  • Single-molecule fluorescence techniques to visualize oligomerization dynamics in real-time

How can researchers effectively design LL-37 derivatives with enhanced therapeutic potential?

Designing LL-37 derivatives with enhanced therapeutic potential requires systematic structure-activity relationship studies that modify specific peptide properties while maintaining desired functions:

  • Helix stabilization: Introducing helix-stabilizing residues (e.g., α-aminoisobutyric acid) or disulfide bridges to maintain the amphipathic helical structure that is critical for antimicrobial activity, while potentially reducing proteolytic degradation.

  • Charge modulation: Systematic substitution of basic residues to optimize the cationic charge (+6 in native LL-37) for specific applications. Higher positive charge typically enhances antimicrobial activity but may increase cytotoxicity.

  • Hydrophobicity tuning: Careful adjustment of hydrophobic residues to balance antimicrobial potency against unwanted aggregation and cytotoxicity. Substituting natural amino acids with non-natural hydrophobic residues can provide finer control.

  • Truncation studies: Identifying the minimum active sequence (such as the KR-12 fragment) that retains specific functions while reducing synthesis costs and potentially minimizing side effects.

  • Domain-focused modifications: Selectively modifying regions responsible for specific functions, based on the understanding that different segments of LL-37 contribute differently to its multiple activities.

Methodologically, researchers should employ:

  • Alanine scanning to identify critical residues

  • D-amino acid substitutions to enhance proteolytic stability

  • Cyclic derivatives to constrain conformation

  • High-throughput screening of peptide libraries

  • In silico modeling to predict structure-activity relationships before synthesis

  • Iterative optimization based on biological testing

What are the most promising approaches for studying LL-37's role in cancer progression?

LL-37 exhibits complex and sometimes contradictory roles in cancer biology, acting as both a tumor promoter and tumor suppressor depending on cancer type and microenvironment. To effectively study these roles, researchers should employ multifaceted approaches:

Methodological approaches:

  • Patient sample analysis:

    • Quantify LL-37 expression in tumor tissue versus matched normal tissue

    • Correlate expression with clinical outcomes and molecular subtypes

    • Use immunohistochemistry to determine cellular sources of LL-37 within the tumor microenvironment

  • Mechanistic in vitro studies:

    • Assess direct effects on cancer cell proliferation, migration, and invasion

    • Investigate effects on angiogenesis using endothelial cell tube formation assays

    • Evaluate impact on immune cell recruitment and polarization

    • Examine receptor-mediated signaling, particularly through FPRL1, P2X7, and EGFR pathways

  • In vivo models:

    • Develop conditional knockout or overexpression of cathelicidin in specific cell types

    • Use orthotopic tumor models to maintain appropriate microenvironment

    • Employ patient-derived xenografts to better reflect human tumor heterogeneity

    • Implement syngeneic models to properly assess immune component interactions

  • Therapeutic exploration:

    • Test LL-37 or derivatives as direct anti-cancer agents

    • Explore combination approaches with immunotherapies

    • Investigate LL-37's potential as a cancer vaccine adjuvant

    • Develop strategies to block LL-37 in contexts where it promotes tumor growth

This comprehensive approach can help resolve the seemingly contradictory findings regarding LL-37's role in cancer and potentially identify new therapeutic strategies based on context-specific modulation of LL-37 activity .

What are the emerging technologies that will advance our understanding of LL-37 biology?

Several cutting-edge technologies are poised to transform our understanding of LL-37 biology and accelerate therapeutic applications:

  • Single-cell technologies: Single-cell RNA sequencing and mass cytometry will reveal cell-specific responses to LL-37 within heterogeneous populations, helping to resolve contradictory findings that may result from studying bulk cell populations.

  • CRISPR-Cas9 genome editing: Precise modification of LL-37-related genes and regulatory elements will enable detailed mechanistic studies of expression regulation and receptor interactions in relevant human cell types.

  • Cryo-electron microscopy: This technique can provide atomic-resolution structures of LL-37 oligomers and membrane-associated complexes, offering unprecedented insights into the molecular mechanisms of membrane disruption.

  • Advanced computational modeling: Machine learning approaches combined with molecular dynamics simulations will allow prediction of structure-activity relationships and enable rational design of improved LL-37 derivatives.

  • Biomimetic membrane systems: Nanodiscs, lipid cubic phases, and microfluidic organ-on-chip platforms will provide more physiologically relevant contexts for studying LL-37-membrane interactions.

  • Advanced imaging techniques: Super-resolution microscopy and correlative light-electron microscopy will visualize LL-37's interactions with cellular structures at nanometer resolution in real-time.

  • Systems biology approaches: Multi-omics integration (genomics, transcriptomics, proteomics, metabolomics) will map the complex networks through which LL-37 influences cellular physiology and disease processes .

How can researchers address the challenge of LL-37's context-dependent functions in developing therapeutic applications?

Addressing the context-dependency of LL-37's functions is crucial for successful therapeutic development. Researchers should implement the following methodological strategies:

  • Systematic microenvironment mapping: Characterize LL-37's activities across various physiologically relevant microenvironments, varying parameters such as pH, ionic strength, presence of serum proteins, and extracellular matrix components.

  • Receptor-specific derivatives: Design LL-37 variants that selectively engage specific receptors (FPRL1, P2X7, EGFR) to elicit desired functions while avoiding unwanted effects mediated through other pathways.

  • Tissue-targeted delivery systems: Develop delivery platforms (nanoparticles, hydrogels, tissue-specific antibody conjugates) that direct LL-37 or its derivatives to specific anatomical locations where beneficial effects predominate.

  • Temporal control strategies: Implement controlled release systems to manage exposure kinetics, potentially avoiding adverse effects associated with prolonged exposure.

  • Combination therapy approaches: Pair LL-37 with complementary agents that enhance desired activities or counteract unwanted effects in specific contexts.

  • Biomarker-guided personalization: Identify biomarkers that predict favorable responses to LL-37-based interventions in individual patients or disease states.

  • Feedback-responsive systems: Design smart delivery systems that respond to environmental cues (e.g., bacterial products, inflammatory mediators) to release LL-37 only when and where beneficial effects are likely to predominate.

By systematically addressing context-dependency rather than viewing it as an obstacle, researchers can harness the multifunctionality of LL-37 for precision medicine applications tailored to specific disease contexts .

Product Science Overview

Origin and Structure

LL-37 is the only human cathelicidin-derived antimicrobial peptide. It is a 37-amino acid cationic peptide generated by the extracellular cleavage of the C-terminal end of the 18-kDa precursor protein, hCAP18, by serine proteases such as kallikreins in keratinocytes and proteinase 3 in neutrophils . The peptide is named “LL-37” based on its sequence, which starts with two leucine residues.

Classification

LL-37 belongs to the family of antimicrobial peptides (AMPs), which are crucial components of the innate immune system. AMPs are evolutionarily conserved molecules that provide a first line of defense against a wide range of pathogens, including bacteria, viruses, fungi, and parasites .

Biological Properties

LL-37 exhibits broad-spectrum antimicrobial activity, making it effective against various pathogens. Beyond its antimicrobial properties, LL-37 has been shown to modulate immune responses, influence cell proliferation, and promote wound healing . It can neutralize endotoxins, bind to lipopolysaccharides, and inhibit biofilm formation, further enhancing its protective role in the immune system .

Expression Patterns and Tissue Distribution

LL-37 is expressed in various tissues and cell types, including epithelial cells, neutrophils, and macrophages. It is found in the skin, respiratory tract, gastrointestinal tract, and reproductive organs . The expression of LL-37 can be induced by inflammatory stimuli, infections, and tissue injury, highlighting its role in immune defense and tissue repair .

Biological Functions and Modes of Action

LL-37’s primary function is to act as an antimicrobial agent, directly killing pathogens through membrane disruption. However, it also plays a significant role in modulating the immune system. LL-37 can attract immune cells to sites of infection, promote the release of cytokines and chemokines, and enhance the phagocytic activity of macrophages . Additionally, LL-37 has been implicated in the regulation of adaptive immune responses and the modulation of inflammatory pathways .

Regulatory Mechanisms

The expression and activity of LL-37 are tightly regulated to maintain immune homeostasis and prevent excessive inflammation. Various factors, including microbial components, cytokines, and growth factors, can influence the production of LL-37 . Dysregulation of LL-37 expression has been associated with several inflammatory and autoimmune diseases, such as psoriasis, systemic lupus erythematosus, and rheumatoid arthritis .

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.