The triple mutation disrupts critical residues in the AB loop (amino acids 39–42), a region essential for ObR activation via site III binding. PEGylation at a single site minimizes steric hindrance while maximizing stability .
Leptin tA Human, PEG binds to ObR but blocks downstream signaling:
Binding Affinity: Retains high affinity for ObR due to preserved binding sites I and II (helices A/B and C) .
Antagonism: Mutations in the AB loop (site III) prevent receptor dimerization and JAK2/STAT3 pathway activation .
Blood-Brain Barrier (BBB) Interaction: PEGylation enhances BBB penetration, inducing central leptin deficiency .
Weight Loss Trials: In a 12-week study (20 mg/week PEG-OB in obese men), no significant weight loss vs. placebo, but triglyceride reduction trends noted .
Antagonist Efficacy: Pegylated leptin antagonists (e.g., PEG-MLA) show 13.4-fold increased half-life and potent orexigenic effects in mice .
Parameter | Pegylated Leptin tA Human, PEG | Non-Pegylated Leptin Antagonist |
---|---|---|
Half-Life | 24–48 hours (vs. 30 minutes native) | 8–30 minutes (rapid renal clearance) |
Biological Activity | 3–19.5% of non-PEGylated (due to steric hindrance) | Full activity |
BBB Penetration | Enhanced (central leptin deficiency) | Limited (peripheral action only) |
Cachexia: Induced leptin deficiency mimics wasting syndromes, enabling mechanistic studies .
Metabolic Disorders: Potential for modulating insulin sensitivity and lipid metabolism .
Obesity: Limited efficacy in hyperleptinemic states due to leptin resistance .
Selectivity: Balancing BBB penetration and peripheral effects remains a challenge.
Dosing Optimization: Weekly vs. daily regimens require further testing .
Property | Value | Methodology |
---|---|---|
Molecular Weight | 35.6 kDa (calculated) | SDS-PAGE, gel filtration |
PEG Size | 20 kDa | Proprietary conjugation techniques |
Mutations | L39A, D40A, F41A (AB loop) | Site-directed mutagenesis |
Source | E. coli recombinant | Lyophilized powder formulation |
Parameter | Pegylated Leptin tA Human, PEG | Non-Pegylated Leptin Antagonist |
---|---|---|
Half-Life | 24–48 hours | 8–30 minutes |
Clearance Route | Hepatic/renal (reduced) | Renal dominant |
Stability | High (protease resistance) | Moderate |
Human leptin contains numerous hydrophobic residues (Trp100, Phe92, Leu142, Trp138, and Phe41) responsible for its self-association and aggregation. Two cysteine residues (C96 in the CD loop and C146 at the C-terminal end) form a disulfide bridge crucial for structural stability and biological activity .
Pegylation, the process of attaching polyethylene glycol (PEG) molecules to proteins, significantly enhances leptin's bioavailability and half-life. This modification reduces renal clearance, protects against proteolytic degradation, and decreases immunogenicity while maintaining therapeutic efficacy. For leptin antagonists specifically, pegylation has been shown to substantially enhance their in vivo potency by extending their circulation time, allowing for less frequent administration while maintaining biological activity .
Leptin binds to LEP-R (leptin receptor), triggering a conformational change that activates associated JAK2 (Janus kinase 2). JAK2 autophosphorylates and simultaneously phosphorylates tyrosine residues on the functional LEP-R's intracellular domain, allowing STAT proteins to bind and subsequently translocate to the nucleus where they function as transcription factors .
Multiple signaling cascades are activated by leptin binding, including:
JAK2/STAT3 pathway - primarily responsible for regulating gene expression changes
Phosphoinositol-3 kinase (PI3K) pathway - signals more rapidly through phosphorylation of cytoplasmic proteins
Mitogen-activated protein kinases/extracellular signal-regulated kinase (MAPK/ERK) pathway
The activation of these pathways collectively contributes to leptin's anorexigenic effects (suppressing appetite, stimulating weight loss, and increasing thermogenesis). Notably, the PI3K pathway plays a particularly important role in leptin's acute effects, such as regulating food intake and arterial hypertension .
Leptin acts on the hypothalamus by inhibiting orexigenic (appetite-stimulating) neural pathways while activating anorexigenic (appetite-suppressing) pathways . This regulation occurs through a simple but elegant model:
Leptin affects the transcription of proopiomelanocortin (POMC), whose product α-MSH is released into synapses to activate neurons via melanocortin receptor (MCR) binding, leading to appetite suppression
Simultaneously, leptin inhibits NPY/AgRP synthesis in neurons, reducing the agonistic effect of AgRP on MCR
Research has shown that leptin treatment normalizes synaptic density on NPY/AgRP and POMC neurons within 6 hours, preceding its effects on food intake. This indicates that leptin's initial action involves modulating neuronal plasticity rather than direct metabolic effects .
Leptin antagonists are primarily designed through strategic amino acid substitutions in regions critical for receptor activation. The sequence 39-42 in the loop AB of leptin is fundamental for leptin receptor (ObR) activation and therefore represents a prime target for modification .
Several specific mutations have proven effective:
Triple mutein L39A/D40A/F41A (LDF or Lan-1) and quadruple mutein L39A/D40A/F41A/I42A (LDFI or Lan-2): Created by replacing amino acids in sequence 39-42 with alanine residues
D23L mutation: Significantly enhances human leptin's affinity toward the leptin receptor
S120A/T121A leptin mutant: Replaces Ser120 and Thr121 on the N-terminus of helix D with alanine, enabling selective binding to the CRH2 domain of ObR without activation
R128Q leptin antagonist: Increases body weight in mice by indirectly affecting binding site III through modification of the AB and CD loops' orientation
Pegylation substantially enhances the bioavailability of these antagonists in vivo, creating more potent and longer-acting therapeutic candidates .
Pegylated leptin peptide receptor antagonist 2 (PEG-LPrA2) has demonstrated significant anti-tumor activity in breast cancer models. Treatment with PEG-LPrA2 reduced the expression of vascular endothelial growth factor (VEGF), vascular endothelial growth factor receptor type 2 (VEGFR2), and inhibited growth of 4T1-breast cancer in syngeneic mice .
PEG-LPrA2 treatment also effectively reduced human leptin levels in tumors from mice hosting MCF-7 breast cancer xenografts . This research suggests leptin signaling inhibition may represent a novel therapeutic approach for breast cancer.
Current investigations are exploring differential impacts of leptin signaling inhibition on the expression of pro-angiogenic and pro-proliferative molecules between estrogen receptor-positive (ER+) and estrogen receptor-negative (ER-) breast cancer xenografts . This research area is particularly promising given the observed overexpression of leptin receptors (ObR) in many human cancer types, which correlates strongly with leptin presence .
Integration studies should focus on potential synergistic effects between leptin antagonists and other hormone receptor modulators. The search results highlight successful examples with incretin/glucagon system triagonists where combination therapies yielded enhanced metabolic outcomes .
For example, dual-incretin agonism (combining GLP-1 and GIP analogs) improved body weight and hyperglycemia in metabolically compromised mice more effectively than single agonists . When a stabilized GIP analog (ZP4165) was coadministered with liraglutide (a GLP-1 receptor agonist), researchers observed synergistic lowering of HbA1c and body weight, as well as reduction in total cholesterol that was not seen with either treatment alone .
For leptin antagonist integration studies, researchers should:
Establish clear baseline effects of each modulator independently
Use factorial experimental designs to systematically test combinations
Identify potential mechanisms of interaction through signaling pathway analysis
Evaluate effects across multiple metabolic parameters (weight, glucose, lipids)
Consider potential tissue-specific effects where receptors may be co-expressed
Reliable assessment of leptin antagonist binding and efficacy requires a multi-faceted approach:
In vitro binding assays:
Surface plasmon resonance (SPR) to measure binding kinetics to LEP-R
Competition binding assays with labeled leptin
BRET/FRET techniques to assess receptor conformational changes
Signaling pathway validation:
Western blot analysis for JAK2/STAT3 phosphorylation inhibition
Reporter gene assays using STAT3-responsive elements
Phosphoproteomic analysis of downstream effectors
Functional validation:
Food intake and body weight monitoring in animal models
Hypothalamic slice culture electrophysiology to measure effects on POMC and NPY/AgRP neurons
Metabolic phenotyping (indirect calorimetry, glucose tolerance)
The most reliable approach combines binding assays with both signaling and functional readouts. For instance, the S120A/T121A leptin mutant was validated through binding assays showing selective interaction with the CRH2 domain of ObR without activation, followed by in vivo studies demonstrating increased body weight in mice .
While specific pegylation protocols for leptin antagonists aren't explicitly detailed in the search results, general principles and considerations can be derived from successful examples like PEG-LPrA2 :
Site selection considerations:
Target non-essential residues away from receptor binding interfaces
Consider lysine residues for NHS-ester chemistry or cysteine residues for maleimide chemistry
N-terminal pegylation may be preferable to minimize activity interference
PEG size optimization:
Smaller PEG molecules (5-20 kDa) maintain better biological activity
Larger PEG molecules (30-40 kDa) provide longer half-life
Branched PEG structures may offer better protection from proteolysis
Reaction conditions:
pH 7.2-7.4 for lysine-targeted pegylation
pH 6.5-7.0 for cysteine-targeted pegylation
Temperature: typically 4-25°C depending on protein stability
Reaction time: 1-4 hours with continuous gentle mixing
Protein:PEG molar ratio typically 1:3 to 1:10
Purification and characterization:
Size exclusion chromatography to separate pegylated from non-pegylated protein
Mass spectrometry to confirm pegylation site(s) and degree
Activity assays to ensure antagonist function is preserved
The optimal protocol should achieve a balance between enhanced pharmacokinetic properties and preserved antagonist activity.
Robust experimental design for evaluating pegylated leptin antagonist effects should include:
Study design elements:
Power analysis to determine appropriate sample size (typically n=8-12 per group)
Randomization and blinding procedures to minimize bias
Appropriate controls (vehicle, non-pegylated antagonist, wild-type leptin)
Dose-response studies to establish optimal dosing
Time-course evaluations to determine duration of effect
Key measurements:
Body weight and food intake (daily measurements)
Body composition analysis (MRI or DEXA)
Energy expenditure via indirect calorimetry
Core body temperature (implanted transponders)
Glucose homeostasis (GTT, ITT, clamp studies)
Molecular assessments:
Hypothalamic neuropeptide expression (qPCR, in situ hybridization)
Synaptic density on NPY/AgRP and POMC neurons (immunohistochemistry)
Signaling pathway activation state (phospho-specific Western blots)
Circulating hormone levels (leptin, insulin, ghrelin)
Data collection schedule:
Baseline measurements (3-7 days)
Treatment period (acute: 24-72h; chronic: 2-12 weeks)
Washout period to assess reversibility
When evaluating antagonists like S120A/T121A or R128Q that increase body weight in mice , researchers should ensure sufficient study duration to capture the full extent of metabolic adaptations.
When encountering contradictory findings regarding leptin receptor modulation, researchers should consider several key factors:
Context-dependent effects:
Physiological state (lean vs. obese, insulin sensitive vs. resistant)
Disease model specifics (cancer type, metabolic disorder severity)
Acute vs. chronic treatment paradigms
Central vs. peripheral actions
Technical considerations:
Dose-dependent effects (hormetic responses are common)
Timing of assessments relative to treatment
Bioavailability differences between formulations
Specificity of the antagonist for leptin vs. other pathways
Biological complexity:
Compensatory mechanisms and feedback loops
Interactions with other hormonal systems
Genetic background effects
Age and sex differences
For example, the search results reveal apparent contradictions regarding leptin modulation in cancer. While many human cancers show overexpression of leptin receptor (ObR) correlated with leptin presence , suggesting potential benefits of antagonism, the precise mechanisms and context-dependence of these effects require careful interpretation.
Researchers should design comparative studies that directly test hypotheses about contradictory findings, using consistent methodologies across different disease models. Meta-analysis approaches can also help identify patterns across seemingly contradictory studies.
Complex datasets from pegylated leptin antagonist studies require sophisticated statistical approaches:
For longitudinal data (body weight, food intake over time):
Mixed-effects models with appropriate covariance structures
Repeated measures ANOVA with post-hoc tests
Area under the curve (AUC) analysis followed by between-group comparisons
For multidimensional datasets:
Principal component analysis to identify major sources of variation
Multivariate ANOVA for simultaneous consideration of multiple endpoints
Path analysis to evaluate relationships between variables
For dose-response relationships:
Non-linear regression models (four-parameter logistic models)
EC50/IC50 calculations with confidence intervals
Comparison of dose-response curves between treatment groups
For controlling Type I error in multiple comparisons:
Bonferroni correction (conservative)
False discovery rate (FDR) methods (Benjamini-Hochberg procedure)
Planned orthogonal contrasts to test specific hypotheses
For reproducibility and robustness:
Bootstrap and jackknife resampling techniques
Sensitivity analyses with varying assumptions
Bayesian approaches incorporating prior knowledge
When analyzing data from cancer studies with PEG-LPrA2 , researchers should consider tumor growth kinetics models and survival analysis techniques alongside standard statistical approaches.
Several promising research directions for pegylated leptin antagonists emerge from the current literature:
Cancer applications:
Combination therapy with standard chemotherapeutics for breast cancer, leveraging PEG-LPrA2's ability to reduce VEGF and VEGFR2 expression
Targeting specific cancer subtypes with known leptin receptor overexpression
Development of tumor-targeted delivery systems for leptin antagonists
Investigation of leptin antagonism in cancer-cachexia syndrome
Metabolic disorder applications:
Leptin-melanocortin system combination therapies leveraging leptin's effects on POMC and α-MSH pathways
Development of tissue-selective antagonists that preferentially target peripheral vs. central leptin receptors
Intermittent leptin antagonism protocols to reset leptin sensitivity
Exploration of leptin antagonists in lipodystrophy and other conditions of leptin deficiency
Novel formulation approaches:
Development of oral delivery systems for pegylated leptin antagonists
Sustained-release formulations for once-monthly administration
Blood-brain barrier penetrant variants for enhanced central effects
Research specifically targeting the amino acid sequence 39-42 in the AB loop of leptin appears particularly promising, as this region is fundamental for ObR activation and represents an important target sequence for developing ObR antagonists with high specificity and potency .
Emerging technologies offer numerous opportunities to enhance pegylated leptin antagonists:
Protein engineering advances:
Structure-guided design of antagonists based on the critical binding sites identified in leptin-receptor interactions
Non-natural amino acid incorporation for site-specific pegylation
Fusion protein approaches combining leptin antagonist domains with stability-enhancing scaffolds
Development of switchable antagonists that respond to physiological cues
Advanced pegylation strategies:
Site-specific enzymatic pegylation to ensure homogeneous products
Releasable PEG linkers that detach under specific conditions
Hydrolytically degradable PEG to address concerns about PEG accumulation
Alternative polymer conjugation (e.g., hyperbrached polyglycerol) for improved properties
Delivery system innovations:
Nanoparticle formulations for targeted delivery to specific tissues
Controlled-release systems using biodegradable polymers
Cell-specific targeting using peptide or antibody conjugation
Stimuli-responsive systems triggered by disease-specific conditions
Particularly promising is the development of leptin antagonists that leverage the D23L mutation, which greatly enhances human leptin's affinity toward the leptin receptor , combined with advanced pegylation strategies to optimize pharmacokinetics while maintaining this enhanced binding affinity.
To elucidate tissue-specific mechanisms of pegylated leptin antagonists, researchers should consider:
Advanced imaging techniques:
PET imaging with labeled antagonists to track tissue distribution
CLARITY or iDISCO tissue clearing with fluorescent antagonists for 3D visualization
Intravital microscopy to observe antagonist effects in real-time in vivo
Tissue-specific conditional models:
Cre-lox systems to delete LEP-R in specific cell types
Inducible expression systems to temporally control LEP-R expression
CRISPR-based approaches to introduce specific LEP-R mutations
Single-cell technologies:
Single-cell RNA-seq to identify cell populations responsive to antagonist treatment
Single-cell proteomics to characterize signaling pathway alterations
Spatial transcriptomics to map antagonist effects while preserving tissue architecture
System biology approaches:
Comprehensive multi-omics profiling (transcriptomics, proteomics, metabolomics)
Network analysis to identify key nodes in leptin signaling networks
Computational modeling to predict antagonist effects across tissues
For mutations such as S120A/T121A that bind specifically to the CRH2 domain of ObR without causing activation , understanding the structural basis and downstream consequences of this selective binding could facilitate development of next-generation antagonists with enhanced specificity and efficacy.
While specific comparative data is not provided in the search results, this table structure illustrates how such data should be presented:
Leptin is a hormone predominantly made by adipose cells and enterocytes in the small intestine that helps to regulate energy balance by inhibiting hunger, which in turn diminishes fat storage in adipocytes. The Leptin Antagonist Triple Mutant (Human Recombinant, Pegylated) is a modified form of leptin designed to counteract the effects of endogenous leptin, making it a valuable tool in research related to obesity and metabolic disorders.
The Leptin Antagonist Triple Mutant is a single non-glycosylated polypeptide chain consisting of 146 amino acids with an additional alanine at the N-terminus. The molecular weight of this protein is approximately 35.6 kDa. However, due to its pegylation, it exhibits an enlarged hydrodynamic volume, causing it to run on SDS-PAGE as a 48 kDa protein and in gel-filtration on Superdex 200 as over 200 kDa protein .
Pegylation refers to the process of attaching polyethylene glycol (PEG) chains to a molecule, in this case, the leptin antagonist. The pegylation of the Leptin Antagonist Triple Mutant involves the attachment of a 20 kDa PEG molecule. This modification enhances the stability and solubility of the protein, prolongs its half-life in circulation, and reduces immunogenicity .
The recombinant production of the Leptin Antagonist Triple Mutant is typically carried out in Escherichia coli. The protein is expressed, purified, and then pegylated using proprietary chromatographic techniques to ensure high purity and consistency. The final product is lyophilized from a concentrated solution containing 0.003 mM NaHCO3 .
The Leptin Antagonist Triple Mutant is primarily used in research to study the role of leptin in energy homeostasis, obesity, and related metabolic disorders. By inhibiting the action of endogenous leptin, researchers can better understand the physiological and pathological processes regulated by this hormone.